Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Curr Opin Neurobiol ; 86: 102858, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38547564

RESUMEN

Advances in autism spectrum disorder (ASD) genetics have identified many genetic causes, reflecting remarkable progress while at the same time identifying challenges such as heterogeneity and pleiotropy, which complicate attempts to connect genetic risk to mechanisms. High-throughput functional genomic approaches have yielded progress by defining a molecular pathology in the brain of individuals with ASD and in identifying convergent biological pathways through which risk genes are predicted to act. These studies indicate that ASD genetic risk converges in early brain development, primarily during the period of cortical neurogenesis. Over development, genetic perturbations in turn lead to broad neuronal signaling dysregulation, most prominent in glutamatergic cortical-cortical projecting neurons and somatostatin positive interneurons, which is accompanied by glial dyshomeostasis throughout the cerebral cortex. Connecting these developmental perturbations to disrupted neuronal and glial function in the postnatal brain is an important direction in current research. Coupling functional genomic approaches with advances in induced pluripotent stem cell-derived neural organoid development provides a promising avenue for connecting brain pathology to developmental mechanisms.


Asunto(s)
Trastorno del Espectro Autista , Genómica , Humanos , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/patología , Trastorno del Espectro Autista/metabolismo , Genómica/métodos , Animales , Encéfalo/patología , Encéfalo/metabolismo
2.
Mol Psychiatry ; 23(4): 993-1000, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-28533516

RESUMEN

Autism spectrum disorder (ASD) is a behaviorally defined condition that manifests in infancy or early childhood as deficits in communication skills and social interactions. Often, restricted and repetitive behaviors (RRBs) accompany this disorder. ASD is polygenic and genetically complex, so we hypothesized that focusing analyses on intermediate core component phenotypes, such as RRBs, can reduce genetic heterogeneity and improve statistical power. Applying this approach, we mined Caucasian genome-wide association studies (GWAS) data from two of the largest ASD family cohorts, the Autism Genetics Resource Exchange and Autism Genome Project (AGP). Of the 12 RRBs measured by the Autism Diagnostic Interview-Revised, seven were found to be significantly familial and substantially variable, and hence, were tested for genome-wide association in 3104 ASD-affected children from 2045 families. Using a stringent significance threshold (P<7.1 × 10-9), GWAS in the AGP revealed an association between 'the degree of the repetitive use of objects or interest in parts of objects' and rs2898883 (P<6.8 × 10-9), which resides within the sixth intron of PHB. To identify the candidate target genes of the associated single-nucleotide polymorphisms at that locus, we applied chromosome conformation studies in developing human brains and implicated three additional genes: SLC35B1, CALCOCO2 and DLX3. Gene expression, brain imaging and fetal brain expression quantitative trait locus studies prioritize SLC35B1 and PHB. These analyses indicate that GWAS of single heritable features of genetically complex disorders followed by chromosome conformation studies in relevant tissues can be successful in revealing novel risk genes for single core features of ASD.


Asunto(s)
Trastorno del Espectro Autista/genética , Cromosomas Humanos Par 17 , Conducta Compulsiva/genética , Adolescente , Adulto , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/psicología , Encéfalo/metabolismo , Niño , Preescolar , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo/métodos , Edad Gestacional , Proteínas de Homeodominio/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Transporte de Monosacáridos/genética , Herencia Multifactorial , Proteínas Nucleares/genética , Polimorfismo de Nucleótido Simple , Prohibitinas , Sitios de Carácter Cuantitativo , Proteínas Represoras/genética , Factores de Transcripción/genética , Transcriptoma
3.
Mol Psychiatry ; 22(8): 1134-1139, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27843152

RESUMEN

Several common alleles in the oxytocin receptor gene (OXTR) are associated with altered brain function in reward circuitry in neurotypical adults and may increase risk for autism spectrum disorders (ASD). Yet, it is currently unknown how variation in the OXTR relates to brain functioning in individuals with ASD, and, critically, whether neural endophenotypes vary as a function of aggregate genetic risk. Here, for we believe the first time, we use a multi-locus approach to examine how genetic variation across several OXTR single-nucleotide polymorphisms (SNPs) affect functional connectivity of the brain's reward network. Using data from 41 children with ASD and 41 neurotypical children, we examined functional connectivity of the nucleus accumbens (NAcc) - a hub of the reward network - focusing on how connectivity varies with OXTR risk-allele dosage. Youth with ASD showed reduced NAcc connectivity with other areas in the reward circuit as a function of increased OXTR risk-allele dosage, as well as a positive association between risk-allele dosage and symptom severity, whereas neurotypical youth showed increased NAcc connectivity with frontal brain regions involved in mentalizing. In addition, we found that increased NAcc-frontal cortex connectivity in typically developing youth was related to better scores on a standardized measure of social functioning. Our results indicate that cumulative genetic variation on the OXTR impacts reward system connectivity in both youth with ASD and neurotypical controls. By showing differential genetic effects on neuroendophenotypes, these pathways elucidate mechanisms of vulnerability versus resilience in carriers of disease-associated risk alleles.


Asunto(s)
Trastorno del Espectro Autista/genética , Receptores de Oxitocina/genética , Adolescente , Alelos , Trastorno Autístico/genética , Encéfalo , Estudios de Casos y Controles , Niño , Femenino , Lóbulo Frontal , Dosificación de Gen/genética , Frecuencia de los Genes/genética , Variación Genética , Humanos , Masculino , Neuroimagen/métodos , Núcleo Accumbens/fisiopatología , Oxitocina/metabolismo , Polimorfismo de Nucleótido Simple/genética , Receptores de Oxitocina/metabolismo , Recompensa , Conducta Social
4.
Transl Psychiatry ; 6(8): e864, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27483382

RESUMEN

Genes for autism spectrum disorders (ASDs) are also implicated in fragile X syndrome (FXS), intellectual disabilities (ID) or schizophrenia (SCZ), and converge on neuronal function and differentiation. The SH-SY5Y neuroblastoma cell line, the most widely used system to study neurodevelopment, is currently discussed for its applicability to model cortical development. We implemented an optimal neuronal differentiation protocol of this system and evaluated neurodevelopment at the transcriptomic level using the CoNTeXT framework, a machine-learning algorithm based on human post-mortem brain data estimating developmental stage and regional identity of transcriptomic signatures. Our improved model in contrast to currently used SH-SY5Y models does capture early neurodevelopmental processes with high fidelity. We applied regression modelling, dynamic time warping analysis, parallel independent component analysis and weighted gene co-expression network analysis to identify activated gene sets and networks. Finally, we tested and compared these sets for enrichment of risk genes for neuropsychiatric disorders. We confirm a significant overlap of genes implicated in ASD with FXS, ID and SCZ. However, counterintuitive to this observation, we report that risk genes affect pathways specific for each disorder during early neurodevelopment. Genes implicated in ASD, ID, FXS and SCZ were enriched among the positive regulators, but only ID-implicated genes were also negative regulators of neuronal differentiation. ASD and ID genes were involved in dendritic branching modules, but only ASD risk genes were implicated in histone modification or axonal guidance. Only ID genes were over-represented among cell cycle modules. We conclude that the underlying signatures are disorder-specific and that the shared genetic architecture results in overlaps across disorders such as ID in ASD. Thus, adding developmental network context to genetic analyses will aid differentiating the pathophysiology of neuropsychiatric disorders.


Asunto(s)
Trastorno del Espectro Autista/genética , Síndrome del Cromosoma X Frágil/genética , Regulación del Desarrollo de la Expresión Génica , Discapacidad Intelectual/genética , Neurogénesis/genética , Esquizofrenia/genética , Transcriptoma , Algoritmos , Encéfalo/crecimiento & desarrollo , Línea Celular Tumoral , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Humanos , Aprendizaje Automático , Plasticidad Neuronal/genética , ARN Mensajero/metabolismo , Análisis de Regresión
6.
Mol Psychiatry ; 20(1): 118-25, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25199916

RESUMEN

Autism is a heritable disorder, with over 250 associated genes identified to date, yet no single gene accounts for >1-2% of cases. The clinical presentation, behavioural symptoms, imaging and histopathology findings are strikingly heterogeneous. A more complete understanding of autism can be obtained by examining multiple genetic or behavioural mouse models of autism using magnetic resonance imaging (MRI)-based neuroanatomical phenotyping. Twenty-six different mouse models were examined and the consistently found abnormal brain regions across models were parieto-temporal lobe, cerebellar cortex, frontal lobe, hypothalamus and striatum. These models separated into three distinct clusters, two of which can be linked to the under and over-connectivity found in autism. These clusters also identified previously unknown connections between Nrxn1α, En2 and Fmr1; Nlgn3, BTBR and Slc6A4; and also between X monosomy and Mecp2. With no single treatment for autism found, clustering autism using neuroanatomy and identifying these strong connections may prove to be a crucial step in predicting treatment response.


Asunto(s)
Trastorno Autístico/patología , Encéfalo/patología , Modelos Animales de Enfermedad , Familia de Multigenes/genética , Animales , Trastorno Autístico/genética , Humanos , Procesamiento de Imagen Asistido por Computador , Imagen por Resonancia Magnética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos
7.
Mol Psychiatry ; 20(9): 1069-78, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25311365

RESUMEN

Rare maternally inherited duplications at 15q11-13 are observed in ~1% of individuals with an autism spectrum disorder (ASD), making it among the most common causes of ASD. 15q11-13 comprises a complex region, and as this copy number variation encompasses many genes, it is important to explore individual genotype-phenotype relationships. Cytoplasmic FMR1-interacting protein 1 (CYFIP1) is of particular interest because of its interaction with Fragile X mental retardation protein (FMRP), its upregulation in transformed lymphoblastoid cell lines from patients with duplications at 15q11-13 and ASD and the presence of smaller overlapping deletions of CYFIP1 in patients with schizophrenia and intellectual disability. Here, we confirm that CYFIP1 is upregulated in transformed lymphoblastoid cell lines and demonstrate its upregulation in the post-mortem brain from 15q11-13 duplication patients for the first time. To investigate how increased CYFIP1 dosage might predispose to neurodevelopmental disease, we studied the consequence of its overexpression in multiple systems. We show that overexpression of CYFIP1 results in morphological abnormalities including cellular hypertrophy in SY5Y cells and differentiated mouse neuronal progenitors. We validate these results in vivo by generating a BAC transgenic mouse, which overexpresses Cyfip1 under the endogenous promotor, observing an increase in the proportion of mature dendritic spines and dendritic spine density. Gene expression profiling on embryonic day 15 suggested the dysregulation of mammalian target of rapamycin (mTOR) signaling, which was confirmed at the protein level. Importantly, similar evidence of mTOR-related dysregulation was seen in brains from 15q11-13 duplication patients with ASD. Finally, treatment of differentiated mouse neuronal progenitors with an mTOR inhibitor (rapamycin) rescued the morphological abnormalities resulting from CYFIP1 overexpression. Together, these data show that CYFIP1 overexpression results in specific cellular phenotypes and implicate modulation by mTOR signaling, further emphasizing its role as a potential convergent pathway in some forms of ASD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Células Dendríticas/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/patología , Células Cultivadas , Cromosomas Humanos Par 15 , Variaciones en el Número de Copia de ADN , Células Dendríticas/metabolismo , Células Dendríticas/patología , Espinas Dendríticas/genética , Espinas Dendríticas/patología , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Regulación hacia Arriba
10.
Mol Psychiatry ; 18(2): 226-35, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22105621

RESUMEN

Autism Spectrum Disorder (ASD) has a heterogeneous etiology that is genetically complex. It is defined by deficits in communication and social skills and the presence of restricted and repetitive behaviors. Genetic analyses of heritable quantitative traits that correlate with ASD may reduce heterogeneity. With this in mind, deficits in nonverbal communication (NVC) were quantified based on items from the Autism Diagnostic Interview Revised. Our previous analysis of 228 families from the Autism Genetics Research Exchange (AGRE) repository reported 5 potential quantitative trait loci (QTL). Here we report an NVC QTL replication study in an independent sample of 213 AGRE families. One QTL was replicated (P<0.0004). It was investigated using a targeted-association analysis of 476 haplotype blocks with 708 AGRE families using the Family Based Association Test (FBAT). Blocks in two QTL genes were associated with NVC with a P-value of 0.001. Three associated haplotype blocks were intronic to the Nerve Growth Factor (NGF) gene (P=0.001, 0.001, 0.002), and one was intronic to KCND3 (P=0.001). Individual haplotypes within the associated blocks drove the associations (0.003, 0.0004 and 0.0002) for NGF and 0.0001 for KCND3. Using the same methods, these genes were tested for association with NVC in an independent sample of 1517 families from an Autism Genome Project (AGP). NVC was associated with a haplotype in an adjacent NGF block (P=0.0005) and one 46 kb away from the associated block in KCND3 (0.008). These analyses illustrate the value of QTL and targeted association studies for genetically complex disorders such as ASD. NGF is a promising risk gene for NVC deficits.


Asunto(s)
Trastornos de la Comunicación/genética , Predisposición Genética a la Enfermedad , Factor de Crecimiento Nervioso/genética , Comunicación no Verbal/fisiología , Carácter Cuantitativo Heredable , Niño , Trastornos Generalizados del Desarrollo Infantil/complicaciones , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos de la Comunicación/etiología , Salud de la Familia , Femenino , Estudios de Seguimiento , Estudios de Asociación Genética , Ligamiento Genético , Genotipo , Humanos , Masculino , Canales de Potasio Shal/genética
11.
Mol Psychiatry ; 18(10): 1090-5, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23044707

RESUMEN

Copy number variants (CNVs) have a major role in the etiology of autism spectrum disorders (ASD), and several of these have reached statistical significance in case-control analyses. Nevertheless, current ASD cohorts are not large enough to detect very rare CNVs that may be causative or contributory (that is, risk alleles). Here, we use a tiered approach, in which clinically significant CNVs are first identified in large clinical cohorts of neurodevelopmental disorders (including but not specific to ASD), after which these CNVs are then systematically identified within well-characterized ASD cohorts. We focused our initial analysis on 48 recurrent CNVs (segmental duplication-mediated 'hotspots') from 24 loci in 31 516 published clinical cases with neurodevelopmental disorders and 13 696 published controls, which yielded a total of 19 deletion CNVs and 11 duplication CNVs that reached statistical significance. We then investigated the overlap of these 30 CNVs in a combined sample of 3955 well-characterized ASD cases from three published studies. We identified 73 deleterious recurrent CNVs, including 36 deletions from 11 loci and 37 duplications from seven loci, for a frequency of 1 in 54; had we considered the ASD cohorts alone, only 58 CNVs from eight loci (24 deletions from three loci and 34 duplications from five loci) would have reached statistical significance. In conclusion, until there are sufficiently large ASD research cohorts with enough power to detect very rare causative or contributory CNVs, data from larger clinical cohorts can be used to infer the likely clinical significance of CNVs in ASD.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil/genética , Dosificación de Gen , Trastorno Autístico/epidemiología , Trastorno Autístico/genética , Causalidad , Trastornos Generalizados del Desarrollo Infantil/epidemiología , Anomalías Congénitas/epidemiología , Anomalías Congénitas/genética , Minería de Datos , Discapacidades del Desarrollo/epidemiología , Discapacidades del Desarrollo/genética , Eliminación de Gen , Duplicación de Gen , Estudios de Asociación Genética , Heterogeneidad Genética , Predisposición Genética a la Enfermedad , Recombinación Homóloga , Humanos , Prevalencia , Tamaño de la Muestra
12.
Mol Psychiatry ; 17(1): 62-70, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21079609

RESUMEN

Approximately 40-50% of individuals affected by tuberous sclerosis (TSC) develop autism spectrum disorders (ASDs). One possible explanation for this partial penetrance is an interaction between TSC gene mutations and other risk factors such as gestational immune activation. In this study, we report the interactive effects of these two ASD risk factors in a mouse model of TSC. Combined, but not single, exposure had adverse effects on intrauterine survival. Additionally, provisional results suggest that these factors synergize to disrupt social approach behavior in adult mice. Moreover, studies in human populations are consistent with an interaction between high seasonal flu activity in late gestation and TSC mutations in ASD. Taken together, our studies raise the possibility of a gene × environment interaction between heterozygous TSC gene mutations and gestational immune activation in the pathogenesis of TSC-related ASD.


Asunto(s)
Trastornos Generalizados del Desarrollo Infantil , Haploinsuficiencia/genética , Inmunidad Activa/fisiología , Complicaciones del Embarazo/fisiopatología , Conducta Social , Proteínas Supresoras de Tumor/deficiencia , Factores de Edad , Animales , Animales Recién Nacidos , Conducta Animal , Trastornos Generalizados del Desarrollo Infantil/etiología , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/inmunología , Modelos Animales de Enfermedad , Embrión de Mamíferos , Conducta Exploratoria , Femenino , Humanos , Inmunidad Activa/efectos de los fármacos , Recién Nacido , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Poli I-C/efectos adversos , Embarazo , Complicaciones del Embarazo/inducido químicamente , Complicaciones del Embarazo/inmunología , Complicaciones del Embarazo/mortalidad , Proteína 2 del Complejo de la Esclerosis Tuberosa
13.
Mol Psychiatry ; 17(2): 202-14, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21647150

RESUMEN

Human neural progenitors from a variety of sources present new opportunities to model aspects of human neuropsychiatric disease in vitro. Such in vitro models provide the advantages of a human genetic background combined with rapid and easy manipulation, making them highly useful adjuncts to animal models. Here, we examined whether a human neuronal culture system could be utilized to assess the transcriptional program involved in human neural differentiation and to model some of the molecular features of a neurodevelopmental disorder, such as autism. Primary normal human neuronal progenitors (NHNPs) were differentiated into a post-mitotic neuronal state through addition of specific growth factors and whole-genome gene expression was examined throughout a time course of neuronal differentiation. After 4 weeks of differentiation, a significant number of genes associated with autism spectrum disorders (ASDs) are either induced or repressed. This includes the ASD susceptibility gene neurexin 1, which showed a distinct pattern from neurexin 3 in vitro, and which we validated in vivo in fetal human brain. Using weighted gene co-expression network analysis, we visualized the network structure of transcriptional regulation, demonstrating via this unbiased analysis that a significant number of ASD candidate genes are coordinately regulated during the differentiation process. As NHNPs are genetically tractable and manipulable, they can be used to study both the effects of mutations in multiple ASD candidate genes on neuronal differentiation and gene expression in combination with the effects of potential therapeutic molecules. These data also provide a step towards better understanding of the signaling pathways disrupted in ASD.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/patología , Regulación del Desarrollo de la Expresión Génica/genética , Genómica , Modelos Genéticos , Neuronas/metabolismo , Diferenciación Celular/fisiología , Células Cultivadas , Feto , Perfilación de la Expresión Génica , Genotipo , Edad Gestacional , Humanos , Antígeno Ki-67/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos
14.
Neurology ; 76(5): 467-74, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21178100

RESUMEN

OBJECTIVES: To determine whether TMEM106B single nucleotide polymorphisms (SNPs) are associated with frontotemporal lobar degeneration (FTLD) in patients with and without mutations in progranulin (GRN) and to determine whether TMEM106B modulates GRN expression. METHODS: We performed a case-control study of 3 SNPs in TMEM106B in 482 patients with clinical and 80 patients with pathologic FTLD-TAR DNA-binding protein 43 without GRN mutations, 78 patients with FTLD with GRN mutations, and 822 controls. Association analysis of TMEM106B with GRN plasma levels was performed in 1,013 controls and TMEM106B and GRN mRNA expression levels were correlated in peripheral blood samples from 33 patients with FTLD and 150 controls. RESULTS: In our complete FTLD patient cohort, nominal significance was identified for 2 TMEM106B SNPs (top SNP rs1990622, p(allelic) = 0.036). However, the most significant association with risk of FTLD was observed in the subgroup of GRN mutation carriers compared to controls (corrected p(allelic) = 0.0009), where there was a highly significant decrease in the frequency of homozygote carriers of the minor alleles of all TMEM106B SNPs (top SNP rs1990622, CC genotype frequency 2.6% vs 19.1%, corrected p(recessive) = 0.009). We further identified a significant association of TMEM106B SNPs with plasma GRN levels in controls (top SNP rs1990622, corrected p = 0.002) and in peripheral blood samples a highly significant correlation was observed between TMEM106B and GRN mRNA expression in patients with FTLD (r = -0.63, p = 7.7 × 10(-5)) and controls (r = -0.49, p = 2.2 × 10(-10)). CONCLUSIONS: In our study, TMEM106B SNPs significantly reduced the disease penetrance in patients with GRN mutations, potentially by modulating GRN levels. These findings hold promise for the development of future protective therapies for FTLD.


Asunto(s)
Degeneración Lobar Frontotemporal/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Penetrancia , Polimorfismo de Nucleótido Simple/genética , Precursores de Proteínas/genética , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Degeneración Lobar Frontotemporal/sangre , Degeneración Lobar Frontotemporal/diagnóstico , Estudios de Asociación Genética , Tamización de Portadores Genéticos , Predisposición Genética a la Enfermedad/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Masculino , Proteínas de la Membrana/sangre , Persona de Mediana Edad , Proteínas del Tejido Nervioso/sangre , Progranulinas , Precursores de Proteínas/sangre
15.
Mol Psychiatry ; 15(10): 996-1005, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19455149

RESUMEN

Chromosome 17q11-q21 is a region of the genome likely to harbor susceptibility to autism (MIM(209850)) based on earlier evidence of linkage to the disorder. This linkage is specific to multiplex pedigrees containing only male probands (MO) within the Autism Genetic Resource Exchange (AGRE). Earlier, Stone et al.(1) completed a high-density single nucleotide polymorphism association study of 13.7 Mb within this interval, but common variant association was not sufficient to account for the linkage signal. Here, we extend this single nucleotide polymorphism-based association study to complete the coverage of the two-LOD support interval around the chromosome 17q linkage peak by testing the majority of common alleles in 284 MO trios. Markers within an interval containing the gene, CACNA1G, were found to be associated with Autism Spectrum Disorder at a locally significant level (P=1.9 × 10(-5)). While establishing CACNA1G as a novel candidate gene for autism, these alleles do not contribute a sufficient genetic effect to explain the observed linkage, indicating that there is substantial genetic heterogeneity despite the clear linkage signal. The region thus likely harbors a combination of multiple common and rare alleles contributing to the genetic risk. These data, along with earlier studies of chromosomes 5 and 7q3, suggest few if any major common risk alleles account for Autism Spectrum Disorder risk under major linkage peaks in the AGRE sample. This provides important evidence for strategies to identify Autism Spectrum Disorder genes, suggesting that they should focus on identifying rare variants and common variants of small effect.


Asunto(s)
Trastorno Autístico/genética , Canales de Calcio Tipo T/genética , Cromosomas Humanos Par 17 , Polimorfismo de Nucleótido Simple , Trastorno Autístico/epidemiología , Niño , Femenino , Estudios de Seguimiento , Dosificación de Gen , Marcadores Genéticos , Predisposición Genética a la Enfermedad/epidemiología , Haplotipos , Humanos , Desequilibrio de Ligamiento , Escala de Lod , Masculino , Factores de Riesgo
16.
Physiol Genomics ; 38(3): 362-71, 2009 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-19531577

RESUMEN

Type I interferons (IFNs) are pleiotropic cytokines that modulate both innate and adaptive immune responses. They have been used to treat autoimmune disorders, cancers, and viral infection and have been demonstrated to elicit differential responses within cells, despite sharing a single receptor. The molecular basis for such differential responses has remained elusive. To identify the mechanisms underlying differential type I IFN signaling, we used whole genome microarrays to measure longitudinal transcriptional events within human CD4(+) T cells treated with IFN-alpha(2b) or IFN-beta(1a). We identified differentially regulated genes, analyzed them for the enrichment of known promoter elements and pathways, and constructed a network module based on weighted gene coexpression network analysis (WGCNA). WGCNA uses advanced statistical measures to find interconnected modules of correlated genes. Overall, differential responses to IFN in CD4(+) T cells related to three dominant themes: migration, antigen presentation, and the cytotoxic response. For migration, WGCNA identified subtype-specific regulation of pre-mRNA processing factor 4 homolog B and eukaryotic translation initiation factor 4A2, which work at various levels within the cell to affect the expression of the chemokine CCL5. WGCNA also identified sterile alpha-motif domain-containing 9-like (SAMD9L) as critical in subtype-independent effects of IFN treatment. RNA interference of SAMD9L expression enhanced the migratory phenotype of activated T cells treated with IFN-beta compared with controls. Through the analysis of the dynamic transcriptional events after differential IFN treatment, we were able to identify specific signatures and to uncover novel genes that may underpin the type I IFN response.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Interferón Tipo I/farmacología , Adulto , Linfocitos T CD4-Positivos/citología , Células Cultivadas , Análisis por Conglomerados , Redes Reguladoras de Genes , Humanos , Interferón alfa-2 , Interferón beta-1a , Interferón-alfa/farmacología , Interferón beta/farmacología , Péptidos y Proteínas de Señalización Intracelular , Masculino , Modelos Genéticos , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas/genética , Interferencia de ARN , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Cell Death Differ ; 16(6): 807-25, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19282867

RESUMEN

Insights into early human development are fundamental for our understanding of human biology. Efficient differentiation of human embryonic stem cells (hESCs) into neural precursor cells is critical for future cell-based therapies. Here, using defined conditions, we characterized a new method for rapid and uniform differentiation of hESCs into committed neural precursor cells (designated C-NPCs). Dynamic gene expression analysis identified several distinct stages of ESC neuralization and revealed functional modules of coregulated genes and pathways. The first wave of gene expression changes, likely corresponding to the transition through primitive ectoderm, started at day 3, preceding the formation of columnar neuroepithelial rosettes. The second wave started at day 5, coinciding with the formation of rosettes. The majority of C-NPCs were positive for both anterior and posterior markers of developing neuroepithelium. In culture, C-NPCs became electrophysiologically functional neurons; on transplantation into neonatal mouse brains, C-NPCs integrated into the cortex and olfactory bulb, acquiring appropriate neuronal morphologies and markers. Compared to rosette-NPCs,(1) C-NPCs exhibited limited in vitro expansion capacity and did not express potent oncogenes such as PLAG1 or RSPO3. Concordantly, we never detected tumors or excessive neural proliferation after transplantation of C-NPCs into mouse brains. In conclusion, our study provides a framework for future analysis of molecular signaling during ESC neuralization.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Neuronas/citología , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Línea Celular , Ectodermo/metabolismo , Electrofisiología , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos ICR , Proteómica , Formación de Roseta , Trasplante Heterólogo
18.
Neurology ; 71(2): 85-92, 2008 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-18509095

RESUMEN

BACKGROUND: Persons at risk for familial Alzheimer disease (FAD) provide a model in which biomarkers can be studied in presymptomatic disease. METHODS: Twenty-one subjects at risk for presenilin-1 (n = 17) or amyloid precursor protein (n = 4) mutations underwent evaluation with the Clinical Dementia Rating (CDR) scale. We obtained plasma from all subjects and CSF from 11. Plasma (Abeta(40), Abeta(42), F(2)-isoprostanes) and CSF (F(2)-isoprostanes, t-tau, p-tau(181), Abeta(40), Abeta(42), and Abeta(42)/Abeta(40) ratio) levels were compared between FAD mutation carriers (MCs) and noncarriers (NCs). RESULTS: Plasma Abeta(42) levels (25.1 pM vs 15.5 pM, p = 0.031) and the ratio of Abeta(42)/Abeta(40) (0.16 vs 0.11, p = 0.045) were higher in presymptomatic MCs. Among MCs, those with CDR scores of 0.5 had lower plasma Abeta(42) levels than those with CDR scores of 0 (14.1 pM vs 25.1, p = 0.02). The ratio of Abeta(42) to Abeta(40) was also reduced in the CSF (0.08 vs 0.15, p = 0.046) of nondemented MCs compared to NCs. Total CSF tau and p-tau(181) levels were elevated in presymptomatic FAD MCs. CSF levels of F(2)-isoprostanes were also elevated in MCs (n = 7, 48.6 pg/mL) compared to NCs (n = 4, 21.6 pg/mL, p = 0.031). CONCLUSIONS: Our data indicate that Abeta(42) is elevated in plasma in familial Alzheimer disease (FAD) mutation carriers (MCs) and suggests that this level may decrease with disease progression prior to the development of overt dementia. We also demonstrated that the ratio of Abeta(42) to Abeta(40) was reduced in the CSF of nondemented MCs and that elevations of t-tau and p-tau(181) are sensitive indicators of presymptomatic disease. Our finding of elevated F(2)-isoprostane levels in the CSF of preclinical FAD MCs suggests that oxidative stress occurs downstream to mismetabolism of amyloid precursor protein.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/prevención & control , Heterocigoto , Adulto , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/líquido cefalorraquídeo , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Femenino , Humanos , Isoprostanos/sangre , Isoprostanos/líquido cefalorraquídeo , Masculino , Mutación , Examen Neurológico , Presenilina-1/genética , Nexinas de Proteasas , Receptores de Superficie Celular/genética , Sensibilidad y Especificidad , Proteínas tau/líquido cefalorraquídeo
19.
Mol Psychiatry ; 13(3): 285-92, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17968353

RESUMEN

Neural stem cells give rise to new hippocampal neurons throughout adulthood, and defects in neurogenesis may predispose an individual to mood disorders, such as major depression. Our understanding of the signals controlling this process is limited, so we explored potential pathways regulating adult hippocampal progenitor (AHP) proliferation and neuronal differentiation. We demonstrate that the mood stabilizer lithium directly expands pools of AHPs in vitro, and induces them to become neurons at therapeutically relevant concentrations. We show that these effects are independent of inositol monophosphatase, but dependent on Wnt pathway components. Both downregulation of glycogen synthase kinase-3beta, a lithium-sensitive component of the canonical Wnt signaling pathway, and elevated beta-catenin, a downstream component of the same pathway produce effects similar to lithium. In contrast, RNAi-mediated inhibition of beta-catenin abolishes the proliferative effects of lithium, suggesting that Wnt signal transduction may underlie lithium's therapeutic effect. Together, these data strengthen the connection between psychopharmacologic treatment and the process of adult neurogenesis, while also suggesting the pursuit of modulators of Wnt signaling as a new class of more effective mood stabilizers/antidepressants.


Asunto(s)
Células Madre Adultas/efectos de los fármacos , Antipsicóticos/farmacología , Proliferación Celular/efectos de los fármacos , Hipocampo/citología , Litio/farmacología , Proteínas Wnt/metabolismo , Animales , Bromodesoxiuridina/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inositol/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Ratas , Ratas Endogámicas F344 , Transducción de Señal/efectos de los fármacos , Transfección/métodos , beta Catenina/genética , beta Catenina/metabolismo
20.
Proc Natl Acad Sci U S A ; 104(45): 17849-54, 2007 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-17978184

RESUMEN

Despite the well established role of the frontal and posterior perisylvian cortices in many facets of human-cognitive specializations, including language, little is known about the developmental patterning of these regions in the human brain. We performed a genome-wide analysis of human cerebral patterning during midgestation, a critical epoch in cortical regionalization. A total of 345 genes were identified as differentially expressed between superior temporal gyrus (STG) and the remaining cerebral cortex. Gene ontology categories representing transcription factors were enriched in STG, whereas cell-adhesion and extracellular matrix molecules were enriched in the other cortical regions. Quantitative RT-PCR or in situ hybridization was performed to validate differential expression in a subset of 32 genes, most of which were confirmed. LIM domain-binding 1 (LDB1), which we show to be enriched in the STG, is a recently identified interactor of LIM domain only 4 (LMO4), a gene known to be involved in the asymmetric pattering of the perisylvian region in the developing human brain. Protocadherin 17 (PCDH17), a neuronal cell adhesion molecule, was highly enriched in focal regions of the human prefrontal cortex. Contactin associated protein-like 2 (CNTNAP2), in which mutations are known to cause autism, epilepsy, and language delay, showed a remarkable pattern of anterior-enriched cortical expression in human that was not observed in mouse or rat. These data highlight the importance of expression analysis of human brain and the utility of cross-species comparisons of gene expression. Genes identified here provide a foundation for understanding molecular aspects of human-cognitive specializations and the disorders that disrupt them.


Asunto(s)
Corteza Cerebral/embriología , Corteza Cerebral/fisiología , Cognición/fisiología , Genoma Humano , Conocimiento , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Embarazo , Segundo Trimestre del Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...