Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Cell ; 84(4): 621-639.e9, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38244545

RESUMO

The DNA-binding protein SATB2 is genetically linked to human intelligence. We studied its influence on the three-dimensional (3D) epigenome by mapping chromatin interactions and accessibility in control versus SATB2-deficient cortical neurons. We find that SATB2 affects the chromatin looping between enhancers and promoters of neuronal-activity-regulated genes, thus influencing their expression. It also alters A/B compartments, topologically associating domains, and frequently interacting regions. Genes linked to SATB2-dependent 3D genome changes are implicated in highly specialized neuronal functions and contribute to cognitive ability and risk for neuropsychiatric and neurodevelopmental disorders. Non-coding DNA regions with a SATB2-dependent structure are enriched for common variants associated with educational attainment, intelligence, and schizophrenia. Our data establish SATB2 as a cell-type-specific 3D genome modulator, which operates both independently and in cooperation with CCCTC-binding factor (CTCF) to set up the chromatin landscape of pyramidal neurons for cognitive processes.


Assuntos
Proteínas de Ligação à Região de Interação com a Matriz , Fatores de Transcrição , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Neurônios/metabolismo , Fator de Ligação a CCCTC/metabolismo , Cromatina/genética , Cromatina/metabolismo , Genoma , Cognição , Proteínas de Ligação à Região de Interação com a Matriz/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo
2.
Cell Genom ; 3(8): 100356, 2023 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-37601975

RESUMO

While germline copy-number variants (CNVs) contribute to schizophrenia (SCZ) risk, the contribution of somatic CNVs (sCNVs)-present in some but not all cells-remains unknown. We identified sCNVs using blood-derived genotype arrays from 12,834 SCZ cases and 11,648 controls, filtering sCNVs at loci recurrently mutated in clonal blood disorders. Likely early-developmental sCNVs were more common in cases (0.91%) than controls (0.51%, p = 2.68e-4), with recurrent somatic deletions of exons 1-5 of the NRXN1 gene in five SCZ cases. Hi-C maps revealed ectopic, allele-specific loops forming between a potential cryptic promoter and non-coding cis-regulatory elements upon 5' deletions in NRXN1. We also observed recurrent intragenic deletions of ABCB11, encoding a transporter implicated in anti-psychotic response, in five treatment-resistant SCZ cases and showed that ABCB11 is specifically enriched in neurons forming mesocortical and mesolimbic dopaminergic projections. Our results indicate potential roles of sCNVs in SCZ risk.

3.
Biol Psychiatry ; 92(2): 127-138, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34895699

RESUMO

BACKGROUND: Cannabis remains one of the most widely abused drugs during pregnancy. In utero exposure to its principal psychoactive component, Δ9-tetrahydrocannabinol (THC), can result in long-term neuropsychiatric risk for the progeny. This study investigated epigenetic signatures underlying these enduring consequences. METHODS: Rat dams were exposed daily to THC (0.15 mg/kg) during pregnancy, and adult male offspring were examined for reward and depressive-like behavioral endophenotypes. Using unbiased sequencing approaches, we explored transcriptional and epigenetic profiles in the nucleus accumbens (NAc), a brain area central to reward and emotional processing. An in vitro CRISPR (clustered regularly interspaced short palindromic repeats) activation model coupled with RNA sequencing was also applied to study specific consequences of epigenetic dysregulation, and altered molecular signatures were compared with human major depressive disorder transcriptome datasets. RESULTS: Prenatal THC exposure induced increased motivation for food, heightened learned helplessness and anhedonia, and altered stress sensitivity. We identified a robust increase specific to males in the expression of Kmt2a (histone-lysine N-methyltransferase 2A) that targets H3K4 (lysine 4 on histone H3) in cellular chromatin. Normalizing Kmt2a in the NAc rescued the motivational phenotype of prenatally THC-exposed animals. Comparison of RNA- and H3K4me3-sequencing datasets from the NAc of rat offspring with the in vitro model of Kmt2a upregulation revealed overlapping, significant disturbances in pathways that mediate synaptic plasticity. Similar transcriptional alterations were detected in human major depressive disorder. CONCLUSIONS: These studies provide direct evidence for the persistent effects of prenatal cannabis exposure on transcriptional and epigenetic deviations in the NAc via Kmt2a dysregulation and associated psychiatric vulnerability.


Assuntos
Cannabis , Transtorno Depressivo Maior , Animais , Transtorno Depressivo Maior/metabolismo , Dronabinol/farmacologia , Epigênese Genética , Feminino , Masculino , Motivação , Núcleo Accumbens , Gravidez , Ratos
4.
Nat Commun ; 12(1): 7243, 2021 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-34903713

RESUMO

Regulatory mechanisms associated with repeat-rich sequences and chromosomal conformations in mature neurons remain unexplored. Here, we map cell-type specific chromatin domain organization in adult mouse cerebral cortex and report strong enrichment of Endogenous Retrovirus 2 (ERV2) repeat sequences in the neuron-specific heterochromatic B2NeuN+ megabase-scaling subcompartment. Single molecule long-read sequencing and comparative Hi-C chromosomal contact mapping in wild-derived SPRET/EiJ (Mus spretus) and laboratory inbred C57BL/6J (Mus musculus) reveal neuronal reconfigurations tracking recent ERV2 expansions in the murine germline, with significantly higher B2NeuN+ contact frequencies at sites with ongoing insertions in Mus musculus. Neuronal ablation of the retrotransposon silencer Kmt1e/Setdb1 triggers B2NeuN+ disintegration and rewiring with open chromatin domains enriched for cellular stress response genes, along with severe neuroinflammation and proviral assembly with infiltration of dendrites . We conclude that neuronal megabase-scale chromosomal architectures include an evolutionarily adaptive heterochromatic organization which, upon perturbation, results in transcriptional dysregulation and unleashes ERV2 proviruses with strong neuronal tropism.


Assuntos
Cromossomos/metabolismo , Neurônios/metabolismo , Retroelementos/genética , Animais , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Cromossomos/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Retrovirus Endógenos/genética , Evolução Molecular , Amplificação de Genes , Inativação Gênica , Genes de Partícula A Intracisternal/genética , Genoma Viral/genética , Gliose/genética , Gliose/metabolismo , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Camundongos , Microglia/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/virologia , Provírus/genética , Vírion/genética , Vírion/metabolismo
5.
Nat Neurosci ; 23(4): 583-593, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32152537

RESUMO

Most risk variants for brain disorders identified by genome-wide association studies reside in the noncoding genome, which makes deciphering biological mechanisms difficult. A commonly used tool, multimarker analysis of genomic annotation (MAGMA), addresses this issue by aggregating single nucleotide polymorphism associations to nearest genes. Here we developed a platform, Hi-C-coupled MAGMA (H-MAGMA), that advances MAGMA by incorporating chromatin interaction profiles from human brain tissue across two developmental epochs and two brain cell types. By analyzing gene regulatory relationships in the disease-relevant tissue, H-MAGMA identified neurobiologically relevant target genes. We applied H-MAGMA to five psychiatric disorders and four neurodegenerative disorders to interrogate biological pathways, developmental windows and cell types implicated for each disorder. Psychiatric-disorder risk genes tended to be expressed during mid-gestation and in excitatory neurons, whereas neurodegenerative-disorder risk genes showed increasing expression over time and more diverse cell-type specificities. H-MAGMA adds to existing analytic frameworks to help identify the neurobiological principles of brain disorders.


Assuntos
Encefalopatias/genética , Encéfalo/metabolismo , Cromatina/metabolismo , Predisposição Genética para Doença , Polimorfismo de Nucleotídeo Único , Encefalopatias/metabolismo , Genômica , Humanos , Fatores de Risco
6.
Genome Med ; 12(1): 19, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32075678

RESUMO

BACKGROUND: Midbrain dopaminergic neurons (MDN) represent 0.0005% of the brain's neuronal population and mediate cognition, food intake, and metabolism. MDN are also posited to underlay the neurobiological dysfunction of schizophrenia (SCZ), a severe neuropsychiatric disorder that is characterized by psychosis as well as multifactorial medical co-morbidities, including metabolic disease, contributing to markedly increased morbidity and mortality. Paradoxically, however, the genetic risk sequences of psychosis and traits associated with metabolic disease, such as body mass, show very limited overlap. METHODS: We investigated the genomic interaction of SCZ with medical conditions and traits, including body mass index (BMI), by exploring the MDN's "spatial genome," including chromosomal contact landscapes as a critical layer of cell type-specific epigenomic regulation. Low-input Hi-C protocols were applied to 5-10 × 103 dopaminergic and other cell-specific nuclei collected by fluorescence-activated nuclei sorting from the adult human midbrain. RESULTS: The Hi-C-reconstructed MDN spatial genome revealed 11 "Euclidean hot spots" of clustered chromatin domains harboring risk sequences for SCZ and elevated BMI. Inter- and intra-chromosomal contacts interconnecting SCZ and BMI risk sequences showed massive enrichment for brain-specific expression quantitative trait loci (eQTL), with gene ontologies, regulatory motifs and proteomic interactions related to adipogenesis and lipid regulation, dopaminergic neurogenesis and neuronal connectivity, and reward- and addiction-related pathways. CONCLUSIONS: We uncovered shared nuclear topographies of cognitive and metabolic risk variants. More broadly, our PsychENCODE sponsored Hi-C study offers a novel genomic approach for the study of psychiatric and medical co-morbidities constrained by limited overlap of their respective genetic risk architectures on the linear genome.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Polimorfismo Genético , Locos de Características Quantitativas , Esquizofrenia/genética , Adipogenia , Animais , Índice de Massa Corporal , Cromossomos/genética , Cognição , Humanos , Metabolismo dos Lipídeos , Mesencéfalo/citologia , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese , Esquizofrenia/metabolismo , Esquizofrenia/patologia
7.
Schizophr Res ; 217: 26-36, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31277978

RESUMO

As expanding genetic and genomic studies continue to implicate a growing list of variants contributing risk to neuropsychiatric disease, an important next step is to understand the functional impact and points of convergence of these risk factors. Here, with a focus on schizophrenia, we survey the most recent findings of the rare and common variants underlying genetic risk for schizophrenia. We discuss the ongoing efforts to validate these variants in post-mortem brain tissue, as well as new approaches to combine CRISPR-based genome engineering with patient-specific human induced pluripotent stem cell (hiPSC)-based models, in order to identify putative causal schizophrenia loci that regulate gene expression and cellular function. We consider the current limitations of hiPSC-based approaches as well as the future advances necessary to improve the fidelity of this human model. With the objective of utilizing patient genotype data to improve diagnosis and predict treatment response, the integration of CRISPR-genome engineering and hiPSC-based models represent an important strategy with which to systematically demonstrate the cell-type-specific effects of schizophrenia-associated variants.


Assuntos
Células-Tronco Pluripotentes Induzidas , Esquizofrenia , Encéfalo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Genoma , Humanos , Esquizofrenia/genética
8.
Curr Opin Neurobiol ; 59: 112-119, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31255842

RESUMO

The 'non-linear' genome, or the spatial proximity of non-contiguous sequences, emerges as an important regulatory layer for genome organization and function, including transcriptional regulation. Here, we review recent genome-scale chromosome conformation mappings ('Hi-C') in developing and adult human and mouse brain. Neural differentiation is associated with widespread remodeling of the chromosomal contact map, reflecting dynamic changes in cell-type-specific gene expression programs, with a massive (estimated 20-50%) net loss of chromosomal contacts that is specific for the neuronal lineage. Hi-C datasets provided an unexpected link between locus-specific abnormal expansion of repeat sequences positioned at the boundaries of self-associating topological chromatin domains, and monogenic neurodevelopmental and neurodegenerative disease. Furthermore, integrative cell-type-specific Hi-C and transcriptomic analysis uncovered an expanded genomic risk space for sequences conferring liability for schizophrenia and other cognitive disease. We predict that spatial genome exploration will deliver radically new insights into the brain nucleome in health and disease.


Assuntos
Doenças Neurodegenerativas , Animais , Cromatina , Cognição , Genoma , Genômica , Humanos
9.
Dialogues Clin Neurosci ; 21(4): 407-416, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31949408

RESUMO

Schizophrenia is a debilitating psychiatric disorder with a complex genetic architecture and limited understanding of its neuropathology, reflected by the lack of diagnostic measures and effective pharmacological treatments. Geneticists have recently identified more than 145 risk loci comprising hundreds of common variants of small effect sizes, most of which lie in noncoding genomic regions. This review will discuss how the epigenetic toolbox can be applied to contextualize genetic findings in schizophrenia. Progress in next-generation sequencing, along with increasing methodological complexity, has led to the compilation of genome-wide maps of DNA methylation, histone modifications, RNA expression, and more. Integration of chromatin conformation datasets is one of the latest efforts in deciphering schizophrenia risk, allowing the identification of genes in contact with regulatory variants across 100s of kilobases. Large-scale multiomics studies will facilitate the prioritization of putative causal risk variants and gene networks that contribute to schizophrenia etiology, informing clinical diagnostics and treatment downstream.
.


La esquizofrenia es un trastorno psiquiátrico invalidante con una arquitectura genética compleja y una comprensión limitada de su neuropatología, lo que se traduce en la ausencia de mediciones diagnósticas y tratamientos farmacológicos eficaces. Recientemente los genetistas han identificado más de 145 loci de riesgo que comprenden cientos de variantes comunes de pequeño efecto, la mayoría de los cuales se encuentran en regiones genómicas no codificantes. Este artículo revisará cómo se pueden aplicar las herramientas epigenéticas para contextualizar los hallazgos genéticos en la esquizofrenia. El progreso en la secuenciación de segunda generación, junto con la creciente complejidad metodológica, ha llevado a la recopilación de mapas de metilación de ADN, modificaciones de histonas, expresión de ARN y otros más. Uno de los últimos esfuerzos para descifrar el riesgo de presentar esquizofrenia es la integración de los conjuntos de datos de conformación de cromatina, lo que permite la identificación de genes en contacto con variantes reguladoras en cientos de kilobases. Los estudios multiómicos a gran escala facilitarán la priorización de las posibles variantes de riesgo causal y las redes de genes que contribuyen a la etiología de la esquizofrenia, lo que incidirá en el diagnóstico clínico y en el tratamiento.


La schizophrénie est une maladie psychiatrique invalidante dont l'architecture génétique est complexe : sa neuropathologie est mal comprise comme le montre le manque de mesures diagnostiques et de traitements médicamenteux efficaces. Les généticiens ont récemment identifié plus de 145 loci de risque comprenant des centaines de variants courants de faible effet, la plupart situés dans des régions non codantes du génome. Cet article discute de la façon d'appliquer les outils épigénétiques pour replacer les résultats génétiques dans le contexte de la schizophrénie. Les avancées du séquençage de nouvelle génération associées à une méthodologie de plus en plus complexe, ont permis de regrouper les cartes pangénomiques de la méthylation de l'ADN, des modifications de l'histone, de l'expression de l'ARN et plus encore. L'un des derniers progrès en date dans le déchiffrage du risque schizophrénique porte sur l'intégration de séries de données sur l'architecture de la chromatine, permettant l'identification de gènes en contact avec les variants de régulation parmi des centaines de kilobases. Des études multiomiques à grande échelle aideront à prioriser les variants du risque causal putatif et les réseaux de gènes qui contribuent à l'étiologie de la schizophrénie, pour renseigner en aval les diagnostics cliniques et les traitements.


Assuntos
Metilação de DNA/genética , Epigênese Genética/genética , Predisposição Genética para Doença , Esquizofrenia/genética , Epigenômica/métodos , Humanos , Medição de Risco/métodos , Esquizofrenia/diagnóstico
10.
Science ; 362(6420)2018 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-30545851

RESUMO

To explore the developmental reorganization of the three-dimensional genome of the brain in the context of neuropsychiatric disease, we monitored chromosomal conformations in differentiating neural progenitor cells. Neuronal and glial differentiation was associated with widespread developmental remodeling of the chromosomal contact map and included interactions anchored in common variant sequences that confer heritable risk for schizophrenia. We describe cell type-specific chromosomal connectomes composed of schizophrenia risk variants and their distal targets, which altogether show enrichment for genes that regulate neuronal connectivity and chromatin remodeling, and evidence for coordinated transcriptional regulation and proteomic interaction of the participating genes. Developmentally regulated chromosomal conformation changes at schizophrenia-relevant sequences disproportionally occurred in neurons, highlighting the existence of cell type-specific disease risk vulnerabilities in spatial genome organization.


Assuntos
Cromossomos Humanos/química , Conectoma , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Predisposição Genética para Doença , Células-Tronco Neurais/citologia , Neurogênese/genética , Esquizofrenia/genética , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Células Cultivadas , Cromatina/química , Montagem e Desmontagem da Cromatina , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Masculino , Células-Tronco Neurais/metabolismo , Neuroglia/citologia , Neurônios/citologia , Neurônios/metabolismo , Conformação de Ácido Nucleico , Mapas de Interação de Proteínas/genética , Proteômica , Risco , Transcrição Gênica , Transcriptoma
11.
Prog Mol Biol Transl Sci ; 157: 21-40, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29933951

RESUMO

Chromosomal conformations, including promoter-enhancer loops, provide a critical regulatory layer for the transcriptional machinery. Therefore, schizophrenia, a common psychiatric disorder associated with broad changes in neuronal gene expression in prefrontal cortex and other brain regions implicated in psychosis, could be associated with alterations in higher-order chromatin. Here, we review early studies on spatial genome organization in the schizophrenia postmortem brain and discuss how integrative approaches using cell culture and animal model systems could gain deeper insight into the potential roles of higher-order chromatin for the neurobiology of and novel treatment avenues for common psychiatric disease.


Assuntos
Cromossomos/química , Epigenômica , Conformação de Ácido Nucleico , Esquizofrenia/genética , Animais , Modelos Animais de Doenças , Humanos , Sistema Nervoso/embriologia
12.
Stem Cell Reports ; 9(2): 615-628, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28757163

RESUMO

Modulation of transcription, either synthetic activation or repression, via dCas9-fusion proteins is a relatively new methodology with the potential to facilitate high-throughput up- or downregulation studies of gene function. Genetic studies of neurodevelopmental disorders have identified a growing list of risk variants, including both common single-nucleotide variants and rare copy-number variations, many of which are associated with genes having limited functional annotations. By applying a CRISPR-mediated gene-activation/repression platform to populations of human-induced pluripotent stem cell-derived neural progenitor cells, neurons, and astrocytes, we demonstrate that it is possible to manipulate endogenous expression levels of candidate neuropsychiatric risk genes across these three cell types. Although proof-of-concept studies using catalytically inactive Cas9-fusion proteins to modulate transcription have been reported, here we present a detailed survey of the reproducibility of gRNA positional effects across a variety of neurodevelopmental disorder-relevant risk genes, donors, neural cell types, and dCas9 effectors.


Assuntos
Astrócitos/citologia , Astrócitos/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Cálcio/metabolismo , Diferenciação Celular , Células Cultivadas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Imagem Molecular , Transcriptoma
13.
Nat Genet ; 49(8): 1239-1250, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28671686

RESUMO

We report locus-specific disintegration of megabase-scale chromosomal conformations in brain after neuronal ablation of Setdb1 (also known as Kmt1e; encodes a histone H3 lysine 9 methyltransferase), including a large topologically associated 1.2-Mb domain conserved in humans and mice that encompasses >70 genes at the clustered protocadherin locus (hereafter referred to as cPcdh). The cPcdh topologically associated domain (TADcPcdh) in neurons from mutant mice showed abnormal accumulation of the transcriptional regulator and three-dimensional (3D) genome organizer CTCF at cryptic binding sites, in conjunction with DNA cytosine hypomethylation, histone hyperacetylation and upregulated expression. Genes encoding stochastically expressed protocadherins were transcribed by increased numbers of cortical neurons, indicating relaxation of single-cell constraint. SETDB1-dependent loop formations bypassed 0.2-1 Mb of linear genome and radiated from the TADcPcdh fringes toward cis-regulatory sequences within the cPcdh locus, counterbalanced shorter-range facilitative promoter-enhancer contacts and carried loop-bound polymorphisms that were associated with genetic risk for schizophrenia. We show that the SETDB1 repressor complex, which involves multiple KRAB zinc finger proteins, shields neuronal genomes from excess CTCF binding and is critically required for structural maintenance of TADcPcdh.


Assuntos
Cromatina/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Neurônios/metabolismo , Animais , Fator de Ligação a CCCTC , Caderinas/genética , Linhagem Celular , Metilação de DNA , Epigênese Genética , Feminino , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Humanos , Masculino , Camundongos , Mutação , Conformação de Ácido Nucleico , Ligação Proteica , Domínios Proteicos , Proteínas Repressoras/metabolismo
14.
BMC Med Educ ; 16(1): 256, 2016 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-27687285

RESUMO

BACKGROUND: Trained medical interpreters are instrumental to patient satisfaction and quality of care. They are especially important in student-run clinics, where many patients have limited English proficiency. Because student-run clinics have ties to their medical schools, they have access to bilingual students who may volunteer to interpret, but are not necessarily formally trained. METHODS: To study the feasibility and efficacy of leveraging medical student volunteers to improve interpretation services, we performed a pilot study at the student-run clinic at the Icahn School of Medicine at Mount Sinai. In each fall semester in 2012-2015, we implemented a 6-h course providing didactic and interactive training on medical Spanish interpreting techniques and language skills to bilingual students. We then assessed the impact of the course on interpreter abilities. RESULTS: Participants' comfort levels, understanding of their roles, and understanding of terminology significantly increased after the course (p < 0.05), and these gains remained several months later (p < 0.05) and were repeated in an independent cohort. Patients and student clinicians also rated participants highly (averages above 4.5 out of 5) on these measures in real clinical encounters. CONCLUSIONS: These findings suggest that a formal interpreter training course tailored for medical students in the setting of a student-run clinic is feasible and effective. This program for training qualified student interpreters can serve as a model for other settings where medical students serve as interpreters.

15.
Nat Rev Neurosci ; 17(11): 681-691, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27708356

RESUMO

Nonrandom chromosomal conformations, including promoter-enhancer loopings that bypass kilobases or megabases of linear genome, provide a crucial layer of transcriptional regulation and move vast amounts of non-coding sequence into the physical proximity of genes that are important for neurodevelopment, cognition and behaviour. Activity-regulated changes in the neuronal '3D genome' could govern transcriptional mechanisms associated with learning and plasticity, and loop-bound intergenic and intronic non-coding sequences have been implicated in psychiatric and adult-onset neurodegenerative disease. Recent studies have begun to clarify the roles of spatial genome organization in normal and abnormal cognition.


Assuntos
Cognição/fisiologia , Metilação de DNA/fisiologia , Genoma/fisiologia , Transtornos do Neurodesenvolvimento/genética , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Transtornos do Neurodesenvolvimento/metabolismo , Transtornos do Neurodesenvolvimento/patologia , Neurônios/patologia , Neurônios/fisiologia , RNA não Traduzido/genética , RNA não Traduzido/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...