Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 110(12): 2103-2111, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-37924809

RESUMEN

Hereditary spastic parapareses (HSPs) are clinically heterogeneous motor neuron diseases with variable age of onset and severity. Although variants in dozens of genes are implicated in HSPs, much of the genetic basis for pediatric-onset HSP remains unexplained. Here, we re-analyzed clinical exome-sequencing data from siblings with HSP of unknown genetic etiology and identified an inherited nonsense mutation (c.523C>T [p.Arg175Ter]) in the highly conserved RAB1A. The mutation is predicted to produce a truncated protein with an intact RAB GTPase domain but without two C-terminal cysteine residues required for proper subcellular protein localization. Additional RAB1A mutations, including two frameshift mutations and a mosaic missense mutation (c.83T>C [p.Leu28Pro]), were identified in three individuals with similar neurodevelopmental presentations. In rescue experiments, production of the full-length, but not the truncated, RAB1a rescued Golgi structure and cell proliferation in Rab1-depleted cells. In contrast, the missense-variant RAB1a disrupted Golgi structure despite intact Rab1 expression, suggesting a dominant-negative function of the mosaic missense mutation. Knock-down of RAB1A in cultured human embryonic stem cell-derived neurons resulted in impaired neuronal arborization. Finally, RAB1A is located within the 2p14-p15 microdeletion syndrome locus. The similar clinical presentations of individuals with RAB1A loss-of-function mutations and the 2p14-p15 microdeletion syndrome implicate loss of RAB1A in the pathogenesis of neurodevelopmental manifestations of this microdeletion syndrome. Our study identifies a RAB1A-related neurocognitive disorder with speech and motor delay, demonstrates an essential role for RAB1a in neuronal differentiation, and implicates RAB1A in the etiology of the neurodevelopmental sequelae associated with the 2p14-p15 microdeletion syndrome.


Asunto(s)
Haploinsuficiencia , Paraplejía Espástica Hereditaria , Niño , Humanos , Haploinsuficiencia/genética , Mutación , Mutación Missense/genética , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Aparato de Golgi/metabolismo , Paraplejía Espástica Hereditaria/genética
2.
Nature ; 539(7628): 242-247, 2016 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-27830782

RESUMEN

Sensory stimuli drive the maturation and function of the mammalian nervous system in part through the activation of gene expression networks that regulate synapse development and plasticity. These networks have primarily been studied in mice, and it is not known whether there are species- or clade-specific activity-regulated genes that control features of brain development and function. Here we use transcriptional profiling of human fetal brain cultures to identify an activity-dependent secreted factor, Osteocrin (OSTN), that is induced by membrane depolarization of human but not mouse neurons. We find that OSTN has been repurposed in primates through the evolutionary acquisition of DNA regulatory elements that bind the activity-regulated transcription factor MEF2. In addition, we demonstrate that OSTN is expressed in primate neocortex and restricts activity-dependent dendritic growth in human neurons. These findings suggest that, in response to sensory input, OSTN regulates features of neuronal structure and function that are unique to primates.


Asunto(s)
Evolución Molecular , Proteínas Musculares/metabolismo , Neocórtex/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Transcriptoma , Animales , Secuencia de Bases , Huesos/metabolismo , Dendritas/metabolismo , Elementos de Facilitación Genéticos/genética , Femenino , Humanos , Factores de Transcripción MEF2/metabolismo , Macaca mulatta , Masculino , Ratones , Datos de Secuencia Molecular , Proteínas Musculares/genética , Músculos/metabolismo , Neocórtex/citología , Neuronas/citología , Especificidad de Órganos , Especificidad de la Especie , Factores de Transcripción/genética
3.
Proc Natl Acad Sci U S A ; 116(9): 3662-3667, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808755

RESUMEN

Kaufman oculocerebrofacial syndrome (KOS) is a recessive neurodevelopmental disorder characterized by intellectual disability and lack of speech. KOS is caused by inactivating mutations in UBE3B, but the underlying biological mechanisms are completely unknown. We found that loss of Ube3b in mice resulted in growth retardation, decreased grip strength, and loss of vocalization. The brains of Ube3b-/- mice had hypoplasia of the corpus callosum, enlarged ventricles, and decreased thickness of the somatosensory cortex. Ube3b-/- cortical neurons had abnormal dendritic morphology and synapses. We identified 22 UBE3B interactors and found that branched-chain α-ketoacid dehydrogenase kinase (BCKDK) is an in vivo UBE3B substrate. Since BCKDK targets several metabolic pathways, we profiled plasma and cortical metabolomes from Ube3b-/- mice. Nucleotide metabolism and the tricarboxylic acid cycle were among the pathways perturbed. Substrate-induced mitochondrial respiration was reduced in skeletal muscle but not in liver of Ube3b-/- mice. To assess the relevance of these findings to humans, we identified three KOS patients who had compound heterozygous UBE3B mutations. We discovered changes in metabolites from similar pathways in plasma from these patients. Collectively, our results implicate a disease mechanism in KOS, suggest that it is a metabolic encephalomyopathy, and provide an entry to targeted therapies.


Asunto(s)
Anomalías del Ojo/genética , Discapacidad Intelectual/genética , Trastornos del Desarrollo del Lenguaje/genética , Deformidades Congénitas de las Extremidades/genética , Microcefalia/genética , Proteínas Quinasas/genética , Ubiquitina-Proteína Ligasas/genética , Adolescente , Adulto , Animales , Encéfalo/fisiopatología , Niño , Anomalías del Ojo/fisiopatología , Facies , Humanos , Discapacidad Intelectual/fisiopatología , Trastornos del Desarrollo del Lenguaje/fisiopatología , Deformidades Congénitas de las Extremidades/fisiopatología , Masculino , Redes y Vías Metabólicas , Ratones , Ratones Noqueados , Microcefalia/fisiopatología , Mutación , Fenotipo , Ubiquitina/genética
4.
Nature ; 515(7526): 209-15, 2014 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-25363760

RESUMEN

The genetic architecture of autism spectrum disorder involves the interplay of common and rare variants and their impact on hundreds of genes. Using exome sequencing, here we show that analysis of rare coding variation in 3,871 autism cases and 9,937 ancestry-matched or parental controls implicates 22 autosomal genes at a false discovery rate (FDR) < 0.05, plus a set of 107 autosomal genes strongly enriched for those likely to affect risk (FDR < 0.30). These 107 genes, which show unusual evolutionary constraint against mutations, incur de novo loss-of-function mutations in over 5% of autistic subjects. Many of the genes implicated encode proteins for synaptic formation, transcriptional regulation and chromatin-remodelling pathways. These include voltage-gated ion channels regulating the propagation of action potentials, pacemaking and excitability-transcription coupling, as well as histone-modifying enzymes and chromatin remodellers-most prominently those that mediate post-translational lysine methylation/demethylation modifications of histones.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Cromatina/genética , Predisposición Genética a la Enfermedad/genética , Mutación/genética , Sinapsis/metabolismo , Transcripción Genética/genética , Secuencia de Aminoácidos , Trastornos Generalizados del Desarrollo Infantil/patología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Exoma/genética , Femenino , Mutación de Línea Germinal/genética , Humanos , Masculino , Datos de Secuencia Molecular , Mutación Missense/genética , Red Nerviosa/metabolismo , Oportunidad Relativa
5.
Neurobiol Learn Mem ; 165: 106791, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-29398581

RESUMEN

The ubiquitin proteasome system (UPS) is a highly conserved pathway that tightly regulates protein turnover in cells. This process is integral to neuronal development, differentiation, and function. Several members of the UPS are disrupted in neuropsychiatric disorders, highlighting the importance of this pathway in brain development and function. In this review, we discuss some of these pathway members, the molecular processes they regulate, and the potential for targeting the UPS in an effort to develop therapeutic strategies in neuropsychiatric and neurodevelopmental disorders.


Asunto(s)
Trastornos Mentales/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Trastorno por Déficit de Atención con Hiperactividad/metabolismo , Trastorno del Espectro Autista/metabolismo , Encefalopatías/metabolismo , Humanos , Discapacidad Intelectual/metabolismo , Redes y Vías Metabólicas/fisiología , Complejo de la Endopetidasa Proteasomal/fisiología , Esquizofrenia/metabolismo , Ubiquitina/fisiología
6.
Am J Med Genet B Neuropsychiatr Genet ; 177(8): 736-745, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30421579

RESUMEN

Protein homeostasis is tightly regulated by the ubiquitin proteasome pathway. Disruption of this pathway gives rise to a host of neurological disorders. Through whole exome sequencing (WES) in families with neurodevelopmental disorders, we identified mutations in PSMD12, a core component of the proteasome, underlying a neurodevelopmental disorder with intellectual disability (ID) and features of autism spectrum disorder (ASD). We performed WES on six affected siblings from a multiplex family with ID and autistic features, the affected father, and two unaffected mothers, and a trio from a simplex family with one affected child with ID and periventricular nodular heterotopia. We identified an inherited heterozygous nonsense mutation in PSMD12 (NM_002816: c.367C>T: p.R123X) in the multiplex family and a de novo nonsense mutation in the same gene (NM_002816: c.601C>T: p.R201X) in the simplex family. PSMD12 encodes a non-ATPase regulatory subunit of the 26S proteasome. We confirm the association of PSMD12 with ID, present the first cases of inherited PSMD12 mutation, and demonstrate the heterogeneity of phenotypes associated with PSMD12 mutations.


Asunto(s)
Discapacidad Intelectual/genética , Complejo de la Endopetidasa Proteasomal/genética , Adolescente , Adulto , Trastorno del Espectro Autista/genética , Trastorno Autístico/genética , Niño , Preescolar , Familia , Femenino , Predisposición Genética a la Enfermedad , Haploinsuficiencia/genética , Humanos , Masculino , Mutación , Trastornos del Neurodesarrollo/genética , Linaje , Complejo de la Endopetidasa Proteasomal/metabolismo , Hermanos , Secuenciación del Exoma
7.
J Neurosci ; 36(45): 11402-11410, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27911742

RESUMEN

Autism spectrum disorder (ASD) is a constellation of neurodevelopmental presentations with high heritability and both phenotypic and genetic heterogeneity. To date, mutations in hundreds of genes have been associated to varying degrees with increased ASD risk. A better understanding of the functions of these genes and whether they fit together in functional groups or impact similar neuronal circuits is needed to develop rational treatment strategies. We will review current areas of emphasis in ASD research, starting from human genetics and exploring how mouse models of human mutations have helped identify specific molecular pathways (protein synthesis and degradation, chromatin remodeling, intracellular signaling), which are linked to alterations in circuit function and cognitive/social behavior. We will conclude by discussing how we can leverage the findings on molecular and cellular alterations found in ASD to develop therapies for neurodevelopmental disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/terapia , Encéfalo/metabolismo , Terapia Genética/métodos , Proteínas del Tejido Nervioso/genética , Trastorno del Espectro Autista/diagnóstico , Medicina Basada en la Evidencia , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Pruebas Genéticas/métodos , Humanos , Terapia Molecular Dirigida/métodos , Proteínas del Tejido Nervioso/metabolismo , Resultado del Tratamiento
8.
Annu Rev Genomics Hum Genet ; 15: 195-213, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25184530

RESUMEN

Advances in genetic tools and sequencing technology in the past few years have vastly expanded our understanding of the genetics of neurodevelopmental disorders. Recent high-throughput sequencing analyses of structural brain malformations, cognitive and neuropsychiatric disorders, and localized cortical dysplasias have uncovered a diverse genetic landscape beyond classic Mendelian patterns of inheritance. The underlying genetic causes of neurodevelopmental disorders implicate numerous cell biological pathways critical for normal brain development.


Asunto(s)
Corteza Cerebral/patología , Trastornos Generalizados del Desarrollo Infantil/genética , Malformaciones del Desarrollo Cortical/genética , Neuronas/patología , Alelos , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Niño , Trastornos Generalizados del Desarrollo Infantil/patología , Citoesqueleto/genética , Citoesqueleto/patología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Dedos/anomalías , Dedos/patología , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Malformaciones del Desarrollo Cortical/patología , Microcefalia/genética , Microcefalia/patología , Hipotonía Muscular/genética , Hipotonía Muscular/patología , Miopía/genética , Miopía/patología , Neuronas/metabolismo , Obesidad/genética , Obesidad/patología , Degeneración Retiniana
9.
Nature ; 468(7321): 263-9, 2010 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21068835

RESUMEN

Mutations in the X-linked MECP2 gene, which encodes the transcriptional regulator methyl-CpG-binding protein 2 (MeCP2), cause Rett syndrome and several neurodevelopmental disorders including cognitive disorders, autism, juvenile-onset schizophrenia and encephalopathy with early lethality. Rett syndrome is characterized by apparently normal early development followed by regression, motor abnormalities, seizures and features of autism, especially stereotyped behaviours. The mechanisms mediating these features are poorly understood. Here we show that mice lacking Mecp2 from GABA (γ-aminobutyric acid)-releasing neurons recapitulate numerous Rett syndrome and autistic features, including repetitive behaviours. Loss of MeCP2 from a subset of forebrain GABAergic neurons also recapitulates many features of Rett syndrome. MeCP2-deficient GABAergic neurons show reduced inhibitory quantal size, consistent with a presynaptic reduction in glutamic acid decarboxylase 1 (Gad1) and glutamic acid decarboxylase 2 (Gad2) levels, and GABA immunoreactivity. These data demonstrate that MeCP2 is critical for normal function of GABA-releasing neurons and that subtle dysfunction of GABAergic neurons contributes to numerous neuropsychiatric phenotypes.


Asunto(s)
Trastorno Autístico/fisiopatología , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/metabolismo , Síndrome de Rett/fisiopatología , Transducción de Señal , Trastorno de Movimiento Estereotipado/fisiopatología , Ácido gamma-Aminobutírico/metabolismo , Animales , Trastorno Autístico/complicaciones , Trastorno Autístico/genética , Trastorno Autístico/patología , Encéfalo/citología , Conducta Compulsiva/complicaciones , Conducta Compulsiva/genética , Conducta Compulsiva/fisiopatología , Modelos Animales de Enfermedad , Electroencefalografía , Genotipo , Glutamato Descarboxilasa/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Proteínas de Homeodominio/genética , Potenciales Postsinápticos Inhibidores , Potenciación a Largo Plazo , Masculino , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Transgénicos , Inhibición Neural , Plasticidad Neuronal , Neuronas/metabolismo , Fenotipo , Terminales Presinápticos/metabolismo , Trastornos Psicomotores/complicaciones , Trastornos Psicomotores/genética , Trastornos Psicomotores/fisiopatología , Reflejo de Sobresalto/genética , Respiración , Síndrome de Rett/complicaciones , Síndrome de Rett/genética , Síndrome de Rett/patología , Conducta Autodestructiva/complicaciones , Conducta Autodestructiva/genética , Conducta Autodestructiva/fisiopatología , Trastorno de Movimiento Estereotipado/complicaciones , Trastorno de Movimiento Estereotipado/genética , Trastorno de Movimiento Estereotipado/patología , Tasa de Supervivencia , Transmisión Sináptica , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores/genética
10.
PLoS Genet ; 8(4): e1002635, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22511880

RESUMEN

Although autism has a clear genetic component, the high genetic heterogeneity of the disorder has been a challenge for the identification of causative genes. We used homozygosity analysis to identify probands from nonconsanguineous families that showed evidence of distant shared ancestry, suggesting potentially recessive mutations. Whole-exome sequencing of 16 probands revealed validated homozygous, potentially pathogenic recessive mutations that segregated perfectly with disease in 4/16 families. The candidate genes (UBE3B, CLTCL1, NCKAP5L, ZNF18) encode proteins involved in proteolysis, GTPase-mediated signaling, cytoskeletal organization, and other pathways. Furthermore, neuronal depolarization regulated the transcription of these genes, suggesting potential activity-dependent roles in neurons. We present a multidimensional strategy for filtering whole-exome sequence data to find candidate recessive mutations in autism, which may have broader applicability to other complex, heterogeneous disorders.


Asunto(s)
Trastorno Autístico/genética , Exones , Genes Recesivos , Mutación , Neuronas , Proteínas Adaptadoras Transductoras de Señales/genética , Cadenas Pesadas de Clatrina/genética , Exones/genética , Genoma Humano , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Neuronas/metabolismo , Neuronas/fisiología , Proteínas Oncogénicas/genética , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética
11.
Biochem Biophys Res Commun ; 437(1): 67-73, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23791832

RESUMEN

Rett syndrome (RTT) and Angelman syndrome (AS) are devastating neurological disorders that share many clinical features. The disease-causing mutations have been identified for both syndromes. Mutations in Methyl-CpG Binding Protein 2 (MECP2) are found in a majority of patients with classical RTT while absence of maternal allele or intragenic mutation in the maternal copy of UBE3A gene encoding the human papilloma virus E6-associated protein (E6AP) cause most cases of AS. Extensive studies have been performed to determine the cause of the neurological problems in each disease. However, the genetic and molecular basis of the overlap in phenotypes between RTT and AS remains largely unknown. Here we present evidence that the phenotypic similarities between the two syndromes might be due to the shared molecular functions between MeCP2 and E6AP in gene expression. Our genetic and biochemical studies suggest that E6AP acts as an essential cofactor for a subset of MeCP2 functions. Specifically, decreased expression of Ube3a was able to rescue the cellular phenotypes induced by MECP2-overexpression in Drosophila. And biochemical assays using mice and cell culture systems show that MeCP2 and E6AP physically interact and regulate the expression of shared target genes. Together these data suggest that MeCP2 and E6AP play a role in the transcriptional control of common target gene expression and provide some insight into why RTT and AS share several neurological phenotypes.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteína 2 de Unión a Metil-CpG/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas Co-Represoras/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/ultraestructura , Ojo/metabolismo , Ojo/ultraestructura , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Fenotipo , Unión Proteica
12.
Autism Res ; 16(1): 31-39, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36415077

RESUMEN

Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by deficits in communication, diminished social skills, and restrictive and repetitive behaviors and interests. ASD affects approximately 2.3% of the population and is highly heterogeneous, both phenotypically and genetically. As genomic technologies advance, our understanding of the genetic architecture of ASD is becoming clearer, encompassing spontaneous and inherited alterations throughout the genome, and delineating alterations that are either rare or common in the population. This commentary provides an overview of the genomic strategies and resulting major findings of genetic alterations associated with ASD.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Humanos , Trastorno Autístico/genética , Trastorno del Espectro Autista/genética , Genómica , Causalidad , Mutación
13.
Transl Psychiatry ; 13(1): 404, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38129378

RESUMEN

The landscape of autism spectrum disorder (ASD) in Lebanon is unique because of high rates of consanguinity, shared ancestry, and increased remote consanguinity. ASD prevalence in Lebanon is 1 in 68 with a male-to-female ratio of 2:1. This study aims to investigate the impact of an inherited deletion in UBLCP1 (Ubiquitin-Like Domain-Containing CTD Phosphatase 1) on the ubiquitin-proteasome system (UPS) and proteolysis. Whole exome sequencing in a Lebanese family with ASD without pathogenic copy number variations (CNVs) uncovered a deletion in UBLCP1. Functional evaluation of the identified variant is described in fibroblasts from the affected. The deletion in UBLCP1 exon 10 (g.158,710,261CAAAG > C) generates a premature stop codon interrupting the phosphatase domain and is predicted as pathogenic. It is absent from databases of normal variation worldwide and in Lebanon. Wild-type UBLCP1 is widely expressed in mouse brains. The mutation results in decreased UBLCP1 protein expression in patient-derived fibroblasts from the autistic patient compared to controls. The truncated UBLCP1 protein results in increased proteasome activity decreased ubiquitinated protein levels, and downregulation in expression of other proteasome subunits in samples from the affected compared to controls. Inhibition of the proteasome by using MG132 in proband cells reverses alterations in gene expression due to the restoration of protein levels of the common transcription factor, NRF1. Finally, treatment with gentamicin, which promotes premature termination codon read-through, restores UBLCP1 expression and function. Discovery of an ASD-linked mutation in UBLCP1 leading to overactivation of cell proteolysis is reported. This, in turn, leads to dysregulation of proteasome subunit transcript levels as a compensatory response.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Animales , Femenino , Humanos , Masculino , Ratones , Trastorno del Espectro Autista/genética , Variaciones en el Número de Copia de ADN , Mutación , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitinas/genética
14.
Sci Adv ; 9(47): eadi0074, 2023 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-37992166

RESUMEN

Chromatin regulation plays a pivotal role in establishing and maintaining cellular identity and is one of the top pathways disrupted in autism spectrum disorder (ASD). The hippocampus, composed of distinct cell types, is often affected in patients with ASD. However, the specific hippocampal cell types and their transcriptional programs that are dysregulated in ASD are unknown. Using single-nucleus RNA sequencing, we show that the ASD gene, lysine demethylase 5A (KDM5A), regulates the development of specific subtypes of excitatory and inhibitory neurons. We found that KDM5A is essential for establishing hippocampal cell identity by controlling a differentiation switch early in development. Our findings define a role for the chromatin regulator KDM5A in establishing hippocampal cell identity and contribute to the emerging convergent mechanisms across ASD.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Humanos , Cromatina/genética , Histonas/genética , Histonas/metabolismo , Trastorno Autístico/genética , Trastorno del Espectro Autista/genética , Diferenciación Celular/genética , Proteína 2 de Unión a Retinoblastoma/genética
15.
Cell Genom ; 3(7): 100322, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37492102

RESUMEN

Autism spectrum disorder (ASD) is a group of complex neurodevelopmental conditions affecting communication and social interaction in 2.3% of children. Studies that demonstrated its complex genetic architecture have been mainly performed in populations of European ancestry. We investigate the genetics of ASD in an East African cohort (129 individuals) from a population with higher prevalence (5%). Whole-genome sequencing identified 2.13 million private variants in the cohort and potentially pathogenic variants in known ASD genes (including CACNA1C, CHD7, FMR1, and TCF7L2). Admixture analysis demonstrated that the cohort comprises two ancestral populations, African and Eurasian. Admixture mapping discovered 10 regions that confer ASD risk on the African haplotypes, containing several known ASD genes. The increased ASD prevalence in this population suggests decreased heterogeneity in the underlying genetic etiology, enabling risk allele identification. Our approach emphasizes the power of African genetic variation and admixture analysis to inform the architecture of complex disorders.

16.
NPJ Genom Med ; 7(1): 13, 2022 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-35190550

RESUMEN

Autism spectrum disorder (ASD) is a collection of neurodevelopmental disorders characterized by deficits in social communication and restricted, repetitive patterns of behavior or interests. ASD is highly heritable, but genetically and phenotypically heterogeneous, reducing the power to identify causative genes. We performed whole genome sequencing (WGS) in an ASD cohort of 68 individuals from 22 families enriched for recent shared ancestry. We identified an average of 3.07 million variants per genome, of which an average of 112,512 were rare. We mapped runs of homozygosity (ROHs) in affected individuals and found an average genomic homozygosity of 9.65%, consistent with expectations for multiple generations of consanguineous unions. We identified potentially pathogenic rare exonic or splice site variants in 12 known (including KMT2C, SCN1A, SPTBN1, SYNE1, ZNF292) and 12 candidate (including CHD5, GRB10, PPP1R13B) ASD genes. Furthermore, we annotated noncoding variants in ROHs with brain-specific regulatory elements and identified putative disease-causing variants within brain-specific promoters and enhancers for 5 known ASD and neurodevelopmental disease genes (ACTG1, AUTS2, CTNND2, CNTNAP4, SPTBN4). We also identified copy number variants in two known ASD and neurodevelopmental disease loci in two affected individuals. In total we identified potentially etiological variants in known ASD or neurodevelopmental disease genes for ~61% (14/23) of affected individuals. We combined WGS with homozygosity mapping and regulatory element annotations to identify candidate ASD variants. Our analyses add to the growing number of ASD genes and variants and emphasize the importance of leveraging recent shared ancestry to map disease variants in complex neurodevelopmental disorders.

17.
Neuron ; 56(3): 422-37, 2007 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-17988628

RESUMEN

The postnatal neurodevelopmental disorder Rett syndrome (RTT) is caused by mutations in the gene encoding methyl-CpG binding protein 2 (MeCP2), a transcriptional repressor involved in chromatin remodeling and the modulation of RNA splicing. MECP2 aberrations result in a constellation of neuropsychiatric abnormalities, whereby both loss of function and gain in MECP2 dosage lead to similar neurological phenotypes. Recent studies demonstrate disease reversibility in RTT mouse models, suggesting that the neurological defects in MECP2 disorders are not permanent. To investigate the potential for restoring neuronal function in RTT patients, it is essential to identify MeCP2 targets or modifiers of the phenotype that can be therapeutically modulated. Moreover, deciphering the molecular underpinnings of RTT is likely to contribute to the understanding of the pathogenesis of a broader class of neuropsychiatric disorders.


Asunto(s)
Encéfalo/metabolismo , Predisposición Genética a la Enfermedad/genética , Proteína 2 de Unión a Metil-CpG/genética , Mutación/genética , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Adolescente , Animales , Encéfalo/fisiopatología , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/genética , Humanos , Lactante , Masculino , Ratones , Fenotipo , Síndrome de Rett/fisiopatología , Inactivación del Cromosoma X/genética
18.
Hum Mol Genet ; 18(13): 2431-42, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19369296

RESUMEN

A group of post-natal neurodevelopmental disorders collectively referred to as MeCP2 disorders are caused by aberrations in the gene encoding methyl-CpG-binding protein 2 (MECP2). Loss of MeCP2 function causes Rett syndrome (RTT), whereas increased copy number of the gene causes MECP2 duplication or triplication syndromes. MeCP2 acts as a transcriptional repressor, however the gene expression changes observed in the hypothalamus of MeCP2 disorder mouse models suggest that MeCP2 can also upregulate gene expression, given that the majority of genes are downregulated upon loss of MeCP2 and upregulated in its presence. To determine if this dual role of MeCP2 extends beyond the hypothalamus, we studied gene expression patterns in the cerebellum of Mecp2-null and MECP2-Tg mice, modeling RTT and MECP2 duplication syndrome, respectively. We found that abnormal MeCP2 dosage causes alterations in the expression of hundreds of genes in the cerebellum. The majority of genes were upregulated in MECP2-Tg mice and downregulated in Mecp2-null mice, consistent with a role for MeCP2 as a modulator that can both increase and decrease gene expression. Interestingly, many of the genes altered in the cerebellum, particularly those increased by the presence of MeCP2 and decreased in its absence, were similarly altered in the hypothalamus. Our data suggest that either gain or loss of MeCP2 results in gene expression changes in multiple brain regions and that some of these changes are global. Further delineation of the expression pattern of MeCP2 target genes throughout the brain might identify subsets of genes that are more amenable to manipulation, and can thus be used to modulate some of the disease phenotypes.


Asunto(s)
Cerebelo/metabolismo , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Síndrome de Rett/genética , Animales , Modelos Animales de Enfermedad , Dosificación de Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Síndrome de Rett/metabolismo
19.
Afr J Health Sci ; 34(3): 364-373, 2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37920188

RESUMEN

BACKGROUND: Autism spectrum disorder (ASD) continues to climb in prevalence worldwide. Developed nations have focused on aligning their medical and research communities in order to investigate the mechanisms of pathogenesis, diagnosis, and societal impact of this disorder. A simultaneous rise of ASD has impacted developing nations, such as Ethiopia, without a commensurate ability to research the knowledge, beliefs, resources, and training regarding this condition in the country. MATERIALS AND METHODS: We administered a brief survey during a medical conference in Bahir Dar, Ethiopia, to investigate some of the education, information, and experiences with ASD within a small sample of medical and mental health providers in Ethiopia. RESULTS: The data provided insight into the following areas pertaining to ASD in Ethiopia: perceived causes, knowledge, training, and areas of need. CONCLUSIONS: Understanding local beliefs for causes and cures, as well as gaining indigenous opinions regarding what is needed for ASD education and resources in their nation, is the first step towards understanding the impact of this disorder and the approach to its treatment in Ethiopia.

20.
Sci Rep ; 10(1): 14045, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32820185

RESUMEN

More than 98% of the human genome is made up of non-coding DNA, but techniques to ascertain its contribution to human disease have lagged far behind our understanding of protein coding variations. Autism spectrum disorder (ASD) has been mostly associated with coding variations via de novo single nucleotide variants (SNVs), recessive/homozygous SNVs, or de novo copy number variants (CNVs); however, most ASD cases continue to lack a genetic diagnosis. We analyzed 187 consanguineous ASD families for biallelic CNVs. Recessive deletions were significantly enriched in affected individuals relative to their unaffected siblings (17% versus 4%, p < 0.001). Only a small subset of biallelic deletions were predicted to result in coding exon disruption. In contrast, biallelic deletions in individuals with ASD were enriched for overlap with regulatory regions, with 23/28 CNVs disrupting histone peaks in ENCODE (p < 0.009). Overlap with regulatory regions was further demonstrated by comparisons to the 127-epigenome dataset released by the Roadmap Epigenomics project, with enrichment for enhancers found in primary brain tissue and neuronal progenitor cells. Our results suggest a novel noncoding mechanism of ASD, describe a powerful method to identify important noncoding regions in the human genome, and emphasize the potential significance of gene activation and regulation in cognitive and social function.


Asunto(s)
Trastorno del Espectro Autista/genética , Epigénesis Genética , Eliminación de Gen , Homocigoto , Variaciones en el Número de Copia de ADN , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA