Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37425940

RESUMO

Transcription factors (TFs) bind combinatorially to genomic cis-regulatory elements (cREs), orchestrating transcription programs. While studies of chromatin state and chromosomal interactions have revealed dynamic neurodevelopmental cRE landscapes, parallel understanding of the underlying TF binding lags. To elucidate the combinatorial TF-cRE interactions driving mouse basal ganglia development, we integrated ChIP-seq for twelve TFs, H3K4me3-associated enhancer-promoter interactions, chromatin and transcriptional state, and transgenic enhancer assays. We identified TF-cREs modules with distinct chromatin features and enhancer activity that have complementary roles driving GABAergic neurogenesis and suppressing other developmental fates. While the majority of distal cREs were bound by one or two TFs, a small proportion were extensively bound, and these enhancers also exhibited exceptional evolutionary conservation, motif density, and complex chromosomal interactions. Our results provide new insights into how modules of combinatorial TF-cRE interactions activate and repress developmental expression programs and demonstrate the value of TF binding data in modeling gene regulatory wiring.

2.
Proc Natl Acad Sci U S A ; 119(15): e2108760119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35377797

RESUMO

Enhancers integrate transcription factor signaling pathways that drive cell fate specification in the developing brain. We paired enhancer labeling and single-cell RNA-sequencing (scRNA-seq) to delineate and distinguish specification of neuronal lineages in mouse medial, lateral, and caudal ganglionic eminences (MGE, LGE, and CGE) at embryonic day (E)11.5. We show that scRNA-seq clustering using transcription factors improves resolution of regional and developmental populations, and that enhancer activities identify specific and overlapping GE-derived neuronal populations. First, we mapped the activities of seven evolutionarily conserved brain enhancers at single-cell resolution in vivo, finding that the selected enhancers had diverse activities in specific progenitor and neuronal populations across the GEs. We then applied enhancer-based labeling, scRNA-seq, and analysis of in situ hybridization data to distinguish transcriptionally distinct and spatially defined subtypes of MGE-derived GABAergic and cholinergic projection neurons and interneurons. Our results map developmental origins and specification paths underlying neurogenesis in the embryonic basal ganglia and showcase the power of scRNA-seq combined with enhancer-based labeling to resolve the complex paths of neuronal specification underlying mouse brain development.


Assuntos
Gânglios da Base , Neurônios Colinérgicos , Elementos Facilitadores Genéticos , Neurônios GABAérgicos , Neurogênese , Animais , Gânglios da Base/citologia , Gânglios da Base/embriologia , Linhagem da Célula/genética , Neurônios Colinérgicos/metabolismo , Neurônios GABAérgicos/metabolismo , Camundongos , Neurogênese/genética , RNA-Seq , Análise de Célula Única , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Elife ; 102021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34605404

RESUMO

Enhancers are cis-regulatory elements that play critical regulatory roles in modulating developmental transcription programs and driving cell-type-specific and context-dependent gene expression in the brain. The development of massively parallel reporter assays (MPRAs) has enabled high-throughput functional screening of candidate DNA sequences for enhancer activity. Tissue-specific screening of in vivo enhancer function at scale has the potential to greatly expand our understanding of the role of non-coding sequences in development, evolution, and disease. Here, we adapted a self-transcribing regulatory element MPRA strategy for delivery to early postnatal mouse brain via recombinant adeno-associated virus (rAAV). We identified and validated putative enhancers capable of driving reporter gene expression in mouse forebrain, including regulatory elements within an intronic CACNA1C linkage disequilibrium block associated with risk in neuropsychiatric disorder genetic studies. Paired screening and single enhancer in vivo functional testing, as we show here, represents a powerful approach towards characterizing regulatory activity of enhancers and understanding how enhancer sequences organize gene expression in the brain.


Assuntos
Encéfalo/metabolismo , Elementos Facilitadores Genéticos , Animais , Encéfalo/crescimento & desenvolvimento , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos
4.
Genome Med ; 13(1): 69, 2021 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-33910599

RESUMO

BACKGROUND: Genes with multiple co-active promoters appear common in brain, yet little is known about functional requirements for these potentially redundant genomic regulatory elements. SCN1A, which encodes the NaV1.1 sodium channel alpha subunit, is one such gene with two co-active promoters. Mutations in SCN1A are associated with epilepsy, including Dravet syndrome (DS). The majority of DS patients harbor coding mutations causing SCN1A haploinsufficiency; however, putative causal non-coding promoter mutations have been identified. METHODS: To determine the functional role of one of these potentially redundant Scn1a promoters, we focused on the non-coding Scn1a 1b regulatory region, previously described as a non-canonical alternative transcriptional start site. We generated a transgenic mouse line with deletion of the extended evolutionarily conserved 1b non-coding interval and characterized changes in gene and protein expression, and assessed seizure activity and alterations in behavior. RESULTS: Mice harboring a deletion of the 1b non-coding interval exhibited surprisingly severe reductions of Scn1a and NaV1.1 expression throughout the brain. This was accompanied by electroencephalographic and thermal-evoked seizures, and behavioral deficits. CONCLUSIONS: This work contributes to functional dissection of the regulatory wiring of a major epilepsy risk gene, SCN1A. We identified the 1b region as a critical disease-relevant regulatory element and provide evidence that non-canonical and seemingly redundant promoters can have essential function.


Assuntos
Epilepsia/genética , Regulação da Expressão Gênica , Canal de Sódio Disparado por Voltagem NAV1.1/genética , Deleção de Sequência/genética , Animais , Atenção , Sequência de Bases , Encéfalo/metabolismo , Encéfalo/patologia , Cromatina/metabolismo , Sequência Conservada/genética , Modelos Animais de Doenças , Eletroencefalografia , Epilepsia/diagnóstico por imagem , Evolução Molecular , Feminino , Células HEK293 , Heterozigoto , Homozigoto , Humanos , Masculino , Aprendizagem em Labirinto , Transtornos da Memória/genética , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Teste de Campo Aberto , Fenótipo , Ligação Proteica , Sequências Reguladoras de Ácido Nucleico/genética , Análise de Sobrevida , Temperatura , Transativadores/metabolismo
5.
Elife ; 102021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33666173

RESUMO

In utero exposure to maternal immune activation (MIA) is an environmental risk factor for neurodevelopmental and neuropsychiatric disorders. Animal models provide an opportunity to identify mechanisms driving neuropathology associated with MIA. We performed time-course transcriptional profiling of mouse cortical development following induced MIA via poly(I:C) injection at E12.5. MIA-driven transcriptional changes were validated via protein analysis, and parallel perturbations to cortical neuroanatomy were identified via imaging. MIA-induced acute upregulation of genes associated with hypoxia, immune signaling, and angiogenesis, by 6 hr following exposure. This acute response was followed by changes in proliferation, neuronal and glial specification, and cortical lamination that emerged at E14.5 and peaked at E17.5. Decreased numbers of proliferative cells in germinal zones and alterations in neuronal and glial populations were identified in the MIA-exposed cortex. Overall, paired transcriptomic and neuroanatomical characterization revealed a sequence of perturbations to corticogenesis driven by mid-gestational MIA.


Assuntos
Encéfalo/embriologia , Neurogênese , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Transtornos do Neurodesenvolvimento , Poli I-C/imunologia , Gravidez , Transcriptoma
6.
Cell Rep ; 28(8): 2048-2063.e8, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31433982

RESUMO

DLX transcription factors (TFs) are master regulators of the developing vertebrate brain, driving forebrain GABAergic neuronal differentiation. Ablation of Dlx1&2 alters expression of genes that are critical for forebrain GABAergic development. We integrated epigenomic and transcriptomic analyses, complemented with in situ hybridization (ISH), and in vivo and in vitro studies of regulatory element (RE) function. This revealed the DLX-organized gene regulatory network at genomic, cellular, and spatial levels in mouse embryonic basal ganglia. DLX TFs perform dual activating and repressing functions; the consequences of their binding were determined by the sequence and genomic context of target loci. Our results reveal and, in part, explain the paradox of widespread DLX binding contrasted with a limited subset of target loci that are sensitive at the epigenomic and transcriptomic level to Dlx1&2 ablation. The regulatory properties identified here for DLX TFs suggest general mechanisms by which TFs orchestrate dynamic expression programs underlying neurodevelopment.


Assuntos
Neurônios GABAérgicos/metabolismo , Redes Reguladoras de Genes , Genoma , Proteínas de Homeodomínio/metabolismo , Prosencéfalo/embriologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Animais , Sequência de Bases , Cromatina/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Camundongos , Modelos Genéticos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Reprodutibilidade dos Testes
7.
Nat Neurosci ; 20(8): 1062-1073, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28671691

RESUMO

The chromatin remodeling gene CHD8 represents a central node in neurodevelopmental gene networks implicated in autism. We examined the impact of germline heterozygous frameshift Chd8 mutation on neurodevelopment in mice. Chd8+/del5 mice displayed normal social interactions with no repetitive behaviors but exhibited cognitive impairment correlated with increased regional brain volume, validating that phenotypes of Chd8+/del5 mice overlap pathology reported in humans with CHD8 mutations. We applied network analysis to characterize neurodevelopmental gene expression, revealing widespread transcriptional changes in Chd8+/del5 mice across pathways disrupted in neurodevelopmental disorders, including neurogenesis, synaptic processes and neuroimmune signaling. We identified a co-expression module with peak expression in early brain development featuring dysregulation of RNA processing, chromatin remodeling and cell-cycle genes enriched for promoter binding by Chd8, and we validated increased neuronal proliferation and developmental splicing perturbation in Chd8+/del5 mice. This integrative analysis offers an initial picture of the consequences of Chd8 haploinsufficiency for brain development.


Assuntos
Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Redes Reguladoras de Genes/genética , Haploinsuficiência/genética , Animais , Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética , Cromatina/metabolismo , Camundongos Transgênicos , Mutação/genética , Fenótipo , Fatores de Transcrição/genética
8.
Neuron ; 91(6): 1260-1275, 2016 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-27657450

RESUMO

The embryonic basal ganglia generates multiple projection neurons and interneuron subtypes from distinct progenitor domains. Combinatorial interactions of transcription factors and chromatin are thought to regulate gene expression. In the medial ganglionic eminence, the NKX2-1 transcription factor controls regional identity and, with LHX6, is necessary to specify pallidal projection neurons and forebrain interneurons. Here, we dissected the molecular functions of NKX2-1 by defining its chromosomal binding, regulation of gene expression, and epigenetic state. NKX2-1 binding at distal regulatory elements led to a repressed epigenetic state and transcriptional repression in the ventricular zone. Conversely, NKX2-1 is required to establish a permissive chromatin state and transcriptional activation in the sub-ventricular and mantle zones. Moreover, combinatorial binding of NKX2-1 and LHX6 promotes transcriptionally permissive chromatin and activates genes expressed in cortical migrating interneurons. Our integrated approach provides a foundation for elucidating transcriptional networks guiding the development of the MGE and its descendants.


Assuntos
Gânglios da Base/citologia , Gânglios da Base/metabolismo , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Cromatina/metabolismo , Elementos E-Box/genética , Repressão Epigenética/genética , Proteínas de Homeodomínio/metabolismo , Interneurônios/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Fator Nuclear 1 de Tireoide
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA