Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 23(4): 993-1000, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28533516

RESUMO

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.


Assuntos
Transtorno do Espectro Autista/genética , Cromossomos Humanos Par 17 , Comportamento Compulsivo/genética , Adolescente , Adulto , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/psicologia , Encéfalo/metabolismo , Criança , Pré-Escolar , Feminino , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla/métodos , Idade Gestacional , Proteínas de Homeodomínio/genética , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Transporte de Monossacarídeos/genética , Herança Multifatorial , Proteínas Nucleares/genética , Polimorfismo de Nucleotídeo Único , Proibitinas , Locos de Características Quantitativas , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Transcriptoma
2.
Mol Psychiatry ; 22(8): 1134-1139, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-27843152

RESUMO

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.


Assuntos
Transtorno do Espectro Autista/genética , Receptores de Ocitocina/genética , Adolescente , Alelos , Transtorno Autístico/genética , Encéfalo , Estudos de Casos e Controles , Criança , Feminino , Lobo Frontal , Dosagem de Genes/genética , Frequência do Gene/genética , Variação Genética , Humanos , Masculino , Neuroimagem/métodos , Núcleo Accumbens/fisiopatologia , Ocitocina/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Receptores de Ocitocina/metabolismo , Recompensa , Comportamento Social
3.
Mol Psychiatry ; 20(9): 1069-78, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25311365

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Células Dendríticas/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/patologia , Células Cultivadas , Cromossomos Humanos Par 15 , Variações do Número de Cópias de DNA , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Espinhas Dendríticas/genética , Espinhas Dendríticas/patologia , Feminino , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Regulação para Cima
4.
Mol Psychiatry ; 20(1): 118-25, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25199916

RESUMO

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.


Assuntos
Transtorno Autístico/patologia , Encéfalo/patologia , Modelos Animais de Doenças , Família Multigênica/genética , Animais , Transtorno Autístico/genética , Humanos , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos
5.
Mol Psychiatry ; 18(2): 226-35, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22105621

RESUMO

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.


Assuntos
Transtornos da Comunicação/genética , Predisposição Genética para Doença , Fator de Crescimento Neural/genética , Comunicação não Verbal/fisiologia , Característica Quantitativa Herdável , Criança , Transtornos Globais do Desenvolvimento Infantil/complicações , Transtornos Globais do Desenvolvimento Infantil/genética , Transtornos da Comunicação/etiologia , Saúde da Família , Feminino , Seguimentos , Estudos de Associação Genética , Ligação Genética , Genótipo , Humanos , Masculino , Canais de Potássio Shal/genética
6.
Mol Psychiatry ; 18(10): 1090-5, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23044707

RESUMO

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.


Assuntos
Transtornos Globais do Desenvolvimento Infantil/genética , Dosagem de Genes , Transtorno Autístico/epidemiologia , Transtorno Autístico/genética , Causalidade , Transtornos Globais do Desenvolvimento Infantil/epidemiologia , Anormalidades Congênitas/epidemiologia , Anormalidades Congênitas/genética , Mineração de Dados , Deficiências do Desenvolvimento/epidemiologia , Deficiências do Desenvolvimento/genética , Deleção de Genes , Duplicação Gênica , Estudos de Associação Genética , Heterogeneidade Genética , Predisposição Genética para Doença , Recombinação Homóloga , Humanos , Prevalência , Tamanho da Amostra
7.
Curr Opin Neurobiol ; 86: 102858, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38547564

RESUMO

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.


Assuntos
Transtorno do Espectro Autista , Genômica , Humanos , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/patologia , Transtorno do Espectro Autista/metabolismo , Genômica/métodos , Animais , Encéfalo/patologia , Encéfalo/metabolismo
8.
Mol Psychiatry ; 17(2): 202-14, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21647150

RESUMO

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.


Assuntos
Transtorno Autístico/genética , Transtorno Autístico/patologia , Regulação da Expressão Gênica no Desenvolvimento/genética , Genômica , Modelos Genéticos , Neurônios/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Feto , Perfilação da Expressão Gênica , Genótipo , Idade Gestacional , Humanos , Antígeno Ki-67/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos
9.
Mol Psychiatry ; 17(1): 62-70, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21079609

RESUMO

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.


Assuntos
Transtornos Globais do Desenvolvimento Infantil , Haploinsuficiência/genética , Imunidade Ativa/fisiologia , Complicações na Gravidez/fisiopatologia , Comportamento Social , Proteínas Supressoras de Tumor/deficiência , Fatores Etários , Animais , Animais Recém-Nascidos , Comportamento Animal , Transtornos Globais do Desenvolvimento Infantil/etiologia , Transtornos Globais do Desenvolvimento Infantil/genética , Transtornos Globais do Desenvolvimento Infantil/imunologia , Modelos Animais de Doenças , Embrião de Mamíferos , Comportamento Exploratório , Feminino , Humanos , Imunidade Ativa/efeitos dos fármacos , Recém-Nascido , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Poli I-C/efeitos adversos , Gravidez , Complicações na Gravidez/induzido quimicamente , Complicações na Gravidez/imunologia , Complicações na Gravidez/mortalidade , Proteína 2 do Complexo Esclerose Tuberosa
10.
Mol Psychiatry ; 15(10): 996-1005, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19455149

RESUMO

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.


Assuntos
Transtorno Autístico/genética , Canais de Cálcio Tipo T/genética , Cromossomos Humanos Par 17 , Polimorfismo de Nucleotídeo Único , Transtorno Autístico/epidemiologia , Criança , Feminino , Seguimentos , Dosagem de Genes , Marcadores Genéticos , Predisposição Genética para Doença/epidemiologia , Haplótipos , Humanos , Desequilíbrio de Ligação , Escore Lod , Masculino , Fatores de Risco
14.
Physiol Genomics ; 38(3): 362-71, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19531577

RESUMO

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.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Interferon Tipo I/farmacologia , Adulto , Linfócitos T CD4-Positivos/citologia , Células Cultivadas , Análise por Conglomerados , Redes Reguladoras de Genes , Humanos , Interferon alfa-2 , Interferon beta-1a , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Modelos Genéticos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas/genética , Interferência de RNA , Proteínas Recombinantes , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Mol Psychiatry ; 13(3): 285-92, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17968353

RESUMO

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.


Assuntos
Células-Tronco Adultas/efeitos dos fármacos , Antipsicóticos/farmacologia , Proliferação de Células/efeitos dos fármacos , Hipocampo/citologia , Lítio/farmacologia , Proteínas Wnt/metabolismo , Animais , Bromodesoxiuridina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Inositol/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Ratos , Ratos Endogâmicos F344 , Transdução de Sinais/efeitos dos fármacos , Transfecção/métodos , beta Catenina/genética , beta Catenina/metabolismo
16.
Science ; 282(5395): 1914-7, 1998 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-9836646

RESUMO

Tau proteins aggregate as cytoplasmic inclusions in a number of neurodegenerative diseases, including Alzheimer's disease and hereditary frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17). Over 10 exonic and intronic mutations in the tau gene have been identified in about 20 FTDP-17 families. Analyses of soluble and insoluble tau proteins from brains of FTDP-17 patients indicated that different pathogenic mutations differentially altered distinct biochemical properties and stoichiometry of brain tau isoforms. Functional assays of recombinant tau proteins with different FTDP-17 missense mutations implicated all but one of these mutations in disease pathogenesis by reducing the ability of tau to bind microtubules and promote microtubule assembly.


Assuntos
Encéfalo/metabolismo , Demência/genética , Microtúbulos/metabolismo , Doença de Parkinson Secundária/genética , Proteínas tau/genética , Proteínas tau/metabolismo , Processamento Alternativo , Cerebelo/metabolismo , Cromossomos Humanos Par 17 , Demência/metabolismo , Lobo Frontal/metabolismo , Humanos , Mutação , Mutação de Sentido Incorreto , Doença de Parkinson Secundária/metabolismo , Fosforilação , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Recombinantes/metabolismo , Solubilidade , Síndrome , Proteínas tau/química
17.
Neuron ; 29(2): 325-39, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11239426

RESUMO

Genetic mechanisms regulating CNS progenitor function and differentiation are not well understood. We have used microarrays derived from a representational difference analysis (RDA) subtraction in a heterogeneous stem cell culture system to systematically study the gene expression patterns of CNS progenitors. This analysis identified both known and novel genes enriched in progenitor cultures. In situ hybridization in a subset of clones demonstrated that many of these genes were expressed preferentially in germinal zones, some showing distinct ventricular or subventricular zone labeling. Several genes were also enriched in hematopoietic stem cells, suggesting an overlap of gene expression in neural and hematopoietic progenitors. This combination of methods demonstrates the power of using custom microarrays derived from RDA-subtracted libraries for both gene discovery and gene expression analysis in the central nervous system.


Assuntos
Diferenciação Celular/fisiologia , Córtex Cerebral/metabolismo , Perfilação da Expressão Gênica/métodos , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Northern Blotting/métodos , Células Cultivadas , Córtex Cerebral/citologia , Células-Tronco Hematopoéticas/metabolismo , Hibridização In Situ/métodos , Camundongos , Dados de Sequência Molecular , Neurônios/citologia
18.
J Mol Neurosci ; 33(2): 151-4, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17917073

RESUMO

Basal ganglia calcification (striatopallidodentate calcifications) can be caused by several systemic and neurological disorders. Familial Idiopathic Basal Ganglia Calcification (IBGC, "Fahr's disease"), is characterized by basal ganglia and extrabasal ganglia calcifications, parkinsonism and neuropsychiatric symptoms. Because of an increased use of neuroimaging procedures, calcifications of the basal ganglia are visualized more often and precociously. In 1999, a major American family with IBGC was linked to a locus on chromosome 14q (IBGC1). Another small kindred, from Spain, has also been reported as possibly linked to this locus. Here we report the main findings of the first 30 candidate genes sequenced at the IBGC1 locus during the process of searching for a mutation responsible for familial IBGC. During the sequencing process, we identified a heterozygous nonsynonymous single nucleotide polymorphism (exon 20 of the MGEA6/c-TAGE gene) shared by the affected and not present in the controls. This SNP was randomly screened in the general population (348 chromosomes) in a minor allele frequency to 0.0058 (two heterozygous among 174 subjects). Another variation in this gene, in the exon 9, was found in the Spanish family. However, this variation was extremely common in the general population. Functional and population studies are necessary to fully access the implications of the MGEA6 gene in familial IBGC, and a complete sequencing of the IBGC1 locus will be necessary to define a gene responsible for familial IBGC.


Assuntos
Doenças dos Gânglios da Base , Calcinose , Doenças dos Gânglios da Base/genética , Doenças dos Gânglios da Base/patologia , Calcinose/genética , Calcinose/patologia , Análise Mutacional de DNA , Ligação Genética , Predisposição Genética para Doença , Humanos , Mutação , Polimorfismo de Nucleotídeo Único
19.
J Neurosci ; 25(46): 10773-85, 2005 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-16291951

RESUMO

We performed genomic subtraction coupled to microarray-based gene expression profiling and identified the PDZ (postsynaptic density-95/Discs large/zona occludens-1)-binding kinase/T-LAK (lymphokine-activated killer T cell) cell originating protein kinase (PBK/TOPK) as a gene highly enriched in neural stem cell cultures. Previous studies have identified PBK/TOPK as a mitogen-activated protein kinase (MAPK) kinase that phosphorylated P38 MAPK but with no known expression or function in the nervous system. First, using a novel, bioinformatics-based approach to assess cross-correlation in large microarray datasets, we generated the hypothesis of a cell-cycle-related role for PBK/TOPK in neural cells. We then demonstrated that both PBK/TOPK and P38 are activated in a cell-cycle-dependent manner in neuronal progenitor cells in vitro, and inhibition of this pathway disrupts progenitor proliferation and self-renewal, a core feature of progenitors. In vivo, PBK/TOPK is expressed in rapidly proliferating cells in the adult subependymal zone (SEZ) and early postnatal cerebellar external granular layer. Using an approach based on transgenically targeted ablation and lineage tracing in mice, we show that PBK/TOPK-positive cells in the SEZ are GFAP negative but arise from GFAP-positive neural stem cells during adult neurogenesis. Furthermore, ablation of the adult stem cell population leads to concomitant loss of PBK/TOPK-positive cells in the SEZ. Together, these studies demonstrate that PBK/TOPK is a marker for transiently amplifying neural progenitors in the SEZ. Additionally, they suggest that PBK/TOPK plays an important role in these progenitors, and further implicates the P38 MAPK pathway in general, as an important regulator of progenitor proliferation and self-renewal.


Assuntos
Proliferação de Células , Sistema de Sinalização das MAP Quinases/fisiologia , Neurônios/citologia , Neurônios/enzimologia , Proteínas Serina-Treonina Quinases/fisiologia , Células-Tronco/citologia , Células-Tronco/enzimologia , Animais , Bovinos , Células Cultivadas , Galinhas , Cães , Humanos , Camundongos , Camundongos Transgênicos , Quinases de Proteína Quinase Ativadas por Mitógeno , Oncorhynchus mykiss , Pan troglodytes , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Ratos , Tetraodontiformes , Xenopus laevis , Peixe-Zebra , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
20.
Genes Brain Behav ; 5 Suppl 1: 54-63, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16417618

RESUMO

Language is a defining characteristic of our species that has emerged quite recently on an evolutionary timescale. Understanding the neurobiological substrates and genetic underpinnings of language constitutes a basic challenge for both neuroscience and genetics. The functional localization of language in the brain has been progressively refined over the last century through studies of aphasics and more recently through neuroimaging. Concurrently, structural specializations in these brain regions have been identified by virtue of their lateralization in humans and also through comparisons with homologous brain regions in non-human primate species. Comparative genomics has revealed the genome of our closest living relative, the chimpanzee, to be astonishingly similar to our own. To explore the role that changes in the regulation of gene expression have had in recent human evolution, several groups have used microarrays to compare expression levels for thousands of genes in the brain between humans and chimpanzees. By applying this approach to the increasingly refined peri-sylvian network of brain regions involved in language, it may be possible to discern functionally significant changes in gene expression that are universal among humans but unique to our species, thus casting light on the molecular basis of language in the brain.


Assuntos
Lateralidade Funcional/genética , Perfilação da Expressão Gênica , Idioma , Análise de Sequência com Séries de Oligonucleotídeos , Animais , Encéfalo/citologia , Encéfalo/fisiologia , Feminino , Genômica/métodos , Humanos , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA