Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
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
2.
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
3.
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
4.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Sci Rep ; 8(1): 8868, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29892006

RESUMO

Elucidating brain cell type specific gene expression patterns is critical towards a better understanding of how cell-cell communications may influence brain functions and dysfunctions. We set out to compare and contrast five human and murine cell type-specific transcriptome-wide RNA expression data sets that were generated within the past several years. We defined three measures of brain cell type-relative expression including specificity, enrichment, and absolute expression and identified corresponding consensus brain cell "signatures," which were well conserved across data sets. We validated that the relative expression of top cell type markers are associated with proxies for cell type proportions in bulk RNA expression data from postmortem human brain samples. We further validated novel marker genes using an orthogonal ATAC-seq dataset. We performed multiscale coexpression network analysis of the single cell data sets and identified robust cell-specific gene modules. To facilitate the use of the cell type-specific genes for cell type proportion estimation and deconvolution from bulk brain gene expression data, we developed an R package, BRETIGEA. In summary, we identified a set of novel brain cell consensus signatures and robust networks from the integration of multiple datasets and therefore transcend limitations related to technical issues characteristic of each individual study.


Assuntos
Bases de Dados de Ácidos Nucleicos , Células Endoteliais/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Lobo Temporal , Transcriptoma , Animais , Conjuntos de Dados como Assunto , Células Endoteliais/citologia , Feminino , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Marcadores Genéticos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/citologia , Neurônios/citologia , Análise de Célula Única , Lobo Temporal/citologia , Lobo Temporal/metabolismo
14.
Clin Cancer Res ; 24(6): 1355-1363, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29351917

RESUMO

Purpose: The classification of medulloblastoma into WNT, SHH, group 3, and group 4 subgroups has become of critical importance for patient risk stratification and subgroup-tailored clinical trials. Here, we aimed to develop a simplified, clinically applicable classification approach that can be implemented in the majority of centers treating patients with medulloblastoma.Experimental Design: We analyzed 1,577 samples comprising previously published DNA methylation microarray data (913 medulloblastomas, 457 non-medulloblastoma tumors, 85 normal tissues), and 122 frozen and formalin-fixed paraffin-embedded medulloblastoma samples. Biomarkers were identified applying stringent selection filters and Linear Discriminant Analysis (LDA) method, and validated using DNA methylation microarray data, bisulfite pyrosequencing, and direct-bisulfite sequencing.Results: Using a LDA-based approach, we developed and validated a prediction method (EpiWNT-SHH classifier) based on six epigenetic biomarkers that allowed for rapid classification of medulloblastoma into the clinically relevant subgroups WNT, SHH, and non-WNT/non-SHH with excellent concordance (>99%) with current gold-standard methods, DNA methylation microarray, and gene signature profiling analysis. The EpiWNT-SHH classifier showed high prediction capacity using both frozen and formalin-fixed material, as well as diverse DNA methylation detection methods. Similarly, we developed a classifier specific for group 3 and group 4 tumors, based on five biomarkers (EpiG3-G4) with good discriminatory capacity, allowing for correct assignment of more than 92% of tumors. EpiWNT-SHH and EpiG3-G4 methylation profiles remained stable across tumor primary, metastasis, and relapse samples.Conclusions: The EpiWNT-SHH and EpiG3-G4 classifiers represent a new simplified approach for accurate, rapid, and cost-effective molecular classification of single medulloblastoma DNA samples, using clinically applicable DNA methylation detection methods. Clin Cancer Res; 24(6); 1355-63. ©2018 AACR.


Assuntos
Biomarcadores Tumorais , Neoplasias Cerebelares/diagnóstico , Neoplasias Cerebelares/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Meduloblastoma/diagnóstico , Meduloblastoma/genética , Biópsia , Ilhas de CpG , Metilação de DNA , Epigênese Genética , Epigenômica/métodos , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Reprodutibilidade dos Testes
15.
Nat Neurosci ; 20(9): 1247-1259, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28783139

RESUMO

Antipsychotic drugs remain the standard for schizophrenia treatment. Despite their effectiveness in treating hallucinations and delusions, prolonged exposure to antipsychotic medications leads to cognitive deficits in both schizophrenia patients and animal models. The molecular mechanisms underlying these negative effects on cognition remain to be elucidated. Here we demonstrate that chronic antipsychotic drug exposure increases nuclear translocation of NF-κB in both mouse and human frontal cortex, a trafficking event triggered via 5-HT2A-receptor-dependent downregulation of the NF-κB repressor IκBα. This upregulation of NF-κB activity led to its increased binding at the Hdac2 promoter, thereby augmenting Hdac2 transcription. Deletion of HDAC2 in forebrain pyramidal neurons prevented the negative effects of antipsychotic treatment on synaptic remodeling and cognition. Conversely, virally mediated activation of NF-κB signaling decreased cortical synaptic plasticity via HDAC2. Together, these observations may aid in developing therapeutic strategies to improve the outcome of schizophrenia treatment.


Assuntos
Antipsicóticos/efeitos adversos , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/metabolismo , Histona Desacetilase 2/metabolismo , NF-kappa B/metabolismo , Sinapses/metabolismo , Animais , Antipsicóticos/toxicidade , Transtornos Cognitivos/genética , Lobo Frontal/efeitos dos fármacos , Lobo Frontal/metabolismo , Células HEK293 , Histona Desacetilase 2/deficiência , Histona Desacetilase 2/genética , Humanos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , NF-kappa B/genética , Sinapses/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/fisiologia
16.
Nat Med ; 23(6): 733-741, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28459438

RESUMO

Blood vessels in the central nervous system (CNS) are controlled by neuronal activity. For example, widespread vessel constriction (vessel tone) is induced by brainstem neurons that release the monoamines serotonin and noradrenaline, and local vessel dilation is induced by glutamatergic neuron activity. Here we examined how vessel tone adapts to the loss of neuron-derived monoamines after spinal cord injury (SCI) in rats. We find that, months after the imposition of SCI, the spinal cord below the site of injury is in a chronic state of hypoxia owing to paradoxical excess activity of monoamine receptors (5-HT1) on pericytes, despite the absence of monoamines. This monoamine-receptor activity causes pericytes to locally constrict capillaries, which reduces blood flow to ischemic levels. Receptor activation in the absence of monoamines results from the production of trace amines (such as tryptamine) by pericytes that ectopically express the enzyme aromatic L-amino acid decarboxylase (AADC), which synthesizes trace amines directly from dietary amino acids (such as tryptophan). Inhibition of monoamine receptors or of AADC, or even an increase in inhaled oxygen, produces substantial relief from hypoxia and improves motoneuron and locomotor function after SCI.


Assuntos
Monoaminas Biogênicas/metabolismo , Capilares/metabolismo , Hipóxia/metabolismo , Locomoção/fisiologia , Pericitos/metabolismo , Traumatismos da Medula Espinal/metabolismo , Vasoconstrição , Animais , Descarboxilases de Aminoácido-L-Aromático/metabolismo , Capilares/efeitos dos fármacos , Capilares/patologia , Capilares/fisiopatologia , Injeções Espinhais , Locomoção/efeitos dos fármacos , Microscopia Confocal , Microscopia de Interferência , Norepinefrina/metabolismo , Oxigênio/metabolismo , Oxigenoterapia , RNA Mensageiro/metabolismo , Ratos , Receptor 5-HT1B de Serotonina/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Receptores 5-HT1 de Serotonina/metabolismo , Serotonina/metabolismo , Antagonistas do Receptor 5-HT1 de Serotonina/farmacologia , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Transcriptoma , Triptaminas/metabolismo , Tiramina/metabolismo
17.
Genes (Basel) ; 8(6)2017 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-28556790

RESUMO

We employed Illumina 450 K Infinium microarrays to profile DNA methylation (DNAm) in neuronal nuclei separated by fluorescence-activated sorting from the postmortem orbitofrontal cortex (OFC) of heroin users who died from heroin overdose (N = 37), suicide completers (N = 22) with no evidence of heroin use and from control subjects who did not abuse illicit drugs and died of non-suicide causes (N = 28). We identified 1298 differentially methylated CpG sites (DMSs) between heroin users and controls, and 454 DMSs between suicide completers and controls (p < 0.001). DMSs and corresponding genes (DMGs) in heroin users showed significant differences in the preferential context of hyper and hypo DM. HyperDMSs were enriched in gene bodies and exons but depleted in promoters, whereas hypoDMSs were enriched in promoters and enhancers. In addition, hyperDMGs showed preference for genes expressed specifically by glutamatergic as opposed to GABAergic neurons and enrichment for axonogenesis- and synaptic-related gene ontology categories, whereas hypoDMGs were enriched for transcription factor activity- and gene expression regulation-related terms. Finally, we found that the DNAm-based "epigenetic age" of neurons from heroin users was younger than that in controls. Suicide-related results were more difficult to interpret. Collectively, these findings suggest that the observed DNAm differences could represent functionally significant marks of heroin-associated plasticity in the OFC.

18.
Nat Commun ; 7: 10561, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26830004

RESUMO

DNA methylation (DNAm) levels lend themselves for defining an epigenetic biomarker of aging known as the 'epigenetic clock'. Our genome-wide association study (GWAS) of cerebellar epigenetic age acceleration identifies five significant (P<5.0 × 10(-8)) SNPs in two loci: 2p22.1 (inside gene DHX57) and 16p13.3 near gene MLST8 (a subunit of mTOR complex 1 and 2). We find that the SNP in 16p13.3 has a cis-acting effect on the expression levels of MLST8 (P=6.9 × 10(-18)) in most brain regions. In cerebellar samples, the SNP in 2p22.1 has a cis-effect on DHX57 (P=4.4 × 10(-5)). Gene sets found by our GWAS analysis of cerebellar age acceleration exhibit significant overlap with those of Alzheimer's disease (P=4.4 × 10(-15)), age-related macular degeneration (P=6.4 × 10(-6)), and Parkinson's disease (P=2.6 × 10(-4)). Overall, our results demonstrate the utility of a new paradigm for understanding aging and age-related diseases: it will be fruitful to use epigenetic tissue age as endophenotype in GWAS.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cerebelo/fisiologia , Epigênese Genética/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Envelhecimento , Linhagem Celular , Variação Genética , Estudo de Associação Genômica Ampla , Humanos , Desequilíbrio de Ligação , Homólogo LST8 da Proteína Associada a mTOR
19.
Schizophr Res ; 170(2-3): 235-44, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26776227

RESUMO

Increased neuronal densities in subcortical white matter have been reported for some cases with schizophrenia. The underlying cellular and molecular mechanisms remain unresolved. We exposed 26 young adult macaque monkeys for 6 months to either clozapine, haloperidol or placebo and measured by structural MRI frontal gray and white matter volumes before and after treatment, followed by observer-independent, flow-cytometry-based quantification of neuronal and non-neuronal nuclei and molecular fingerprinting of cell-type specific transcripts. After clozapine exposure, the proportion of nuclei expressing the neuronal marker NeuN increased by approximately 50% in subcortical white matter, in conjunction with a more subtle and non-significant increase in overlying gray matter. Numbers and proportions of nuclei expressing the oligodendrocyte lineage marker, OLIG2, and cell-type specific RNA expression patterns, were maintained after antipsychotic drug exposure. Frontal lobe gray and white matter volumes remained indistinguishable between antipsychotic-drug-exposed and control groups. Chronic clozapine exposure increases the proportion of NeuN+ nuclei in frontal subcortical white matter, without alterations in frontal lobe volumes or cell type-specific gene expression. Further exploration of neurochemical plasticity in non-human primate brain exposed to antipsychotic drugs is warranted.


Assuntos
Antipsicóticos/farmacologia , Encéfalo/efeitos dos fármacos , Clozapina/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Substância Branca/efeitos dos fármacos , Administração Oral , Animais , Encéfalo/anatomia & histologia , Encéfalo/metabolismo , Contagem de Células , Feminino , Citometria de Fluxo , Substância Cinzenta/anatomia & histologia , Substância Cinzenta/efeitos dos fármacos , Substância Cinzenta/metabolismo , Haloperidol/farmacologia , Imuno-Histoquímica , Macaca , Imageamento por Ressonância Magnética , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Neurônios/citologia , Neurônios/metabolismo , Oligodendroglia/citologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo , Tamanho do Órgão , Distribuição Aleatória , Substância Branca/anatomia & histologia , Substância Branca/metabolismo
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA