Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 180(6): 1178-1197.e20, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32200800

RESUMEN

Social impairment is frequently associated with mitochondrial dysfunction and altered neurotransmission. Although mitochondrial function is crucial for brain homeostasis, it remains unknown whether mitochondrial disruption contributes to social behavioral deficits. Here, we show that Drosophila mutants in the homolog of the human CYFIP1, a gene linked to autism and schizophrenia, exhibit mitochondrial hyperactivity and altered group behavior. We identify the regulation of GABA availability by mitochondrial activity as a biologically relevant mechanism and demonstrate its contribution to social behavior. Specifically, increased mitochondrial activity causes gamma aminobutyric acid (GABA) sequestration in the mitochondria, reducing GABAergic signaling and resulting in social deficits. Pharmacological and genetic manipulation of mitochondrial activity or GABA signaling corrects the observed abnormalities. We identify Aralar as the mitochondrial transporter that sequesters GABA upon increased mitochondrial activity. This study increases our understanding of how mitochondria modulate neuronal homeostasis and social behavior under physiopathological conditions.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Drosophila/metabolismo , Mitocondrias/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Modificados Genéticamente , Ácido Aspártico/metabolismo , Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Proteínas de Drosophila/fisiología , Drosophila melanogaster/metabolismo , Glucosa/metabolismo , Homeostasis , Humanos , Masculino , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Conducta Social , Transmisión Sináptica , Ácido gamma-Aminobutírico/genética
2.
Cell ; 147(1): 235-46, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21962519

RESUMEN

Although many genes predisposing to autism spectrum disorders (ASD) have been identified, the biological mechanism(s) remain unclear. Mouse models based on human disease-causing mutations provide the potential for understanding gene function and novel treatment development. Here, we characterize a mouse knockout of the Cntnap2 gene, which is strongly associated with ASD and allied neurodevelopmental disorders. Cntnap2(-/-) mice show deficits in the three core ASD behavioral domains, as well as hyperactivity and epileptic seizures, as have been reported in humans with CNTNAP2 mutations. Neuropathological and physiological analyses of these mice before the onset of seizures reveal neuronal migration abnormalities, reduced number of interneurons, and abnormal neuronal network activity. In addition, treatment with the FDA-approved drug risperidone ameliorates the targeted repetitive behaviors in the mutant mice. These data demonstrate a functional role for CNTNAP2 in brain development and provide a new tool for mechanistic and therapeutic research in ASD.


Asunto(s)
Trastorno Autístico/genética , Encéfalo/crecimiento & desarrollo , Modelos Animales de Enfermedad , Proteínas de la Membrana/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Animales , Trastorno Autístico/patología , Encéfalo/metabolismo , Encéfalo/patología , Movimiento Celular , Epilepsia/genética , Humanos , Interneuronas/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones Noqueados , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neuronas/patología
3.
Epilepsia ; 62(11): 2845-2857, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34510432

RESUMEN

OBJECTIVE: Dravet syndrome is a severe developmental and epileptic encephalopathy (DEE) most often caused by de novo pathogenic variants in SCN1A. Individuals with Dravet syndrome rarely achieve seizure control and have significantly elevated risk for sudden unexplained death in epilepsy (SUDEP). Heterozygous deletion of Scn1a in mice (Scn1a+/- ) recapitulates several core phenotypes, including temperature-dependent and spontaneous seizures, SUDEP, and behavioral abnormalities. Furthermore, Scn1a+/- mice exhibit a similar clinical response to standard anticonvulsants. Cholesterol 24-hydroxlase (CH24H) is a brain-specific enzyme responsible for cholesterol catabolism. Recent research has indicated the therapeutic potential of CH24H inhibition for diseases associated with neural excitation, including seizures. METHODS: In this study, the novel compound soticlestat, a CH24H inhibitor, was administered to Scn1a+/- mice to investigate its ability to improve Dravet-like phenotypes in this preclinical model. RESULTS: Soticlestat treatment reduced seizure burden, protected against hyperthermia-induced seizures, and completely prevented SUDEP in Scn1a+/- mice. Video-electroencephalography (EEG) analysis confirmed the ability of soticlestat to reduce occurrence of electroclinical seizures. SIGNIFICANCE: This study demonstrates that soticlestat-mediated inhibition of CH24H provides therapeutic benefit for the treatment of Dravet syndrome in mice and has the potential for treatment of DEEs.


Asunto(s)
Epilepsias Mioclónicas , Epilepsia , Piperidinas , Piridinas , Convulsiones Febriles , Muerte Súbita e Inesperada en la Epilepsia , Animales , Colesterol 24-Hidroxilasa/antagonistas & inhibidores , Epilepsias Mioclónicas/complicaciones , Epilepsias Mioclónicas/tratamiento farmacológico , Epilepsias Mioclónicas/genética , Epilepsia/genética , Síndromes Epilépticos , Ratones , Mortalidad Prematura , Mutación , Canal de Sodio Activado por Voltaje NAV1.1/genética , Piperidinas/farmacología , Piridinas/farmacología , Convulsiones/etiología , Convulsiones/genética , Convulsiones Febriles/tratamiento farmacológico , Muerte Súbita e Inesperada en la Epilepsia/etiología
4.
PLoS Genet ; 10(5): e1004402, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24875834

RESUMEN

DNA mutational events are increasingly being identified in autism spectrum disorder (ASD), but the potential additional role of dysregulation of the epigenome in the pathogenesis of the condition remains unclear. The epigenome is of interest as a possible mediator of environmental effects during development, encoding a cellular memory reflected by altered function of progeny cells. Advanced maternal age (AMA) is associated with an increased risk of having a child with ASD for reasons that are not understood. To explore whether AMA involves covert aneuploidy or epigenetic dysregulation leading to ASD in the offspring, we tested a homogeneous ectodermal cell type from 47 individuals with ASD compared with 48 typically developing (TD) controls born to mothers of ≥35 years, using a quantitative genome-wide DNA methylation assay. We show that DNA methylation patterns are dysregulated in ectodermal cells in these individuals, having accounted for confounding effects due to subject age, sex and ancestral haplotype. We did not find mosaic aneuploidy or copy number variability to occur at differentially-methylated regions in these subjects. Of note, the loci with distinctive DNA methylation were found at genes expressed in the brain and encoding protein products significantly enriched for interactions with those produced by known ASD-causing genes, representing a perturbation by epigenomic dysregulation of the same networks compromised by DNA mutational mechanisms. The results indicate the presence of a mosaic subpopulation of epigenetically-dysregulated, ectodermally-derived cells in subjects with ASD. The epigenetic dysregulation observed in these ASD subjects born to older mothers may be associated with aging parental gametes, environmental influences during embryogenesis or could be the consequence of mutations of the chromatin regulatory genes increasingly implicated in ASD. The results indicate that epigenetic dysregulatory mechanisms may complement and interact with DNA mutations in the pathogenesis of the disorder.


Asunto(s)
Factores de Edad , Trastornos Generalizados del Desarrollo Infantil/genética , Metilación de ADN/genética , Epigénesis Genética , Mosaicismo , Adulto , Trastornos Generalizados del Desarrollo Infantil/patología , Aberraciones Cromosómicas , Femenino , Perfilación de la Expresión Génica , Genoma Humano , Haplotipos , Humanos , Masculino , Relaciones Materno-Fetales , Persona de Mediana Edad , Embarazo
5.
Nature ; 466(7304): 368-72, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20531469

RESUMEN

The autism spectrum disorders (ASDs) are a group of conditions characterized by impairments in reciprocal social interaction and communication, and the presence of restricted and repetitive behaviours. Individuals with an ASD vary greatly in cognitive development, which can range from above average to intellectual disability. Although ASDs are known to be highly heritable ( approximately 90%), the underlying genetic determinants are still largely unknown. Here we analysed the genome-wide characteristics of rare (<1% frequency) copy number variation in ASD using dense genotyping arrays. When comparing 996 ASD individuals of European ancestry to 1,287 matched controls, cases were found to carry a higher global burden of rare, genic copy number variants (CNVs) (1.19 fold, P = 0.012), especially so for loci previously implicated in either ASD and/or intellectual disability (1.69 fold, P = 3.4 x 10(-4)). Among the CNVs there were numerous de novo and inherited events, sometimes in combination in a given family, implicating many novel ASD genes such as SHANK2, SYNGAP1, DLGAP2 and the X-linked DDX53-PTCHD1 locus. We also discovered an enrichment of CNVs disrupting functional gene sets involved in cellular proliferation, projection and motility, and GTPase/Ras signalling. Our results reveal many new genetic and functional targets in ASD that may lead to final connected pathways.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/fisiopatología , Variaciones en el Número de Copia de ADN/genética , Dosificación de Gen/genética , Predisposición Genética a la Enfermedad/genética , Estudios de Casos y Controles , Movimiento Celular , Niño , Trastornos Generalizados del Desarrollo Infantil/patología , Citoprotección , Europa (Continente)/etnología , Estudio de Asociación del Genoma Completo , Humanos , Transducción de Señal , Conducta Social
6.
Nature ; 459(7246): 528-33, 2009 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-19404256

RESUMEN

Autism spectrum disorders (ASDs) represent a group of childhood neurodevelopmental and neuropsychiatric disorders characterized by deficits in verbal communication, impairment of social interaction, and restricted and repetitive patterns of interests and behaviour. To identify common genetic risk factors underlying ASDs, here we present the results of genome-wide association studies on a cohort of 780 families (3,101 subjects) with affected children, and a second cohort of 1,204 affected subjects and 6,491 control subjects, all of whom were of European ancestry. Six single nucleotide polymorphisms between cadherin 10 (CDH10) and cadherin 9 (CDH9)-two genes encoding neuronal cell-adhesion molecules-revealed strong association signals, with the most significant SNP being rs4307059 (P = 3.4 x 10(-8), odds ratio = 1.19). These signals were replicated in two independent cohorts, with combined P values ranging from 7.4 x 10(-8) to 2.1 x 10(-10). Our results implicate neuronal cell-adhesion molecules in the pathogenesis of ASDs, and represent, to our knowledge, the first demonstration of genome-wide significant association of common variants with susceptibility to ASDs.


Asunto(s)
Trastorno Autístico/genética , Cromosomas Humanos Par 5/genética , Predisposición Genética a la Enfermedad/genética , Variación Genética/genética , Encéfalo/metabolismo , Cadherinas/genética , Estudios de Casos y Controles , Adhesión Celular/genética , Moléculas de Adhesión Celular Neuronal/genética , Estudios de Cohortes , Marcadores Genéticos/genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Polimorfismo de Nucleótido Simple/genética , Reproducibilidad de los Resultados
7.
J Neurosci ; 33(7): 2732-53, 2013 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-23407934

RESUMEN

The immense molecular diversity of neurons challenges our ability to understand the genetic and cellular etiology of neuropsychiatric disorders. Leveraging knowledge from neurobiology may help parse the genetic complexity: identifying genes important for a circuit that mediates a particular symptom of a disease may help identify polymorphisms that contribute to risk for the disease as a whole. The serotonergic system has long been suspected in disorders that have symptoms of repetitive behaviors and resistance to change, including autism. We generated a bacTRAP mouse line to permit translational profiling of serotonergic neurons. From this, we identified several thousand serotonergic-cell expressed transcripts, of which 174 were highly enriched, including all known markers of these cells. Analysis of common variants near the corresponding genes in the AGRE collection implicated the RNA binding protein CELF6 in autism risk. Screening for rare variants in CELF6 identified an inherited premature stop codon in one of the probands. Subsequent disruption of Celf6 in mice resulted in animals exhibiting resistance to change and decreased ultrasonic vocalization as well as abnormal levels of serotonin in the brain. This work provides a reproducible and accurate method to profile serotonergic neurons under a variety of conditions and suggests a novel paradigm for gaining information on the etiology of psychiatric disorders.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/psicología , Perfilación de la Expresión Génica/métodos , Modificación Traduccional de las Proteínas/genética , Modificación Traduccional de las Proteínas/fisiología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Neuronas Serotoninérgicas/fisiología , Serotonina/fisiología , Animales , Conducta Animal/fisiología , Proteínas CELF , Estudio de Asociación del Genoma Completo , Humanos , Inmunohistoquímica , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Mutación/genética , Mutación/fisiología , Neurotransmisores/metabolismo , Polimorfismo Genético , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Ribosomas/genética , Ribosomas/metabolismo , Proteínas de Transporte de Serotonina en la Membrana Plasmática/genética , Olfato/fisiología , Conducta Social , Vocalización Animal/fisiología
8.
Hum Mol Genet ; 21(21): 4761-73, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22872700

RESUMEN

Although genetic variations in several genes encoding for synaptic adhesion proteins have been found to be associated with autism spectrum disorders, one of the most consistently replicated genes has been CNTNAP2, encoding for contactin-associated protein-like 2 (CASPR2), a multidomain transmembrane protein of the neurexin superfamily. Using immunofluorescence confocal microscopy and complementary biochemical techniques, we compared wild-type CASPR2 to 12 point mutations identified in individuals with autism. In contrast to the wild-type protein, localized to the cell surface, some of the mutants show altered cellular disposition. In particular, CASPR2-D1129H is largely retained in the endoplasmic reticulum (ER) in HEK-293 cells and in hippocampal neurons. BiP/Grp78, Calnexin and ERp57, key ER chaperones, appear to be responsible for retention of this mutant and activation of one signaling pathway of the unfolded protein response (UPR). The presence of this mutation also lowers expression and activates proteosomal degradation. A frame-shift mutation that causes a form of syndromic epilepsy (CASPR2-1253*), results in a secreted protein with seemingly normal folding and oligomerization. Taken together, these data indicate that CASPR2-D1129H has severe trafficking abnormalities and CASPR2-1253* is a secreted soluble protein, suggesting that the structural or signaling functions of the membrane tethered form are lost. Our data support a complex genetic architecture in which multiple distinct risk factors interact with others to shape autism risk and presentation.


Asunto(s)
Factor de Transcripción Activador 6 , Trastornos Generalizados del Desarrollo Infantil/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Mutación Puntual , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Niño , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Chaperón BiP del Retículo Endoplásmico , Regulación de la Expresión Génica , Células HEK293 , Hipocampo/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Pliegue de Proteína , Transporte de Proteínas/genética , Transducción de Señal , Respuesta de Proteína Desplegada/genética
9.
Nat Rev Genet ; 9(5): 341-55, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18414403

RESUMEN

Autism is a heterogeneous syndrome defined by impairments in three core domains: social interaction, language and range of interests. Recent work has led to the identification of several autism susceptibility genes and an increased appreciation of the contribution of de novo and inherited copy number variation. Promising strategies are also being applied to identify common genetic risk variants. Systems biology approaches, including array-based expression profiling, are poised to provide additional insights into this group of disorders, in which heterogeneity, both genetic and phenotypic, is emerging as a dominant theme.


Asunto(s)
Trastorno Autístico/genética , Dosificación de Gen , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Análisis de Secuencia por Matrices de Oligonucleótidos , Animales , Humanos , Fenotipo , Biología de Sistemas/métodos , Biología de Sistemas/tendencias
10.
Hum Mol Genet ; 19(20): 4072-82, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20663923

RESUMEN

Although autism spectrum disorders (ASDs) have a substantial genetic basis, most of the known genetic risk has been traced to rare variants, principally copy number variants (CNVs). To identify common risk variation, the Autism Genome Project (AGP) Consortium genotyped 1558 rigorously defined ASD families for 1 million single-nucleotide polymorphisms (SNPs) and analyzed these SNP genotypes for association with ASD. In one of four primary association analyses, the association signal for marker rs4141463, located within MACROD2, crossed the genome-wide association significance threshold of P < 5 × 10(-8). When a smaller replication sample was analyzed, the risk allele at rs4141463 was again over-transmitted; yet, consistent with the winner's curse, its effect size in the replication sample was much smaller; and, for the combined samples, the association signal barely fell below the P < 5 × 10(-8) threshold. Exploratory analyses of phenotypic subtypes yielded no significant associations after correction for multiple testing. They did, however, yield strong signals within several genes, KIAA0564, PLD5, POU6F2, ST8SIA2 and TAF1C.


Asunto(s)
Trastorno Autístico/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Polimorfismo de Nucleótido Simple , Alelos , Variaciones en el Número de Copia de ADN , Bases de Datos Genéticas , Variación Genética , Genoma Humano , Genotipo , Humanos , Factores de Riesgo , Población Blanca/genética
11.
Hum Genet ; 131(4): 565-79, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21996756

RESUMEN

Autism spectrum disorder (ASD) is a highly heritable disorder of complex and heterogeneous aetiology. It is primarily characterized by altered cognitive ability including impaired language and communication skills and fundamental deficits in social reciprocity. Despite some notable successes in neuropsychiatric genetics, overall, the high heritability of ASD (~90%) remains poorly explained by common genetic risk variants. However, recent studies suggest that rare genomic variation, in particular copy number variation, may account for a significant proportion of the genetic basis of ASD. We present a large scale analysis to identify candidate genes which may contain low-frequency recessive variation contributing to ASD while taking into account the potential contribution of population differences to the genetic heterogeneity of ASD. Our strategy, homozygous haplotype (HH) mapping, aims to detect homozygous segments of identical haplotype structure that are shared at a higher frequency amongst ASD patients compared to parental controls. The analysis was performed on 1,402 Autism Genome Project trios genotyped for 1 million single nucleotide polymorphisms (SNPs). We identified 25 known and 1,218 novel ASD candidate genes in the discovery analysis including CADM2, ABHD14A, CHRFAM7A, GRIK2, GRM3, EPHA3, FGF10, KCND2, PDZK1, IMMP2L and FOXP2. Furthermore, 10 of the previously reported ASD genes and 300 of the novel candidates identified in the discovery analysis were replicated in an independent sample of 1,182 trios. Our results demonstrate that regions of HH are significantly enriched for previously reported ASD candidate genes and the observed association is independent of gene size (odds ratio 2.10). Our findings highlight the applicability of HH mapping in complex disorders such as ASD and offer an alternative approach to the analysis of genome-wide association data.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo/métodos , Haplotipos/genética , Adulto , Niño , Análisis por Conglomerados , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Femenino , Genotipo , Homocigoto , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Núcleo Familiar , Polimorfismo de Nucleótido Simple
12.
PLoS Genet ; 5(6): e1000536, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19557195

RESUMEN

The genetics underlying the autism spectrum disorders (ASDs) is complex and remains poorly understood. Previous work has demonstrated an important role for structural variation in a subset of cases, but has lacked the resolution necessary to move beyond detection of large regions of potential interest to identification of individual genes. To pinpoint genes likely to contribute to ASD etiology, we performed high density genotyping in 912 multiplex families from the Autism Genetics Resource Exchange (AGRE) collection and contrasted results to those obtained for 1,488 healthy controls. Through prioritization of exonic deletions (eDels), exonic duplications (eDups), and whole gene duplication events (gDups), we identified more than 150 loci harboring rare variants in multiple unrelated probands, but no controls. Importantly, 27 of these were confirmed on examination of an independent replication cohort comprised of 859 cases and an additional 1,051 controls. Rare variants at known loci, including exonic deletions at NRXN1 and whole gene duplications encompassing UBE3A and several other genes in the 15q11-q13 region, were observed in the course of these analyses. Strong support was likewise observed for previously unreported genes such as BZRAP1, an adaptor molecule known to regulate synaptic transmission, with eDels or eDups observed in twelve unrelated cases but no controls (p = 2.3x10(-5)). Less is known about MDGA2, likewise observed to be case-specific (p = 1.3x10(-4)). But, it is notable that the encoded protein shows an unexpectedly high similarity to Contactin 4 (BLAST E-value = 3x10(-39)), which has also been linked to disease. That hundreds of distinct rare variants were each seen only once further highlights complexity in the ASDs and points to the continued need for larger cohorts.


Asunto(s)
Trastorno Autístico/genética , Exones , Dosificación de Gen , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Adolescente , Proteínas de Unión al Calcio , Estudios de Casos y Controles , Moléculas de Adhesión Celular Neuronal , Niño , Preescolar , Estudios de Cohortes , Femenino , Duplicación de Gen , Humanos , Masculino , Proteínas del Tejido Nervioso/genética , Moléculas de Adhesión de Célula Nerviosa , Linaje , Eliminación de Secuencia , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
13.
Am J Hum Genet ; 82(1): 150-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18179893

RESUMEN

Autism is a genetically complex neurodevelopmental syndrome in which language deficits are a core feature. We describe results from two complimentary approaches used to identify risk variants on chromosome 7 that likely contribute to the etiology of autism. A two-stage association study tested 2758 SNPs across a 10 Mb 7q35 language-related autism QTL in AGRE (Autism Genetic Resource Exchange) trios and found significant association with Contactin Associated Protein-Like 2 (CNTNAP2), a strong a priori candidate. Male-only containing families were identified as primarily responsible for this association signal, consistent with the strong male affection bias in ASD and other language-based disorders. Gene-expression analyses in developing human brain further identified CNTNAP2 as enriched in circuits important for language development. Together, these results provide convergent evidence for involvement of CNTNAP2, a Neurexin family member, in autism, and demonstrate a connection between genetic risk for autism and specific brain structures.


Asunto(s)
Trastorno Autístico/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Encéfalo/embriología , Niño , Cromosomas Humanos Par 7 , Femenino , Expresión Génica , Humanos , Desarrollo del Lenguaje , Masculino , Polimorfismo de Nucleótido Simple
14.
Am J Hum Genet ; 82(1): 165-73, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18179895

RESUMEN

Autism spectrum disorders (ASD) are a group of related neurodevelopmental syndromes with complex genetic etiology. We identified a de novo chromosome 7q inversion disrupting Autism susceptibility candidate 2 (AUTS2) and Contactin Associated Protein-Like 2 (CNTNAP2) in a child with cognitive and social delay. We focused our initial analysis on CNTNAP2 based on our demonstration of disruption of Contactin 4 (CNTN4) in a patient with ASD; the recent finding of rare homozygous mutations in CNTNAP2 leading to intractable seizures and autism; and in situ and biochemical analyses reported herein that confirm expression in relevant brain regions and demonstrate the presence of CNTNAP2 in the synaptic plasma membrane fraction of rat forebrain lysates. We comprehensively resequenced CNTNAP2 in 635 patients and 942 controls. Among patients, we identified a total of 27 nonsynonymous changes; 13 were rare and unique to patients and 8 of these were predicted to be deleterious by bioinformatic approaches and/or altered residues conserved across all species. One variant at a highly conserved position, I869T, was inherited by four affected children in three unrelated families, but was not found in 4010 control chromosomes (p = 0.014). Overall, this resequencing data demonstrated a modest nonsignificant increase in the burden of rare variants in cases versus controls. Nonetheless, when viewed in light of two independent studies published in this issue of AJHG showing a relationship between ASD and common CNTNAP2 alleles, the cytogenetic and mutation screening data suggest that rare variants may also contribute to the pathophysiology of ASD, but place limits on the magnitude of this contribution.


Asunto(s)
Trastorno Autístico/genética , Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Animales , Niño , Femenino , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , ARN Mensajero , Ratas , Lóbulo Temporal/metabolismo
15.
N Engl J Med ; 359(22): 2337-45, 2008 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-18987363

RESUMEN

BACKGROUND: Rare mutations affecting the FOXP2 transcription factor cause a monogenic speech and language disorder. We hypothesized that neural pathways downstream of FOXP2 influence more common phenotypes, such as specific language impairment. METHODS: We performed genomic screening for regions bound by FOXP2 using chromatin immunoprecipitation, which led us to focus on one particular gene that was a strong candidate for involvement in language impairments. We then tested for associations between single-nucleotide polymorphisms (SNPs) in this gene and language deficits in a well-characterized set of 184 families affected with specific language impairment. RESULTS: We found that FOXP2 binds to and dramatically down-regulates CNTNAP2, a gene that encodes a neurexin and is expressed in the developing human cortex. On analyzing CNTNAP2 polymorphisms in children with typical specific language impairment, we detected significant quantitative associations with nonsense-word repetition, a heritable behavioral marker of this disorder (peak association, P=5.0x10(-5) at SNP rs17236239). Intriguingly, this region coincides with one associated with language delays in children with autism. CONCLUSIONS: The FOXP2-CNTNAP2 pathway provides a mechanistic link between clinically distinct syndromes involving disrupted language.


Asunto(s)
Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Trastornos del Desarrollo del Lenguaje/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Polimorfismo de Nucleótido Simple , Niño , Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Femenino , Factores de Transcripción Forkhead/metabolismo , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Haplotipos , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa
16.
Front Behav Neurosci ; 13: 141, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31293404

RESUMEN

Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and autism. FXS is also accompanied by attention problems, hyperactivity, anxiety, aggression, poor sleep, repetitive behaviors, and self-injury. Recent work supports the role of γ-aminobutyric-acid (GABA), the primary inhibitory neurotransmitter in the brain, in mediating symptoms of FXS. Deficits in GABA machinery have been observed in a mouse model of FXS, including a loss of tonic inhibition in the amygdala, which is mediated by extrasynaptic GABAA receptors. Humans with FXS also show reduced GABAA receptor availability. Here, we sought to evaluate the potential of gaboxadol (also called OV101 and THIP), a selective and potent agonist for delta-subunit-containing extrasynaptic GABAA receptors (dSEGA), as a therapeutic agent for FXS by assessing its ability to normalize aberrant behaviors in a relatively uncharacterized mouse model of FXS (Fmr1 KO2 mice). Four behavioral domains (hyperactivity, anxiety, aggression, and repetitive behaviors) were probed using a battery of behavioral assays. The results showed that Fmr1 KO2 mice were hyperactive, had abnormal anxiety-like behavior, were more irritable and aggressive, and had an increased frequency of repetitive behaviors compared to wild-type (WT) littermates, which are all behavioral deficits reminiscent of individuals with FXS. Treatment with gaboxadol normalized all of the aberrant behaviors observed in Fmr1 KO2 mice back to WT levels, providing evidence of its potential benefit for treating FXS. We show that the potentiation of extrasynaptic GABA receptors alone, by gaboxadol, is sufficient to normalize numerous behavioral deficits in the FXS model using endpoints that are directly translatable to the clinical presentation of FXS. Taken together, these data support the future evaluation of gaboxadol in individuals with FXS, particularly with regard to symptoms of hyperactivity, anxiety, irritability, aggression, and repetitive behaviors.

17.
Biol Psychiatry ; 86(4): 306-314, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31202490

RESUMEN

BACKGROUND: Deletions encompassing a four-gene region on chromosome 15 (BP1-BP2 at 15q11.2), seen at a population frequency of 1 in 500, are associated with increased risk for schizophrenia, epilepsy, and other common neurodevelopmental disorders. However, little is known in terms of how these common deletions impact cognition. METHODS: We used a Web-based tool to characterize cognitive function in a novel cohort of adult carriers and their noncarrier family members. Results from 31 carrier and 38 noncarrier parents from 40 families were compared with control data from 6530 individuals who self-registered on the Lumosity platform and opted in to participate in research. We then examined aspects of sensory and cognitive function in flies harboring a mutation in Cyfip, the homologue of one of the genes within the deletion. For the fly studies, 10 or more groups of 50 individuals per genotype were included. RESULTS: Our human studies revealed profound deficits in grammatical reasoning, arithmetic reasoning, and working memory in BP1-BP2 deletion carriers. No such deficits were observed in noncarrier spouses. Our fly studies revealed deficits in associative and nonassociative learning despite intact sensory perception. CONCLUSIONS: Our results provide new insights into outcomes associated with BP1-BP2 deletions and call for a discussion on how to appropriately communicate these findings to unaffected carriers. Findings also highlight the utility of an online tool in characterizing cognitive function in a geographically distributed population.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Disfunción Cognitiva/genética , Proteínas de Drosophila/genética , Drosophila/genética , Trastornos del Neurodesarrollo/genética , Adulto , Animales , Aberraciones Cromosómicas , Cromosomas Humanos Par 15/genética , Disfunción Cognitiva/fisiopatología , Estudios de Cohortes , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Trastornos del Neurodesarrollo/fisiopatología , Fenotipo
18.
Am J Med Genet B Neuropsychiatr Genet ; 147B(6): 880-9, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18205168

RESUMEN

Nuclear receptor 2E1 gene (NR2E1) resides within a 6q21-22 locus for bipolar disorder and schizophrenia. Mice deleted for Nr2e1 show altered neurogenesis, cortical and limbic abnormalities, aggression, hyperexcitability, and cognitive impairment. NR2E1 is therefore a positional and functional candidate for involvement in mental illness. We performed association analyses in 394 patients with bipolar disorder, 396 with schizophrenia, and 479 controls using six common markers and haplotypes. We also performed a comprehensive mutation screen of NR2E1, resequencing its entire coding region, complete 5' and 3' untranslated regions, consensus splice-sites, and evolutionarily conserved regions in 126 humans with bipolar disorder, schizophrenia, or aggressive disorders. NR2E1 was associated with bipolar disorder I and II [odds ratio (OR = 0.77, P = 0.013), bipolar disorder I (OR = 0.77, P = 0.015), bipolar disorder in females (OR = 0.72, P = 0.009), and with age at onset < or = 25 years (OR = 0.67, P = 0.006)], all of which remained significant after correcting for multiple comparisons. We identified eight novel candidate mutations that were absent in 325 controls; four of these were predicted to alter known neural transcription factor binding sites. Analyses of NR2E1 mRNA in human brain revealed forebrain-specific transcription. The data presented support the hypothesis that genetic variation at NR2E1 may be associated with susceptibility to brain-behavior disorders.


Asunto(s)
Agresión/fisiología , Trastorno Bipolar/genética , Análisis Mutacional de ADN/métodos , Ligamiento Genético , Receptores Citoplasmáticos y Nucleares/genética , Esquizofrenia/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Mutación/fisiología , Receptores Nucleares Huérfanos , Polimorfismo de Nucleótido Simple
19.
Brain Lang ; 174: 50-60, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28738218

RESUMEN

Three lines of evidence motivated this study. 1) CNTNAP2 variation is associated with autism risk and speech-language development. 2) CNTNAP2 variations are associated with differences in white matter (WM) tracts comprising the speech-language circuitry. 3) Children with autism show impairment in multisensory speech perception. Here, we asked whether an autism risk-associated CNTNAP2 single nucleotide polymorphism in neurotypical adults was associated with multisensory speech perception performance, and whether such a genotype-phenotype association was mediated through white matter tract integrity in speech-language circuitry. Risk genotype at rs7794745 was associated with decreased benefit from visual speech and lower fractional anisotropy (FA) in several WM tracts (right precentral gyrus, left anterior corona radiata, right retrolenticular internal capsule). These structural connectivity differences were found to mediate the effect of genotype on audiovisual speech perception, shedding light on possible pathogenic pathways in autism and biological sources of inter-individual variation in audiovisual speech processing in neurotypicals.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/fisiopatología , Encéfalo/patología , Encéfalo/fisiopatología , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Vías Nerviosas , Percepción del Habla/genética , Habla , Estimulación Acústica , Adulto , Anisotropía , Femenino , Humanos , Desarrollo del Lenguaje , Masculino , Persona de Mediana Edad , Estimulación Luminosa , Polimorfismo de Nucleótido Simple , Sustancia Blanca/patología , Sustancia Blanca/fisiopatología , Adulto Joven
20.
J Neurosci ; 25(27): 6263-70, 2005 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-16000615

RESUMEN

"Fierce" mice, homozygous for the deletion of nuclear receptor 2E1 (NR2E1), show abnormal brain-eye development and pathological aggression. To evaluate functional equivalency between mouse and human NR2E1, we generated mice transgenic for a genomic clone spanning the human NR2E1 locus and bred these animals to fierce mice deleted for the corresponding mouse gene. In fierce mutants carrying human NR2E1, structural brain defects were eliminated and eye abnormalities ameliorated. Excitingly, behavior in these "rescue" mice was indistinguishable from controls. Because no artificial promoter was used to drive transgene expression, promoter and regulatory elements within the human NR2E1 clone are functional in mouse. Normal behavior in rescue animals suggests that mechanisms underlying the behavioral abnormalities in fierce mice may also be conserved in humans. Our data support the hypothesis that variation at NR2E1 may contribute to human behavioral disorders. Use of this rescue paradigm with other genes will permit the direct evaluation of human genes hypothesized to play a causal role in psychiatric disease but for which evidence is lacking or equivocal.


Asunto(s)
Agresión/fisiología , Encéfalo/anomalías , Anomalías del Ojo/genética , Receptores Citoplasmáticos y Nucleares/deficiencia , Receptores Citoplasmáticos y Nucleares/fisiología , Conducta Agonística/fisiología , Animales , Encéfalo/embriología , Corteza Cerebral/anomalías , Anomalías Congénitas/embriología , Anomalías Congénitas/genética , Anomalías Congénitas/terapia , Cruzamientos Genéticos , Conducta Exploratoria/fisiología , Anomalías del Ojo/embriología , Anomalías del Ojo/terapia , Femenino , Genotipo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Bulbo Olfatorio/anomalías , Receptores Nucleares Huérfanos , Fenotipo , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/genética , Secuencias Reguladoras de Ácidos Nucleicos , Retina/anomalías , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie , Territorialidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA