Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 181(2): 382-395.e21, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32246942

RESUMO

Multiple sclerosis (MS) is an autoimmune disease characterized by attack on oligodendrocytes within the central nervous system (CNS). Despite widespread use of immunomodulatory therapies, patients may still face progressive disability because of failure of myelin regeneration and loss of neurons, suggesting additional cellular pathologies. Here, we describe a general approach for identifying specific cell types in which a disease allele exerts a pathogenic effect. Applying this approach to MS risk loci, we pinpoint likely pathogenic cell types for 70%. In addition to T cell loci, we unexpectedly identified myeloid- and CNS-specific risk loci, including two sites that dysregulate transcriptional pause release in oligodendrocytes. Functional studies demonstrated inhibition of transcriptional elongation is a dominant pathway blocking oligodendrocyte maturation. Furthermore, pause release factors are frequently dysregulated in MS brain tissue. These data implicate cell-intrinsic aberrations outside of the immune system and suggest new avenues for therapeutic development. VIDEO ABSTRACT.


Assuntos
Comunicação Celular/genética , Doença/genética , Oligodendroglia/metabolismo , Animais , Encéfalo/metabolismo , Sistema Nervoso Central/metabolismo , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Humanos , Esclerose Múltipla/genética , Esclerose Múltipla/metabolismo , Esclerose Múltipla/fisiopatologia , Bainha de Mielina/metabolismo , Neurônios/metabolismo , Oligodendroglia/fisiologia , Fatores de Risco
2.
Proc Natl Acad Sci U S A ; 117(45): 28422-28432, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33109720

RESUMO

The human cerebral cortex contains many cell types that likely underwent independent functional changes during evolution. However, cell-type-specific regulatory landscapes in the cortex remain largely unexplored. Here we report epigenomic and transcriptomic analyses of the two main cortical neuronal subtypes, glutamatergic projection neurons and GABAergic interneurons, in human, chimpanzee, and rhesus macaque. Using genome-wide profiling of the H3K27ac histone modification, we identify neuron-subtype-specific regulatory elements that previously went undetected in bulk brain tissue samples. Human-specific regulatory changes are uncovered in multiple genes, including those associated with language, autism spectrum disorder, and drug addiction. We observe preferential evolutionary divergence in neuron subtype-specific regulatory elements and show that a substantial fraction of pan-neuronal regulatory elements undergoes subtype-specific evolutionary changes. This study sheds light on the interplay between regulatory evolution and cell-type-dependent gene-expression programs, and provides a resource for further exploration of human brain evolution and function.


Assuntos
Córtex Cerebral/metabolismo , Evolução Molecular , Neurônios/metabolismo , Animais , Transtorno do Espectro Autista/genética , Encéfalo/metabolismo , Epigênese Genética , Epigenômica , Expressão Gênica , Código das Histonas , Humanos , Interneurônios/metabolismo , Macaca mulatta/genética , Pan troglodytes/genética , Primatas/genética , Elementos Reguladores de Transcrição , Sequências Reguladoras de Ácido Nucleico , Transcriptoma
3.
Mol Psychiatry ; 24(5): 653-673, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-29955163

RESUMO

Delineating the pathophysiology of psychiatric disorders has been extremely challenging but technological advances in recent decades have facilitated a deeper interrogation of molecular processes in the human brain. Initial candidate gene expression studies of the postmortem brain have evolved into genome wide profiling of the transcriptome and the epigenome, a critical regulator of gene expression. Here, we review the potential and challenges of direct molecular characterization of the postmortem human brain, and provide a brief overview of recent transcriptional and epigenetic studies with respect to neuropsychiatric disorders. Such information can now be leveraged and integrated with the growing number of genome-wide association databases to provide a functional context of trait-associated genetic variants linked to psychiatric illnesses and related phenotypes. While it is clear that the field is still developing and challenges remain to be surmounted, these recent advances nevertheless hold tremendous promise for delineating the neurobiological underpinnings of mental diseases and accelerating the development of novel medication strategies.


Assuntos
Encéfalo/fisiopatologia , Transtornos Mentais/genética , Transtornos Mentais/fisiopatologia , Autopsia/métodos , Epigênese Genética/genética , Regulação da Expressão Gênica/genética , Predisposição Genética para Doença/genética , Genoma/genética , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Fenótipo , Transcriptoma/genética
4.
Nucleic Acids Res ; 44(6): 2593-612, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26612861

RESUMO

The brain is built from a large number of cell types which have been historically classified using location, morphology and molecular markers. Recent research suggests an important role of epigenetics in shaping and maintaining cell identity in the brain. To elucidate the role of DNA methylation in neuronal differentiation, we developed a new protocol for separation of nuclei from the two major populations of human prefrontal cortex neurons--GABAergic interneurons and glutamatergic (GLU) projection neurons. Major differences between the neuronal subtypes were revealed in CpG, non-CpG and hydroxymethylation (hCpG). A dramatically greater number of undermethylated CpG sites in GLU versus GABA neurons were identified. These differences did not directly translate into differences in gene expression and did not stem from the differences in hCpG methylation, as more hCpG methylation was detected in GLU versus GABA neurons. Notably, a comparable number of undermethylated non-CpG sites were identified in GLU and GABA neurons, and non-CpG methylation was a better predictor of subtype-specific gene expression compared to CpG methylation. Regions that are differentially methylated in GABA and GLU neurons were significantly enriched for schizophrenia risk loci. Collectively, our findings suggest that functional differences between neuronal subtypes are linked to their epigenetic specification.


Assuntos
Metilação de DNA , Epigênese Genética , Neurônios GABAérgicos/metabolismo , Loci Gênicos , Neurônios/metabolismo , Córtex Pré-Frontal/metabolismo , Adulto , Autopsia , Mapeamento Encefálico , Ilhas de CpG , Neurônios GABAérgicos/citologia , Ácido Glutâmico/metabolismo , Humanos , Masculino , Microtomia , Pessoa de Meia-Idade , Neurônios/citologia , Especificidade de Órgãos , Córtex Pré-Frontal/anatomia & histologia , Fatores de Risco , Esquizofrenia/genética , Esquizofrenia/metabolismo , Esquizofrenia/patologia
5.
Hum Mol Genet ; 23(18): 4801-13, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24781207

RESUMO

Editing of the pre-mRNA for the serotonin receptor 2C (5-HT2CR) by site-specific adenosine deamination (A-to-I pre-mRNA editing) substantially increases the functional plasticity of this key neurotransmitter receptor and is thought to contribute to homeostatic mechanisms in neurons. 5-HT2CR mRNA editing generates up to 24 different receptor isoforms. The extent of editing correlates with 5-HT2CR functional activity: more highly edited isoforms exhibit the least function. Altered 5-HT2CR editing has been reported in postmortem brains of suicide victims. We report a comparative analysis of the connections among 5-HT2CR editing, genome-wide gene expression and DNA methylation in suicide victims, individuals with major depressive disorder and non-psychiatric controls. The results confirm previous findings of an overrepresentation of highly edited mRNA variants (which encode hypoactive 5-HT2CR receptors) in the brains of suicide victims. A large set of genes for which the expression level is associated with editing was detected. This signature set of editing-associated genes is significantly enriched for genes that are involved in synaptic transmission, genes that are preferentially expressed in neurons, and genes whose expression is correlated with the level of DNA methylation. Notably, we report that the link between 5-HT2CR editing and gene expression is disrupted in suicide victims. The results suggest that the postulated homeostatic function of 5-HT2CR editing is dysregulated in individuals who committed suicide.


Assuntos
Transtorno Depressivo Maior/genética , Redes Reguladoras de Genes , Córtex Pré-Frontal/metabolismo , Receptor 5-HT2C de Serotonina/genética , Suicídio , Autopsia , Estudos de Casos e Controles , Metilação de DNA , Perfilação da Expressão Gênica , Humanos , Neurônios/metabolismo , Edição de RNA , Receptor 5-HT2C de Serotonina/metabolismo
6.
Nucleic Acids Res ; 42(1): 109-27, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24057217

RESUMO

We applied Illumina Human Methylation450K array to perform a genomic-scale single-site resolution DNA methylation analysis in neuronal and nonneuronal (primarily glial) nuclei separated from the orbitofrontal cortex of postmortem human brain. The findings were validated using enhanced reduced representation bisulfite sequencing. We identified thousands of sites differentially methylated (DM) between neuronal and nonneuronal cells. The DM sites were depleted within CpG-island-containing promoters but enriched in predicted enhancers. Classification of the DM sites into those undermethylated in neurons (neuronal type) and those undermethylated in nonneuronal cells (glial type), combined with findings of others that methylation within control elements typically negatively correlates with gene expression, yielded large sets of predicted neuron-specific and non-neuron-specific genes. These sets of predicted genes were in excellent agreement with the available direct measurements of gene expression in human and mouse. We also found a distinct set of DNA methylation patterns that were unique for neuronal cells. In particular, neuronal-type differential methylation was overrepresented in CpG island shores, enriched within gene bodies but not in intergenic regions, and preferentially harbored binding motifs for a distinct set of transcription factors, including neuron-specific activity-dependent factors. Finally, non-CpG methylation was substantially more prevalent in neurons than in nonneuronal cells.


Assuntos
Encéfalo/metabolismo , Metilação de DNA , Elementos Facilitadores Genéticos , Neuroglia/metabolismo , Neurônios/metabolismo , Adulto , Animais , Sítios de Ligação , Núcleo Celular/genética , Ilhas de CpG , Evolução Molecular , Expressão Gênica , Genoma Humano , Humanos , Masculino , Camundongos , Motivos de Nucleotídeos , Fatores de Transcrição/metabolismo , Sítio de Iniciação de Transcrição , Adulto Jovem
7.
PLoS Comput Biol ; 8(9): e1002663, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22969417

RESUMO

The serotonin 2C receptor (5-HT(2C)R)-a key regulator of diverse neurological processes-exhibits functional variability derived from editing of its pre-mRNA by site-specific adenosine deamination (A-to-I pre-mRNA editing) in five distinct sites. Here we describe a statistical technique that was developed for analysis of the dependencies among the editing states of the five sites. The statistical significance of the observed correlations was estimated by comparing editing patterns in multiple individuals. For both human and rat 5-HT(2C)R, the editing states of the physically proximal sites A and B were found to be strongly dependent. In contrast, the editing states of sites C and D, which are also physically close, seem not to be directly dependent but instead are linked through the dependencies on sites A and B, respectively. We observed pronounced differences between the editing patterns in humans and rats: in humans site A is the key determinant of the editing state of the other sites, whereas in rats this role belongs to site B. The structure of the dependencies among the editing sites is notably simpler in rats than it is in humans implying more complex regulation of 5-HT(2C)R editing and, by inference, function in the human brain. Thus, exhaustive statistical analysis of the 5-HT(2C)R editing patterns indicates that the editing state of sites A and B is the primary determinant of the editing states of the other three sites, and hence the overall editing pattern. Taken together, these findings allow us to propose a mechanistic model of concerted action of ADAR1 and ADAR2 in 5-HT(2C)R editing. Statistical approach developed here can be applied to other cases of interdependencies among modification sites in RNA and proteins.


Assuntos
Edição de RNA/genética , RNA Mensageiro/genética , Receptor 5-HT2C de Serotonina/genética , Análise de Sequência de Proteína/métodos , Sequência de Bases , Dados de Sequência Molecular
8.
Nat Commun ; 14(1): 5714, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37714849

RESUMO

A repeat expansion in the C9orf72 (C9) gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we investigate single nucleus transcriptomics (snRNA-seq) and epigenomics (snATAC-seq) in postmortem motor and frontal cortices from C9-ALS, C9-FTD, and control donors. C9-ALS donors present pervasive alterations of gene expression with concordant changes in chromatin accessibility and histone modifications. The greatest alterations occur in upper and deep layer excitatory neurons, as well as in astrocytes. In neurons, the changes imply an increase in proteostasis, metabolism, and protein expression pathways, alongside a decrease in neuronal function. In astrocytes, the alterations suggest activation and structural remodeling. Conversely, C9-FTD donors have fewer high-quality neuronal nuclei in the frontal cortex and numerous gene expression changes in glial cells. These findings highlight a context-dependent molecular disruption in C9-ALS and C9-FTD, indicating unique effects across cell types, brain regions, and diseases.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Humanos , Demência Frontotemporal/genética , Esclerose Lateral Amiotrófica/genética , Proteína C9orf72/genética , Transcriptoma/genética , Epigenoma , Mutação
9.
J Psychiatry Neurosci ; 37(4): 267-72, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22469055

RESUMO

BACKGROUND: Dysfunction of glutamate neurotransmission has been implicated in the pathology of schizophrenia and bipolar disorder, and one mechanism by which glutamate signalling can be altered is through RNA editing of ionotropic glutamate receptors (iGluRs). The objectives of the present study were to evaluate the editing status of iGluRs in the human prefrontal cortex, determine whether iGluR editing is associated with psychiatric disease or suicide and evaluate a potential association between editing and alternative splicing in the α-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) iGluR subunits' pre-mRNA. METHODS: We studied specimens derived from patients with antemortem diagnoses of bipolar disorder (n = 31) or schizophrenia (n = 34) who died by suicide or other causes, and from psychiatrically healthy controls (n = 34) who died from causes other than suicide. The RNA editing at all 8 editing sites within AMPA (GluA2-4 subunits) and kainate (GluK1-2 subunits) iGluRs was analyzed using a novel real-time quantitative polymerase chain reaction assay. RESULTS: No differences in editing were detected among schizophrenia, bipolar or control groups or between suicide completers and patients who died from causes other than suicide. The editing efficiency was significantly higher in the flop than in the flip splicoforms of GluA3-4 AMPA subunits (all p < 0.001). LIMITATIONS: The study is limited by the near absence of specimens from medicationnaive psychiatric patients and considerable variation in medication regimens among individuals, both of which introduce considerable uncertainty into the analysis of potential medication effects. CONCLUSION: We found that iGluR RNA editing status was not associated with bipolar disorder, schizophrenia or suicide. Differences in editing between flip and flop splicoforms suggest that glutamate sensitivity of receptors containing GluA3 and/or GluA4 flop subunits is moderated as a result of increased editing.


Assuntos
Transtorno Bipolar/genética , Córtex Pré-Frontal/metabolismo , Edição de RNA/genética , Receptores Ionotrópicos de Glutamato/genética , Esquizofrenia/genética , Adulto , Transtorno Bipolar/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Receptores Ionotrópicos de Glutamato/metabolismo , Esquizofrenia/metabolismo , Suicídio
10.
Nat Commun ; 13(1): 2997, 2022 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-35637184

RESUMO

Posttranscriptional adenosine-to-inosine modifications amplify the functionality of RNA molecules in the brain, yet the cellular and genetic regulation of RNA editing is poorly described. We quantify base-specific RNA editing across three major cell populations from the human prefrontal cortex: glutamatergic neurons, medial ganglionic eminence-derived GABAergic neurons, and oligodendrocytes. We identify more selective editing and hyper-editing in neurons relative to oligodendrocytes. RNA editing patterns are highly cell type-specific, with 189,229 cell type-associated sites. The cellular specificity for thousands of sites is confirmed by single nucleus RNA-sequencing. Importantly, cell type-associated sites are enriched in GTEx RNA-sequencing data, edited ~twentyfold higher than all other sites, and variation in RNA editing is largely explained by neuronal proportions in bulk brain tissue. Finally, we uncover 661,791 cis-editing quantitative trait loci across thirteen brain regions, including hundreds with cell type-associated features. These data reveal an expansive repertoire of highly regulated RNA editing sites across human brain cell types and provide a resolved atlas linking cell types to editing variation and genetic regulatory effects.


Assuntos
Inosina , Edição de RNA , Encéfalo/metabolismo , Humanos , Inosina/genética , Inosina/metabolismo , Locos de Características Quantitativas/genética , RNA/metabolismo , Edição de RNA/genética
11.
Biol Psychiatry ; 91(1): 92-101, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34154796

RESUMO

BACKGROUND: While schizophrenia differs between males and females in the age of onset, symptomatology, and disease course, the molecular mechanisms underlying these differences remain uncharacterized. METHODS: To address questions about the sex-specific effects of schizophrenia, we performed a large-scale transcriptome analysis of RNA sequencing data from 437 controls and 341 cases from two distinct cohorts from the CommonMind Consortium. RESULTS: Analysis across the cohorts identified a reproducible gene expression signature of schizophrenia that was highly concordant with previous work. Differential expression across sex was reproducible across cohorts and identified X- and Y-linked genes, as well as those involved in dosage compensation. Intriguingly, the sex expression signature was also enriched for genes involved in neurexin family protein binding and synaptic organization. Differential expression analysis testing a sex-by-diagnosis interaction effect did not identify any genome-wide signature after multiple testing corrections. Gene coexpression network analysis was performed to reduce dimensionality from thousands of genes to dozens of modules and elucidate interactions among genes. We found enrichment of coexpression modules for sex-by-diagnosis differential expression signatures, which were highly reproducible across the two cohorts and involved a number of diverse pathways, including neural nucleus development, neuron projection morphogenesis, and regulation of neural precursor cell proliferation. CONCLUSIONS: Overall, our results indicate that the effect size of sex differences in schizophrenia gene expression signatures is small and underscore the challenge of identifying robust sex-by-diagnosis signatures, which will require future analyses in larger cohorts.


Assuntos
Esquizofrenia , Transcriptoma , Encéfalo , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Esquizofrenia/genética , Caracteres Sexuais
12.
Nat Neurosci ; 25(4): 474-483, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35332326

RESUMO

Chromosomal organization, scaling from the 147-base pair (bp) nucleosome to megabase-ranging domains encompassing multiple transcriptional units, including heritability loci for psychiatric traits, remains largely unexplored in the human brain. In this study, we constructed promoter- and enhancer-enriched nucleosomal histone modification landscapes for adult prefrontal cortex from H3-lysine 27 acetylation and H3-lysine 4 trimethylation profiles, generated from 388 controls and 351 individuals diagnosed with schizophrenia (SCZ) or bipolar disorder (BD) (n = 739). We mapped thousands of cis-regulatory domains (CRDs), revealing fine-grained, 104-106-bp chromosomal organization, firmly integrated into Hi-C topologically associating domain stratification by open/repressive chromosomal environments and nuclear topography. Large clusters of hyper-acetylated CRDs were enriched for SCZ heritability, with prominent representation of regulatory sequences governing fetal development and glutamatergic neuron signaling. Therefore, SCZ and BD brains show coordinated dysregulation of risk-associated regulatory sequences assembled into kilobase- to megabase-scaling chromosomal domains.


Assuntos
Transtorno Bipolar , Esquizofrenia , Adulto , Transtorno Bipolar/genética , Encéfalo , Cromatina , Humanos , Lisina/genética , Esquizofrenia/genética
13.
Nat Commun ; 12(1): 3968, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34172755

RESUMO

Cellular heterogeneity in the human brain obscures the identification of robust cellular regulatory networks, which is necessary to understand the function of non-coding elements and the impact of non-coding genetic variation. Here we integrate genome-wide chromosome conformation data from purified neurons and glia with transcriptomic and enhancer profiles, to characterize the gene regulatory landscape of two major cell classes in the human brain. We then leverage cell-type-specific regulatory landscapes to gain insight into the cellular etiology of several brain disorders. We find that Alzheimer's disease (AD)-associated epigenetic dysregulation is linked to neurons and oligodendrocytes, whereas genetic risk factors for AD highlighted microglia, suggesting that different cell types may contribute to disease risk, via different mechanisms. Moreover, integration of glutamatergic and GABAergic regulatory maps with genetic risk factors for schizophrenia (SCZ) and bipolar disorder (BD) identifies shared (parvalbumin-expressing interneurons) and distinct cellular etiologies (upper layer neurons for BD, and deeper layer projection neurons for SCZ). Collectively, these findings shed new light on cell-type-specific gene regulatory networks in brain disorders.


Assuntos
Doença de Alzheimer/genética , Transtorno Bipolar/genética , Cromatina/ultraestrutura , Esquizofrenia/genética , Acetilação , Doença de Alzheimer/patologia , Transtorno Bipolar/patologia , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Sequenciamento de Cromatina por Imunoprecipitação , Elementos Facilitadores Genéticos , Epigênese Genética , Neurônios GABAérgicos/metabolismo , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Histonas/metabolismo , Humanos , Lisina/metabolismo , Neuroglia/patologia , Neuroglia/ultraestrutura , Neurônios/patologia , Neurônios/ultraestrutura , Regiões Promotoras Genéticas , Esquizofrenia/patologia
14.
Epigenomics ; 13(15): 1157-1169, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34323598

RESUMO

Aim: Identify grey- and white-matter-specific DNA-methylation differences between schizophrenia (SCZ) patients and controls in postmortem brain cortical tissue. Materials & methods: Grey and white matter were separated from postmortem brain tissue of the superior temporal and medial frontal gyrus from SCZ (n = 10) and control (n = 11) cases. Genome-wide DNA-methylation analysis was performed using the Infinium EPIC Methylation Array (Illumina, CA, USA). Results: Four differentially methylated regions associated with SCZ status and tissue type (grey vs white matter) were identified within or near KLF9, SFXN1, SPRED2 and ALS2CL genes. Gene-expression analysis showed differential expression of KLF9 and SFXN1 in SCZ. Conclusion: Our data show distinct differences in DNA methylation between grey and white matter that are unique to SCZ, providing new leads to unravel the pathogenesis of SCZ.


Lay abstract This study investigated the way gene activity is regulated in brain cells of patients with schizophrenia (SCZ; a severe mental illness characterized by psychosis) compared with unaffected controls. The study focuses on the differences between parts of the brain with many cell bodies (grey matter) in contrast to those parts with mainly conducting fibers (white matter). For that purpose, grey and white matter were separated from brain tissue of ten individuals with SCZ and 11 without. All brains were obtained after the patients died and donated their brains to science. Array technology was used to analyze 800,000 sections of the DNA at once. The study identified regions on four genes that can turn the genes on and off differently in schizophrenic patients compared with controls, these genes were also turned on or off depending on their location either in grey or white matter. Two of these genes showed different activation in schizophrenic patients compared with controls. Overall this study identified distinct differences between grey and white matter that are unique to SCZ, providing new leads to unravel the biology of SCZ.


Assuntos
Metilação de DNA , Regulação da Expressão Gênica , Substância Cinzenta/metabolismo , Esquizofrenia/etiologia , Substância Branca/metabolismo , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Epigênese Genética , Epigenômica/métodos , Feminino , Substância Cinzenta/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Substância Branca/fisiopatologia
15.
Nat Commun ; 11(1): 5581, 2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-33149216

RESUMO

The chromatin landscape of human brain cells encompasses key information to understanding brain function. Here we use ATAC-seq to profile the chromatin structure in four distinct populations of cells (glutamatergic neurons, GABAergic neurons, oligodendrocytes, and microglia/astrocytes) from three different brain regions (anterior cingulate cortex, dorsolateral prefrontal cortex, and primary visual cortex) in human postmortem brain samples. We find that chromatin accessibility varies greatly by cell type and, more moderately, by brain region, with glutamatergic neurons showing the largest regional variability. Transcription factor footprinting implicates cell-specific transcriptional regulators and infers cell-specific regulation of protein-coding genes, long intergenic noncoding RNAs and microRNAs. In vivo transgenic mouse experiments validate the cell type specificity of several of these human-derived regulatory sequences. We find that open chromatin regions in glutamatergic neurons are enriched for neuropsychiatric risk variants, particularly those associated with schizophrenia. Integration of cell-specific chromatin data with a bulk tissue study of schizophrenia brains increases statistical power and confirms that glutamatergic neurons are most affected. These findings illustrate the utility of studying the cell-type-specific epigenome in complex tissues like the human brain, and the potential of such approaches to better understand the genetic basis of human brain function.


Assuntos
Astrócitos/metabolismo , Cromatina/metabolismo , Neurônios GABAérgicos/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Oligodendroglia/metabolismo , Esquizofrenia/metabolismo , Animais , Cromatina/genética , Epigênese Genética , Regulação da Expressão Gênica/genética , Giro do Cíngulo/citologia , Giro do Cíngulo/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , MicroRNAs/metabolismo , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/metabolismo , Regiões Promotoras Genéticas , RNA Longo não Codificante/metabolismo , Fatores de Risco , Esquizofrenia/genética , Fatores de Transcrição/metabolismo , Córtex Visual/citologia , Córtex Visual/metabolismo
16.
Anal Biochem ; 390(2): 173-80, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19371717

RESUMO

Abnormal adenosine to inosine (A-to-I) messenger RNA (mRNA) editing has been linked to several disease states afflicting the central nervous system. Here we report an assay to determine RNA editing frequencies at specific sites that is based on quantitative polymerase chain reaction (qPCR) with TaqMan probes. The assay was tested by measuring the frequency of the A-to-I mRNA editing at the Q/R site of the human kainate receptor subunit GluR5 and was compared with two established methods of assessing RNA editing: sequencing of individual clones and restriction analysis. The qPCR assay displayed high sensitivity and reproducibility, demonstrated exceptional discrimination between edited and unedited transcript variants, and proved to have several advantages over the other editing methods. Due to the fact that TaqMan-based qPCR technology can be easily adapted to different editing targets, the increased capabilities afforded by this new technique should facilitate various RNA editing studies that aim to elucidate the role of this process in normal physiology and in disease.


Assuntos
Reação em Cadeia da Polimerase/métodos , Edição de RNA , RNA Mensageiro/análise , Receptores de Ácido Caínico/genética , Adenosina/análise , Sequência de Bases , Humanos , Inosina/análise , Masculino , Dados de Sequência Molecular , Plasmídeos , Córtex Pré-Frontal/química , RNA Mensageiro/isolamento & purificação , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
17.
Schizophr Res ; 98(1-3): 118-28, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18029146

RESUMO

Considerable evidence based on the study of postmortem brain tissue suggests deficits in both neuronal and myelin systems in schizophrenia (SZ). To date, the majority of the biochemical and molecular biological studies have focused on the cerebral cortex. Most information traveling to or from the cortex is relayed or synaptically gated through the thalamus, and numerous studies suggest structural and functional abnormalities in interconnected regions of the thalamus and cortex in SZ. The present study extends our gene expression studies of neuronal and myelin systems to the thalamus. Quantitative PCR was employed to assess the expression of 10 genes in 5 divisions of the thalamus which were precisely harvested using Laser Capture Microdissection. The divisions studied were present on coronal sections at the level of the centromedian nucleus (CMN) taken from 14 schizophrenic and 16 normal control postmortem brains. The genes examined were specific for oligodendrocytes (MAG, CNP, MBP), neurons (ENO2), glutamatergic neurons (VGlut1, VGlut2, PV, CB) or GABAergic neurons (GAD65, GAD67). Expression levels for each of these markers were quantitated and compared between diagnoses, between sexes, and across nuclei. CB was much more highly expressed in the CMN in SZs compared to NCs. No other diagnosis related differences in gene expression were observed. The expression levels of CNP and MAG, but not MBP, were highly correlated with one another and both, but not MBP, were much more highly expressed in females than in males in all thalamic divisions examined. All markers were differentially expressed across nuclei.


Assuntos
Expressão Gênica/genética , Neurônios/metabolismo , Oligodendroglia/metabolismo , Esquizofrenia/genética , Esquizofrenia/metabolismo , Núcleos Talâmicos/metabolismo , Idoso , Calbindinas , Manual Diagnóstico e Estatístico de Transtornos Mentais , Feminino , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/metabolismo , Humanos , Núcleos Intralaminares do Tálamo/metabolismo , Masculino , Proteína Básica da Mielina/genética , Proteína Básica da Mielina/metabolismo , Proteínas da Mielina/genética , Proteínas da Mielina/metabolismo , Glicoproteína Associada a Mielina/genética , Glicoproteína Associada a Mielina/metabolismo , Reação em Cadeia da Polimerase/métodos , Escalas de Graduação Psiquiátrica , Proteína G de Ligação ao Cálcio S100/genética , Proteína G de Ligação ao Cálcio S100/metabolismo , Esquizofrenia/diagnóstico , Fatores Sexuais , Proteína Vesicular 2 de Transporte de Glutamato/genética , Proteína Vesicular 2 de Transporte de Glutamato/metabolismo
18.
Neuroreport ; 19(3): 379-82, 2008 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-18303585

RESUMO

In an earlier study, we showed increases in serotonin 2C receptor (5-HT2CR) pre-mRNA editing in prefrontal cortex that were specific to suicide victims irrespective of associated psychiatric diagnoses. Here we demonstrate that the ratio between the two 5-HT2CR splice variants is increased in people who committed suicide, but does not vary among the diagnostic groups. This provides further evidence for suicide-specific neurobiology and suggests that, as it was previously shown in vitro, 5-HT2CR editing modulates its splicing in human brain. The association analysis indicates, however, that the efficiency of 5-HT2CR editing is an imperfect predictor of the splicing outcome, and that splice site selection is only partially controlled by the level of editing in vivo.


Assuntos
Córtex Pré-Frontal/metabolismo , Splicing de RNA/genética , Receptor 5-HT2C de Serotonina/biossíntese , Suicídio , Alcoolismo , DNA Complementar/biossíntese , DNA Complementar/genética , Desoxirribonucleases/química , Humanos , Receptor 5-HT2C de Serotonina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica
19.
Brain Res ; 1214: 23-34, 2008 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-18462708

RESUMO

Abnormalities in glutamate neurotransmission are thought to be among the major contributing factors to the pathophysiology of schizophrenia. Although schizophrenia has been regarded mostly as a disorder of higher cortical function, the cortex and thalamus work as a functional unit. Existing data regarding alterations of glutamate receptor subunit expression in the thalamus in schizophrenia remain equivocal. This postmortem study examined mRNA expression of ionotropic glutamate receptor (iGluR) subunits and PSD95 in 5 precisely defined and dissected thalamic subdivisions (medial and lateral sectors of the mediodorsal nucleus; and the ventral lateral posterior, ventral posterior, and centromedian nuclei) of persons with schizophrenia and matched controls using quantitative PCR with normalization to multiple endogenous controls. Among 15 genes examined (NR1 and NR2A-D subunits of the NMDA receptor; GluR1-4 subunits of the AMPA receptor; GluR5-7 and KA1-2 subunits of the kainate receptor; PSD95), all but two (GluR4 and KA1) were expressed at quantifiable levels. Differences in iGluR gene expression were seen between different thalamic nuclei but not between diagnostic groups. The relative abundance of transcripts was: NR1>>NR2A>NR2B>NR2D>NR2C for NMDA, GluR2>GluR1>GluR3 for AMPA, and KA2>GluR5>GluR7>GluR6 for kainate receptors. The expression of PSD95 correlated with the expression of NR1, NR2A, NR2B, NR2D and GluR6 in all nuclei. These results provide detailed and quantitative information on iGluR subunit expression in multiple nuclei of the human thalamus but suggest that alterations in their expression are not a prominent feature of schizophrenia.


Assuntos
Expressão Gênica/fisiologia , RNA Mensageiro/metabolismo , Receptores de Glutamato/genética , Esquizofrenia/patologia , Tálamo/metabolismo , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Feminino , Humanos , Masculino , Mudanças Depois da Morte , Receptores de AMPA/genética , Receptores de AMPA/metabolismo , Receptores de Glutamato/classificação , Receptores de Glutamato/metabolismo , Receptores de Ácido Caínico/genética , Receptores de Ácido Caínico/metabolismo
20.
Sci Adv ; 4(9): eaau6190, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30263963

RESUMO

Brain function depends on interaction of diverse cell types whose gene expression and identity are defined, in part, by epigenetic mechanisms. Neuronal DNA contains two major epigenetic modifications, methylcytosine (mC) and hydroxymethylcytosine (hmC), yet their cell type-specific landscapes and relationship with gene expression are poorly understood. We report high-resolution (h)mC analyses, together with transcriptome and histone modification profiling, in three major cell types in human prefrontal cortex: glutamatergic excitatory neurons, medial ganglionic eminence-derived γ-aminobutyric acid (GABA)ergic inhibitory neurons, and oligodendrocytes. We detected a unique association between hmC and gene expression in inhibitory neurons that differed significantly from the pattern in excitatory neurons and oligodendrocytes. We also found that risk loci associated with neuropsychiatric diseases were enriched near regions of reduced hmC in excitatory neurons and reduced mC in inhibitory neurons. Our findings indicate differential roles for mC and hmC in regulation of gene expression in different brain cell types, with implications for the etiology of human brain diseases.


Assuntos
Encefalopatias/genética , Encéfalo/metabolismo , Metilação de DNA , Epigênese Genética , Inibição Neural/fisiologia , Neurônios/metabolismo , Adulto , Encéfalo/patologia , Encefalopatias/fisiopatologia , Cadáver , Núcleo Celular/genética , Humanos , Masculino , Neurônios/patologia , Polimorfismo de Nucleotídeo Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA