Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Nature ; 612(7939): 301-309, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36450978

RESUMO

Clonal haematopoiesis involves the expansion of certain blood cell lineages and has been associated with ageing and adverse health outcomes1-5. Here we use exome sequence data on 628,388 individuals to identify 40,208 carriers of clonal haematopoiesis of indeterminate potential (CHIP). Using genome-wide and exome-wide association analyses, we identify 24 loci (21 of which are novel) where germline genetic variation influences predisposition to CHIP, including missense variants in the lymphocytic antigen coding gene LY75, which are associated with reduced incidence of CHIP. We also identify novel rare variant associations with clonal haematopoiesis and telomere length. Analysis of 5,041 health traits from the UK Biobank (UKB) found relationships between CHIP and severe COVID-19 outcomes, cardiovascular disease, haematologic traits, malignancy, smoking, obesity, infection and all-cause mortality. Longitudinal and Mendelian randomization analyses revealed that CHIP is associated with solid cancers, including non-melanoma skin cancer and lung cancer, and that CHIP linked to DNMT3A is associated with the subsequent development of myeloid but not lymphoid leukaemias. Additionally, contrary to previous findings from the initial 50,000 UKB exomes6, our results in the full sample do not support a role for IL-6 inhibition in reducing the risk of cardiovascular disease among CHIP carriers. Our findings demonstrate that CHIP represents a complex set of heterogeneous phenotypes with shared and unique germline genetic causes and varied clinical implications.


Assuntos
COVID-19 , Doenças Cardiovasculares , Humanos , Hematopoiese Clonal/genética , Doenças Cardiovasculares/epidemiologia , Doenças Cardiovasculares/genética
3.
Nature ; 611(7936): 532-539, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36323788

RESUMO

Neuropsychiatric disorders classically lack defining brain pathologies, but recent work has demonstrated dysregulation at the molecular level, characterized by transcriptomic and epigenetic alterations1-3. In autism spectrum disorder (ASD), this molecular pathology involves the upregulation of microglial, astrocyte and neural-immune genes, the downregulation of synaptic genes, and attenuation of gene-expression gradients in cortex1,2,4-6. However, whether these changes are limited to cortical association regions or are more widespread remains unknown. To address this issue, we performed RNA-sequencing analysis of 725 brain samples spanning 11 cortical areas from 112 post-mortem samples from individuals with ASD and neurotypical controls. We find widespread transcriptomic changes across the cortex in ASD, exhibiting an anterior-to-posterior gradient, with the greatest differences in primary visual cortex, coincident with an attenuation of the typical transcriptomic differences between cortical regions. Single-nucleus RNA-sequencing and methylation profiling demonstrate that this robust molecular signature reflects changes in cell-type-specific gene expression, particularly affecting excitatory neurons and glia. Both rare and common ASD-associated genetic variation converge within a downregulated co-expression module involving synaptic signalling, and common variation alone is enriched within a module of upregulated protein chaperone genes. These results highlight widespread molecular changes across the cerebral cortex in ASD, extending beyond association cortex to broadly involve primary sensory regions.


Assuntos
Transtorno do Espectro Autista , Córtex Cerebral , Variação Genética , Transcriptoma , Humanos , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Neurônios/metabolismo , RNA/análise , RNA/genética , Transcriptoma/genética , Autopsia , Análise de Sequência de RNA , Córtex Visual Primário/metabolismo , Neuroglia/metabolismo
5.
Front Neurol ; 12: 728700, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34744969

RESUMO

The development of autoimmune antibody panels has improved the diagnosis of paraneoplastic neurological disorders (PNDs) of the brain and spinal cord. Here, we present a case of a woman with a history of breast cancer who presented with a subacute sensory ataxia that progressed over 18 months. Her examination and diagnostic studies were consistent with a myelopathy. Metabolic, infectious, and autoimmune testing were non-diagnostic. However, she responded to empirical immunosuppression, prompting further workup for an autoimmune etiology. An unbiased autoantibody screen utilizing phage display immunoprecipitation sequencing (PhIP-Seq) identified antibodies to the anti-Yo antigens cerebellar degeneration related protein 2 like (CDR2L) and CDR2, which were subsequently validated by immunoblot and cell-based overexpression assays. Furthermore, CDR2L protein expression was restricted to HER2 expressing tumor cells in the patient's breast tissue. Recent evidence suggests that CDR2L is likely the primary antigen in anti-Yo paraneoplastic cerebellar degeneration, but anti-Yo myelopathy is poorly characterized. By immunostaining, we detected neuronal CDR2L protein expression in the murine and human spinal cord. This case demonstrates the diagnostic utility of unbiased assays in patients with suspected PNDs, supports prior observations that anti-Yo PND can be associated with isolated myelopathy, and implicates CDR2L as a potential antigen in the spinal cord.

6.
Hum Mol Genet ; 28(13): 2201-2211, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31220268

RESUMO

Autism spectrum disorder (ASD) encompasses a collection of complex neuropsychiatric disorders characterized by deficits in social functioning, communication and repetitive behaviour. Building on recent studies supporting a role for developmentally moderated regulatory genomic variation in the molecular aetiology of ASD, we quantified genome-wide patterns of DNA methylation in 223 post-mortem tissues samples isolated from three brain regions [prefrontal cortex, temporal cortex and cerebellum (CB)] dissected from 43 ASD patients and 38 non-psychiatric control donors. We identified widespread differences in DNA methylation associated with idiopathic ASD (iASD), with consistent signals in both cortical regions that were distinct to those observed in the CB. Individuals carrying a duplication on chromosome 15q (dup15q), representing a genetically defined subtype of ASD, were characterized by striking differences in DNA methylationacross a discrete domain spanning an imprinted gene cluster within the duplicated region. In addition to the dramatic cis-effects on DNA methylation observed in dup15q carriers, we identified convergent methylomic signatures associated with both iASD and dup15q, reflecting the findings from previous studies of gene expression and H3K27ac. Cortical co-methylation network analysis identified a number of co-methylated modules significantly associated with ASD that are enriched for genomic regions annotated to genes involved in the immune system, synaptic signalling and neuronal regulation. Our study represents the first systematic analysis of DNA methylation associated with ASD across multiple brain regions, providing novel evidence for convergent molecular signatures associated with both idiopathic and syndromic autism.


Assuntos
Transtorno Autístico/genética , Cerebelo/metabolismo , Metilação de DNA , Córtex Pré-Frontal/metabolismo , Lobo Temporal/metabolismo , Transtorno Autístico/metabolismo , Estudos de Casos e Controles , Cerebelo/química , Epigenoma , Feminino , Ontologia Genética , Redes Reguladoras de Genes , Genoma Humano , Humanos , Sistema Imunitário/metabolismo , Masculino , Vias Neurais/fisiologia , Córtex Pré-Frontal/química , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Lobo Temporal/química
7.
J Child Psychol Psychiatry ; 60(5): 585-598, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30362171

RESUMO

BACKGROUND: Williams syndrome (WS) is a neurodevelopmental disorder that has been attributed to heterozygous deletions in chromosome 7q11.23 and exhibits a variety of physical, cognitive, and behavioral features. However, the genetic basis of this phenotypic variability is unclear. In this study, we identified genetic clues underlying these complex phenotypes. METHODS: Neurobehavioral function was assessed in WS patients and healthy controls. Total RNA was extracted from peripheral blood and subjected to microarray analysis, RNA-sequencing, and qRT-PCR. Weighted gene co-expression network analysis was performed to identify specific alterations related to intermediate disease phenotypes. To functionally interpret each WS-related module, gene ontology and disease-related gene enrichment were examined. We also investigated the micro (mi)RNA expression profiles and miRNA co-expression networks to better explain the regulation of the transcriptome in WS. RESULTS: Our analysis identified four significant co-expression modules related to intermediate WS phenotypes. Notably, the three upregulated WS-related modules were composed exclusively of genes located outside the 7q11.23 region. They were significantly enriched in genes related to B-cell activation, RNA processing, and RNA transport. BCL11A, which is known for its association with speech disorders and intellectual disabilities, was identified as one of the hub genes in the top WS-related module. Finally, these key upregulated mRNA co-expression modules appear to be inversely correlated with a specific downregulated WS-related miRNA co-expression module. CONCLUSIONS: Dysregulation of the mRNA/miRNA network involving genes outside of the 7q11.23 region is likely related to the complex phenotypes observed in WS patients.


Assuntos
Transtorno do Espectro Autista/genética , Perfilação da Expressão Gênica , Expressão Gênica/genética , Síndrome de Williams/genética , Criança , Cromossomos Humanos Par 7/genética , Humanos , MicroRNAs/genética , RNA Mensageiro/genética
8.
Focus (Am Psychiatr Publ) ; 17(1): 66-72, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32015716

RESUMO

(Gandal et al., "Shared molecular neuropathology across major psychiatric disorders parallels polygenic overlap" Science 09 Feb 2018:Vol. 359, Issue 6376, pp. 693-697 (DOI: 10.1126/science.aad6469). Reprinted with permission from AAAS).

9.
Nature ; 560(7719): 441-446, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30111840

RESUMO

Common genetic contributions to autism spectrum disorder (ASD) reside in risk gene variants that individually have minimal effect sizes. As environmental factors that perturb neurodevelopment also underlie idiopathic ASD, it is crucial to identify altered regulators that can orchestrate multiple ASD risk genes during neurodevelopment. Cytoplasmic polyadenylation element binding proteins 1-4 (CPEB1-4) regulate the translation of specific mRNAs by modulating their poly(A)-tails and thereby participate in embryonic development and synaptic plasticity. Here we find that CPEB4 binds transcripts of most high-confidence ASD risk genes. The brains of individuals with idiopathic ASD show imbalances in CPEB4 transcript isoforms that result from decreased inclusion of a neuron-specific microexon. In addition, 9% of the transcriptome shows reduced poly(A)-tail length. Notably, this percentage is much higher for high-confidence ASD risk genes, correlating with reduced expression of the protein products of ASD risk genes. An equivalent imbalance in CPEB4 transcript isoforms in mice mimics the changes in mRNA polyadenylation and protein expression of ASD risk genes and induces ASD-like neuroanatomical, electrophysiological and behavioural phenotypes. Together, these data identify CPEB4 as a regulator of ASD risk genes.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/patologia , Predisposição Genética para Doença/genética , Poliadenilação , Splicing de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Éxons/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Neurônios/metabolismo , Fenótipo , Ligação Proteica , RNA Mensageiro/química , RNA Mensageiro/genética , Transcriptoma
10.
Nature ; 560(7718): E30, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29995847

RESUMO

Change history: In this Letter, the labels for splicing events A3SS and A5SS were swapped in column D of Supplementary Table 3a and b. This has been corrected online.

11.
Proc Natl Acad Sci U S A ; 115(28): 7398-7403, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29946024

RESUMO

A fundamental question in the biology of sex differences has eluded direct study in humans: How does sex-chromosome dosage (SCD) shape genome function? To address this, we developed a systematic map of SCD effects on gene function by analyzing genome-wide expression data in humans with diverse sex-chromosome aneuploidies (XO, XXX, XXY, XYY, and XXYY). For sex chromosomes, we demonstrate a pattern of obligate dosage sensitivity among evolutionarily preserved X-Y homologs and update prevailing theoretical models for SCD compensation by detecting X-linked genes that increase expression with decreasing X- and/or Y-chromosome dosage. We further show that SCD-sensitive sex-chromosome genes regulate specific coexpression networks of SCD-sensitive autosomal genes with critical cellular functions and a demonstrable potential to mediate previously documented SCD effects on disease. These gene coexpression results converge with analysis of transcription factor binding site enrichment and measures of gene expression in murine knockout models to spotlight the dosage-sensitive X-linked transcription factor ZFX as a key mediator of SCD effects on wider genome expression. Our findings characterize the effects of SCD broadly across the genome, with potential implications for human phenotypic variation.


Assuntos
Aneuploidia , Cromossomos Humanos X , Cromossomos Humanos Y , Dosagem de Genes , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like , Modelos Genéticos , Animais , Cromossomos Humanos X/genética , Cromossomos Humanos X/metabolismo , Cromossomos Humanos Y/genética , Cromossomos Humanos Y/metabolismo , Feminino , Estudo de Associação Genômica Ampla , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Camundongos , Camundongos Knockout
12.
J Neurodev Disord ; 10(1): 18, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29859039

RESUMO

BACKGROUND: Genetic studies in autism have pinpointed a heterogeneous group of loci and genes. Further, environment may be an additional factor conferring susceptibility to autism. Transcriptome studies investigate quantitative differences in gene expression between patient-derived tissues and control. These studies may pinpoint genes relevant to pathophysiology yet circumvent the need to understand genetic architecture or gene-by-environment interactions leading to disease. METHODS: We conducted alternate gene set enrichment analyses using differentially expressed genes from a previously published RNA-seq study of post-mortem autism cerebral cortex. We used three previously published microarray datasets for validation and one of the microarray datasets for additional differential expression analysis. The RNA-seq study used 26 autism and 33 control brains in differential gene expression analysis, and the largest microarray dataset contained 15 autism and 16 control post-mortem brains. RESULTS: While performing a gene set enrichment analysis of genes differentially expressed in the RNA-seq study, we discovered that genes associated with mitochondrial function were downregulated in autism cerebral cortex, as compared to control. These genes were correlated with genes related to synaptic function. We validated these findings across the multiple microarray datasets. We also did separate differential expression and gene set enrichment analyses to confirm the importance of the mitochondrial pathway among downregulated genes in post-mortem autism cerebral cortex. CONCLUSIONS: We found that genes related to mitochondrial function were differentially expressed in autism cerebral cortex and correlated with genes related to synaptic transmission. Our principal findings replicate across all datasets investigated. Further, these findings may potentially replicate in other diseases, such as in schizophrenia.


Assuntos
Transtorno do Espectro Autista/genética , Córtex Cerebral/metabolismo , Mitocôndrias/metabolismo , Transmissão Sináptica/genética , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Sinapses/genética , Transcriptoma
13.
Science ; 359(6376): 693-697, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29439242

RESUMO

The predisposition to neuropsychiatric disease involves a complex, polygenic, and pleiotropic genetic architecture. However, little is known about how genetic variants impart brain dysfunction or pathology. We used transcriptomic profiling as a quantitative readout of molecular brain-based phenotypes across five major psychiatric disorders-autism, schizophrenia, bipolar disorder, depression, and alcoholism-compared with matched controls. We identified patterns of shared and distinct gene-expression perturbations across these conditions. The degree of sharing of transcriptional dysregulation is related to polygenic (single-nucleotide polymorphism-based) overlap across disorders, suggesting a substantial causal genetic component. This comprehensive systems-level view of the neurobiological architecture of major neuropsychiatric illness demonstrates pathways of molecular convergence and specificity.


Assuntos
Predisposição Genética para Doença , Transtornos Mentais/genética , Herança Multifatorial , Doenças do Sistema Nervoso/genética , Córtex Cerebral/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Polimorfismo de Nucleotídeo Único , Transcrição Gênica
14.
Nature ; 540(7633): 423-427, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27919067

RESUMO

Autism spectrum disorder (ASD) involves substantial genetic contributions. These contributions are profoundly heterogeneous but may converge on common pathways that are not yet well understood. Here, through post-mortem genome-wide transcriptome analysis of the largest cohort of samples analysed so far, to our knowledge, we interrogate the noncoding transcriptome, alternative splicing, and upstream molecular regulators to broaden our understanding of molecular convergence in ASD. Our analysis reveals ASD-associated dysregulation of primate-specific long noncoding RNAs (lncRNAs), downregulation of the alternative splicing of activity-dependent neuron-specific exons, and attenuation of normal differences in gene expression between the frontal and temporal lobes. Our data suggest that SOX5, a transcription factor involved in neuron fate specification, contributes to this reduction in regional differences. We further demonstrate that a genetically defined subtype of ASD, chromosome 15q11.2-13.1 duplication syndrome (dup15q), shares the core transcriptomic signature observed in idiopathic ASD. Co-expression network analysis reveals that individuals with ASD show age-related changes in the trajectory of microglial and synaptic function over the first two decades, and suggests that genetic risk for ASD may influence changes in regional cortical gene expression. Our findings illustrate how diverse genetic perturbations can lead to phenotypic convergence at multiple biological levels in a complex neuropsychiatric disorder.


Assuntos
Processamento Alternativo/genética , Transtorno do Espectro Autista/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genoma Humano/genética , RNA Longo não Codificante/genética , Animais , Autopsia , Estudos de Casos e Controles , Aberrações Cromossômicas , Cromossomos Humanos Par 15/genética , Éxons/genética , Lobo Frontal/metabolismo , Humanos , Deficiência Intelectual/genética , Neurônios/metabolismo , Primatas/genética , Fatores de Transcrição SOXD/metabolismo , Especificidade da Espécie , Lobo Temporal/metabolismo , Transcriptoma/genética
15.
Cell ; 167(5): 1385-1397.e11, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27863250

RESUMO

The association of histone modification changes with autism spectrum disorder (ASD) has not been systematically examined. We conducted a histone acetylome-wide association study (HAWAS) by performing H3K27ac chromatin immunoprecipitation sequencing (ChIP-seq) on 257 postmortem samples from ASD and matched control brains. Despite etiological heterogeneity, ≥68% of syndromic and idiopathic ASD cases shared a common acetylome signature at >5,000 cis-regulatory elements in prefrontal and temporal cortex. Similarly, multiple genes associated with rare genetic mutations in ASD showed common "epimutations." Acetylome aberrations in ASD were not attributable to genetic differentiation at cis-SNPs and highlighted genes involved in synaptic transmission, ion transport, epilepsy, behavioral abnormality, chemokinesis, histone deacetylation, and immunity. By correlating histone acetylation with genotype, we discovered >2,000 histone acetylation quantitative trait loci (haQTLs) in human brain regions, including four candidate causal variants for psychiatric diseases. Due to the relative stability of histone modifications postmortem, we anticipate that the HAWAS approach will be applicable to multiple diseases.


Assuntos
Transtorno do Espectro Autista/genética , Cerebelo/metabolismo , Código das Histonas , Córtex Pré-Frontal/metabolismo , Locos de Características Quantitativas , Lobo Temporal/metabolismo , Acetilação , Transtorno do Espectro Autista/metabolismo , Autopsia , Imunoprecipitação da Cromatina , Elementos Facilitadores Genéticos , Humanos , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo
16.
Nature ; 538(7626): 523-527, 2016 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-27760116

RESUMO

Three-dimensional physical interactions within chromosomes dynamically regulate gene expression in a tissue-specific manner. However, the 3D organization of chromosomes during human brain development and its role in regulating gene networks dysregulated in neurodevelopmental disorders, such as autism or schizophrenia, are unknown. Here we generate high-resolution 3D maps of chromatin contacts during human corticogenesis, permitting large-scale annotation of previously uncharacterized regulatory relationships relevant to the evolution of human cognition and disease. Our analyses identify hundreds of genes that physically interact with enhancers gained on the human lineage, many of which are under purifying selection and associated with human cognitive function. We integrate chromatin contacts with non-coding variants identified in schizophrenia genome-wide association studies (GWAS), highlighting multiple candidate schizophrenia risk genes and pathways, including transcription factors involved in neurogenesis, and cholinergic signalling molecules, several of which are supported by independent expression quantitative trait loci and gene expression analyses. Genome editing in human neural progenitors suggests that one of these distal schizophrenia GWAS loci regulates FOXG1 expression, supporting its potential role as a schizophrenia risk gene. This work provides a framework for understanding the effect of non-coding regulatory elements on human brain development and the evolution of cognition, and highlights novel mechanisms underlying neuropsychiatric disorders.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Cromatina/química , Cromatina/genética , Cromossomos Humanos/química , Cromossomos Humanos/genética , Regulação da Expressão Gênica no Desenvolvimento , Conformação de Ácido Nucleico , Cromatina/metabolismo , Cromossomos Humanos/metabolismo , Cognição , Elementos Facilitadores Genéticos/genética , Epigênese Genética , Fatores de Transcrição Forkhead/genética , Predisposição Genética para Doença/genética , Estudo de Associação Genômica Ampla , Humanos , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/metabolismo , Neurogênese , Especificidade de Órgãos , Polimorfismo de Nucleotídeo Único/genética , Regiões Promotoras Genéticas/genética , Reprodutibilidade dos Testes , Esquizofrenia/genética , Esquizofrenia/patologia
17.
Nat Neurosci ; 19(11): 1397-1407, 2016 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-27786179

RESUMO

Hundreds of genetic loci increasing risk for neuropsychiatric disorders have recently been identified. This success, perhaps paradoxically, has posed challenges for therapeutic development, which are amplified by the highly polygenic and pleiotropic nature of these genetic contributions. Success requires understanding the biological impact of single genetic variants and predicting their effects within an individual. Comprehensive functional genomic annotation of risk loci provides a framework for interpretation of neurobiological impact, requiring experimental validation with in vivo or in vitro model systems. Systems-level, integrative pathway analyses are beginning to elucidate the additive, polygenic contributions of risk variants on specific cellular, molecular, developmental, or circuit-level processes. Although most neuropsychiatric disease modeling has focused on genes disrupted by rare, large-effect-size mutations, common smaller-effect-size variants may also provide solid therapeutic targets to inform precision medicine approaches. Here we enumerate the promise and challenges of a genomics-driven approach to uncovering neuropsychiatric disease mechanisms and facilitating therapeutic development.


Assuntos
Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Herança Multifatorial/genética , Medicina de Precisão , Processos Psicoterapêuticos , Animais , Humanos , Mutação/genética
18.
Nat Neurosci ; 19(11): 1463-1476, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27571009

RESUMO

Genetic variants conferring risk for autism spectrum disorder (ASD) have been identified, but the role of post-transcriptional mechanisms in ASD is not well understood. We performed genome-wide microRNA (miRNA) expression profiling in post-mortem brains from individuals with ASD and controls and identified miRNAs and co-regulated modules that were perturbed in ASD. Putative targets of these ASD-affected miRNAs were enriched for genes that have been implicated in ASD risk. We confirmed regulatory relationships between several miRNAs and their putative target mRNAs in primary human neural progenitors. These include hsa-miR-21-3p, a miRNA of unknown CNS function that is upregulated in ASD and that targets neuronal genes downregulated in ASD, and hsa_can_1002-m, a previously unknown, primate-specific miRNA that is downregulated in ASD and that regulates the epidermal growth factor receptor and fibroblast growth factor receptor signaling pathways involved in neural development and immune function. Our findings support a role for miRNA dysregulation in ASD pathophysiology and provide a rich data set and framework for future analyses of miRNAs in neuropsychiatric diseases.


Assuntos
Transtorno do Espectro Autista/genética , MicroRNAs/genética , Biologia Computacional/métodos , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica/métodos , Estudo de Associação Genômica Ampla , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA Mensageiro/metabolismo , Regulação para Cima
19.
Nat Commun ; 7: 10717, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26892004

RESUMO

Autism spectrum disorder (ASD) is more prevalent in males, and the mechanisms behind this sex-differential risk are not fully understood. Two competing, but not mutually exclusive, hypotheses are that ASD risk genes are sex-differentially regulated, or alternatively, that they interact with characteristic sexually dimorphic pathways. Here we characterized sexually dimorphic gene expression in multiple data sets from neurotypical adult and prenatal human neocortical tissue, and evaluated ASD risk genes for evidence of sex-biased expression. We find no evidence for systematic sex-differential expression of ASD risk genes. Instead, we observe that genes expressed at higher levels in males are significantly enriched for genes upregulated in post-mortem autistic brain, including astrocyte and microglia markers. This suggests that it is not sex-differential regulation of ASD risk genes, but rather naturally occurring sexually dimorphic processes, potentially including neuron-glial interactions, that modulate the impact of risk variants and contribute to the sex-skewed prevalence of ASD.


Assuntos
Transtorno do Espectro Autista/genética , Encéfalo/metabolismo , Expressão Gênica , Adolescente , Adulto , Transtorno do Espectro Autista/metabolismo , Feminino , Humanos , Masculino , Fatores Sexuais , Transcriptoma , Adulto Jovem
20.
Neuron ; 89(1): 113-28, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26687839

RESUMO

Human genetic studies have identified the neuronal RNA binding protein, Rbfox1, as a candidate gene for autism spectrum disorders. While Rbfox1 functions as a splicing regulator in the nucleus, it is also alternatively spliced to produce cytoplasmic isoforms. To investigate the function of cytoplasmic Rbfox1, we knocked down Rbfox proteins in mouse neurons and rescued with cytoplasmic or nuclear Rbfox1. Transcriptome profiling showed that nuclear Rbfox1 rescued splicing changes, whereas cytoplasmic Rbfox1 rescued changes in mRNA levels. iCLIP-seq of subcellular fractions revealed that Rbfox1 bound predominantly to introns in nascent RNA, while cytoplasmic Rbox1 bound to 3' UTRs. Cytoplasmic Rbfox1 binding increased target mRNA stability and translation, and Rbfox1 and miRNA binding sites overlapped significantly. Cytoplasmic Rbfox1 target mRNAs were enriched in genes involved in cortical development and autism. Our results uncover a new Rbfox1 regulatory network and highlight the importance of cytoplasmic RNA metabolism to cortical development and disease.


Assuntos
Transtorno Autístico/genética , Estriol/análogos & derivados , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Processamento Alternativo/genética , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Citoplasma/metabolismo , Estriol/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos Endogâmicos C57BL , Fatores de Processamento de RNA
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...