Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 179(2): 373-391.e27, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585079

RESUMEN

Cells regulate gene expression in response to salient external stimuli. In neurons, depolarization leads to the expression of inducible transcription factors (ITFs) that direct subsequent gene regulation. Depolarization encodes both a neuron's action potential (AP) output and synaptic inputs, via excitatory postsynaptic potentials (EPSPs). However, it is unclear if distinct types of electrical activity can be transformed by an ITF into distinct modes of genomic regulation. Here, we show that APs and EPSPs in mouse hippocampal neurons trigger two spatially segregated and molecularly distinct induction mechanisms that lead to the expression of the ITF NPAS4. These two pathways culminate in the formation of stimulus-specific NPAS4 heterodimers that exhibit distinct DNA binding patterns. Thus, NPAS4 differentially communicates increases in a neuron's spiking output and synaptic inputs to the nucleus, enabling gene regulation to be tailored to the type of depolarizing activity along the somato-dendritic axis of a neuron.


Asunto(s)
Potenciales de Acción , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Potenciales Postsinápticos Excitadores , Neuronas/metabolismo , Activación Transcripcional , Regiones no Traducidas 3' , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Células Cultivadas , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/fisiología , Multimerización de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley
3.
Cell ; 165(5): 1280-1292, 2016 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-27203113

RESUMEN

The cistrome is the complete set of transcription factor (TF) binding sites (cis-elements) in an organism, while an epicistrome incorporates tissue-specific DNA chemical modifications and TF-specific chemical sensitivities into these binding profiles. Robust methods to construct comprehensive cistrome and epicistrome maps are critical for elucidating complex transcriptional networks that underlie growth, behavior, and disease. Here, we describe DNA affinity purification sequencing (DAP-seq), a high-throughput TF binding site discovery method that interrogates genomic DNA with in-vitro-expressed TFs. Using DAP-seq, we defined the Arabidopsis cistrome by resolving motifs and peaks for 529 TFs. Because genomic DNA used in DAP-seq retains 5-methylcytosines, we determined that >75% (248/327) of Arabidopsis TFs surveyed were methylation sensitive, a property that strongly impacts the epicistrome landscape. DAP-seq datasets also yielded insight into the biology and binding site architecture of numerous TFs, demonstrating the value of DAP-seq for cost-effective cistromic and epicistromic annotation in any organism.


Asunto(s)
Arabidopsis/genética , ADN de Plantas/genética , Genoma de Planta , Elementos de Respuesta , Análisis de Secuencia de ADN/métodos , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , ADN de Plantas/metabolismo , Epigénesis Genética , Ácidos Indolacéticos/metabolismo , Proteínas de Plantas/genética
4.
Nature ; 624(7991): 366-377, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092913

RESUMEN

Cytosine DNA methylation is essential in brain development and is implicated in various neurological disorders. Understanding DNA methylation diversity across the entire brain in a spatial context is fundamental for a complete molecular atlas of brain cell types and their gene regulatory landscapes. Here we used single-nucleus methylome sequencing (snmC-seq3) and multi-omic sequencing (snm3C-seq)1 technologies to generate 301,626 methylomes and 176,003 chromatin conformation-methylome joint profiles from 117 dissected regions throughout the adult mouse brain. Using iterative clustering and integrating with companion whole-brain transcriptome and chromatin accessibility datasets, we constructed a methylation-based cell taxonomy with 4,673 cell groups and 274 cross-modality-annotated subclasses. We identified 2.6 million differentially methylated regions across the genome that represent potential gene regulation elements. Notably, we observed spatial cytosine methylation patterns on both genes and regulatory elements in cell types within and across brain regions. Brain-wide spatial transcriptomics data validated the association of spatial epigenetic diversity with transcription and improved the anatomical mapping of our epigenetic datasets. Furthermore, chromatin conformation diversities occurred in important neuronal genes and were highly associated with DNA methylation and transcription changes. Brain-wide cell-type comparisons enabled the construction of regulatory networks that incorporate transcription factors, regulatory elements and their potential downstream gene targets. Finally, intragenic DNA methylation and chromatin conformation patterns predicted alternative gene isoform expression observed in a whole-brain SMART-seq2 dataset. Our study establishes a brain-wide, single-cell DNA methylome and 3D multi-omic atlas and provides a valuable resource for comprehending the cellular-spatial and regulatory genome diversity of the mouse brain.


Asunto(s)
Encéfalo , Metilación de ADN , Epigenoma , Multiómica , Análisis de la Célula Individual , Animales , Ratones , Encéfalo/citología , Encéfalo/metabolismo , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Citosina/metabolismo , Conjuntos de Datos como Asunto , Factores de Transcripción/metabolismo , Transcripción Genética
5.
Nature ; 624(7991): 390-402, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092918

RESUMEN

Divergence of cis-regulatory elements drives species-specific traits1, but how this manifests in the evolution of the neocortex at the molecular and cellular level remains unclear. Here we investigated the gene regulatory programs in the primary motor cortex of human, macaque, marmoset and mouse using single-cell multiomics assays, generating gene expression, chromatin accessibility, DNA methylome and chromosomal conformation profiles from a total of over 200,000 cells. From these data, we show evidence that divergence of transcription factor expression corresponds to species-specific epigenome landscapes. We find that conserved and divergent gene regulatory features are reflected in the evolution of the three-dimensional genome. Transposable elements contribute to nearly 80% of the human-specific candidate cis-regulatory elements in cortical cells. Through machine learning, we develop sequence-based predictors of candidate cis-regulatory elements in different species and demonstrate that the genomic regulatory syntax is highly preserved from rodents to primates. Finally, we show that epigenetic conservation combined with sequence similarity helps to uncover functional cis-regulatory elements and enhances our ability to interpret genetic variants contributing to neurological disease and traits.


Asunto(s)
Secuencia Conservada , Evolución Molecular , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Mamíferos , Neocórtex , Animales , Humanos , Ratones , Callithrix/genética , Cromatina/genética , Cromatina/metabolismo , Secuencia Conservada/genética , Metilación de ADN , Elementos Transponibles de ADN/genética , Epigenoma , Regulación de la Expresión Génica/genética , Macaca/genética , Mamíferos/genética , Corteza Motora/citología , Corteza Motora/metabolismo , Multiómica , Neocórtex/citología , Neocórtex/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Variación Genética/genética
6.
Nature ; 624(7991): 355-365, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092919

RESUMEN

Single-cell analyses parse the brain's billions of neurons into thousands of 'cell-type' clusters residing in different brain structures1. Many cell types mediate their functions through targeted long-distance projections allowing interactions between specific cell types. Here we used epi-retro-seq2 to link single-cell epigenomes and cell types to long-distance projections for 33,034 neurons dissected from 32 different regions projecting to 24 different targets (225 source-to-target combinations) across the whole mouse brain. We highlight uses of these data for interrogating principles relating projection types to transcriptomics and epigenomics, and for addressing hypotheses about cell types and connections related to genetics. We provide an overall synthesis with 926 statistical comparisons of discriminability of neurons projecting to each target for every source. We integrate this dataset into the larger BRAIN Initiative Cell Census Network atlas, composed of millions of neurons, to link projection cell types to consensus clusters. Integration with spatial transcriptomics further assigns projection-enriched clusters to smaller source regions than the original dissections. We exemplify this by presenting in-depth analyses of projection neurons from the hypothalamus, thalamus, hindbrain, amygdala and midbrain to provide insights into properties of those cell types, including differentially expressed genes, their associated cis-regulatory elements and transcription-factor-binding motifs, and neurotransmitter use.


Asunto(s)
Encéfalo , Epigenómica , Vías Nerviosas , Neuronas , Animales , Ratones , Amígdala del Cerebelo , Encéfalo/citología , Encéfalo/metabolismo , Secuencia de Consenso , Conjuntos de Datos como Asunto , Perfilación de la Expresión Génica , Hipotálamo/citología , Mesencéfalo/citología , Vías Nerviosas/citología , Neuronas/metabolismo , Neurotransmisores/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Rombencéfalo/citología , Análisis de la Célula Individual , Tálamo/citología , Factores de Transcripción/metabolismo
7.
Nature ; 598(7879): 120-128, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616061

RESUMEN

Mammalian brain cells show remarkable diversity in gene expression, anatomy and function, yet the regulatory DNA landscape underlying this extensive heterogeneity is poorly understood. Here we carry out a comprehensive assessment of the epigenomes of mouse brain cell types by applying single-nucleus DNA methylation sequencing1,2 to profile 103,982 nuclei (including 95,815 neurons and 8,167 non-neuronal cells) from 45 regions of the mouse cortex, hippocampus, striatum, pallidum and olfactory areas. We identified 161 cell clusters with distinct spatial locations and projection targets. We constructed taxonomies of these epigenetic types, annotated with signature genes, regulatory elements and transcription factors. These features indicate the potential regulatory landscape supporting the assignment of putative cell types and reveal repetitive usage of regulators in excitatory and inhibitory cells for determining subtypes. The DNA methylation landscape of excitatory neurons in the cortex and hippocampus varied continuously along spatial gradients. Using this deep dataset, we constructed an artificial neural network model that precisely predicts single neuron cell-type identity and brain area spatial location. Integration of high-resolution DNA methylomes with single-nucleus chromatin accessibility data3 enabled prediction of high-confidence enhancer-gene interactions for all identified cell types, which were subsequently validated by cell-type-specific chromatin conformation capture experiments4. By combining multi-omic datasets (DNA methylation, chromatin contacts, and open chromatin) from single nuclei and annotating the regulatory genome of hundreds of cell types in the mouse brain, our DNA methylation atlas establishes the epigenetic basis for neuronal diversity and spatial organization throughout the mouse cerebrum.


Asunto(s)
Encéfalo/citología , Metilación de ADN , Epigenoma , Epigenómica , Neuronas/clasificación , Neuronas/metabolismo , Análisis de la Célula Individual , Animales , Atlas como Asunto , Encéfalo/metabolismo , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Citosina/química , Citosina/metabolismo , Conjuntos de Datos como Asunto , Giro Dentado/citología , Elementos de Facilitación Genéticos/genética , Perfilación de la Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Vías Nerviosas , Neuronas/citología
8.
Nature ; 598(7879): 167-173, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616065

RESUMEN

Neuronal cell types are classically defined by their molecular properties, anatomy and functions. Although recent advances in single-cell genomics have led to high-resolution molecular characterization of cell type diversity in the brain1, neuronal cell types are often studied out of the context of their anatomical properties. To improve our understanding of the relationship between molecular and anatomical features that define cortical neurons, here we combined retrograde labelling with single-nucleus DNA methylation sequencing to link neural epigenomic properties to projections. We examined 11,827 single neocortical neurons from 63 cortico-cortical and cortico-subcortical long-distance projections. Our results showed unique epigenetic signatures of projection neurons that correspond to their laminar and regional location and projection patterns. On the basis of their epigenomes, intra-telencephalic cells that project to different cortical targets could be further distinguished, and some layer 5 neurons that project to extra-telencephalic targets (L5 ET) formed separate clusters that aligned with their axonal projections. Such separation varied between cortical areas, which suggests that there are area-specific differences in L5 ET subtypes, which were further validated by anatomical studies. Notably, a population of cortico-cortical projection neurons clustered with L5 ET rather than intra-telencephalic neurons, which suggests that a population of L5 ET cortical neurons projects to both targets. We verified the existence of these neurons by dual retrograde labelling and anterograde tracing of cortico-cortical projection neurons, which revealed axon terminals in extra-telencephalic targets including the thalamus, superior colliculus and pons. These findings highlight the power of single-cell epigenomic approaches to connect the molecular properties of neurons with their anatomical and projection properties.


Asunto(s)
Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Epigenoma , Epigenómica , Vías Nerviosas , Neuronas/clasificación , Neuronas/metabolismo , Animales , Mapeo Encefálico , Femenino , Masculino , Ratones , Neuronas/citología
9.
Nature ; 598(7879): 111-119, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616062

RESUMEN

The primary motor cortex (M1) is essential for voluntary fine-motor control and is functionally conserved across mammals1. Here, using high-throughput transcriptomic and epigenomic profiling of more than 450,000 single nuclei in humans, marmoset monkeys and mice, we demonstrate a broadly conserved cellular makeup of this region, with similarities that mirror evolutionary distance and are consistent between the transcriptome and epigenome. The core conserved molecular identities of neuronal and non-neuronal cell types allow us to generate a cross-species consensus classification of cell types, and to infer conserved properties of cell types across species. Despite the overall conservation, however, many species-dependent specializations are apparent, including differences in cell-type proportions, gene expression, DNA methylation and chromatin state. Few cell-type marker genes are conserved across species, revealing a short list of candidate genes and regulatory mechanisms that are responsible for conserved features of homologous cell types, such as the GABAergic chandelier cells. This consensus transcriptomic classification allows us to use patch-seq (a combination of whole-cell patch-clamp recordings, RNA sequencing and morphological characterization) to identify corticospinal Betz cells from layer 5 in non-human primates and humans, and to characterize their highly specialized physiology and anatomy. These findings highlight the robust molecular underpinnings of cell-type diversity in M1 across mammals, and point to the genes and regulatory pathways responsible for the functional identity of cell types and their species-specific adaptations.


Asunto(s)
Corteza Motora/citología , Neuronas/clasificación , Análisis de la Célula Individual , Animales , Atlas como Asunto , Callithrix/genética , Epigénesis Genética , Epigenómica , Femenino , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Perfilación de la Expresión Génica , Glutamatos/metabolismo , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Persona de Mediana Edad , Corteza Motora/anatomía & histología , Neuronas/citología , Neuronas/metabolismo , Especificidad de Órganos , Filogenia , Especificidad de la Especie , Transcriptoma
10.
Nature ; 598(7879): 103-110, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616066

RESUMEN

Single-cell transcriptomics can provide quantitative molecular signatures for large, unbiased samples of the diverse cell types in the brain1-3. With the proliferation of multi-omics datasets, a major challenge is to validate and integrate results into a biological understanding of cell-type organization. Here we generated transcriptomes and epigenomes from more than 500,000 individual cells in the mouse primary motor cortex, a structure that has an evolutionarily conserved role in locomotion. We developed computational and statistical methods to integrate multimodal data and quantitatively validate cell-type reproducibility. The resulting reference atlas-containing over 56 neuronal cell types that are highly replicable across analysis methods, sequencing technologies and modalities-is a comprehensive molecular and genomic account of the diverse neuronal and non-neuronal cell types in the mouse primary motor cortex. The atlas includes a population of excitatory neurons that resemble pyramidal cells in layer 4 in other cortical regions4. We further discovered thousands of concordant marker genes and gene regulatory elements for these cell types. Our results highlight the complex molecular regulation of cell types in the brain and will directly enable the design of reagents to target specific cell types in the mouse primary motor cortex for functional analysis.


Asunto(s)
Epigenómica , Perfilación de la Expresión Génica , Corteza Motora/citología , Neuronas/clasificación , Análisis de la Célula Individual , Transcriptoma , Animales , Atlas como Asunto , Conjuntos de Datos como Asunto , Epigénesis Genética , Femenino , Masculino , Ratones , Corteza Motora/anatomía & histología , Neuronas/citología , Neuronas/metabolismo , Especificidad de Órganos , Reproducibilidad de los Resultados
14.
Nat Methods ; 16(10): 999-1006, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31501549

RESUMEN

Dynamic three-dimensional chromatin conformation is a critical mechanism for gene regulation during development and disease. Despite this, profiling of three-dimensional genome structure from complex tissues with cell-type specific resolution remains challenging. Recent efforts have demonstrated that cell-type specific epigenomic features can be resolved in complex tissues using single-cell assays. However, it remains unclear whether single-cell chromatin conformation capture (3C) or Hi-C profiles can effectively identify cell types and reconstruct cell-type specific chromatin conformation maps. To address these challenges, we have developed single-nucleus methyl-3C sequencing to capture chromatin organization and DNA methylation information and robustly separate heterogeneous cell types. Applying this method to >4,200 single human brain prefrontal cortex cells, we reconstruct cell-type specific chromatin conformation maps from 14 cortical cell types. These datasets reveal the genome-wide association between cell-type specific chromatin conformation and differential DNA methylation, suggesting pervasive interactions between epigenetic processes regulating gene expression.


Asunto(s)
Metilación de ADN , Genoma Humano , Análisis de la Célula Individual , Algoritmos , Cromatina/metabolismo , Conjuntos de Datos como Asunto , Epigénesis Genética , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos
16.
Nat Methods ; 14(8): 819-825, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28650476

RESUMEN

Broad-scale protein-protein interaction mapping is a major challenge given the cost, time, and sensitivity constraints of existing technologies. Here, we present a massively multiplexed yeast two-hybrid method, CrY2H-seq, which uses a Cre recombinase interaction reporter to intracellularly fuse the coding sequences of two interacting proteins and next-generation DNA sequencing to identify these interactions en masse. We applied CrY2H-seq to investigate sparsely annotated Arabidopsis thaliana transcription factors interactions. By performing ten independent screens testing a total of 36 million binary interaction combinations, and uncovering a network of 8,577 interactions among 1,453 transcription factors, we demonstrate CrY2H-seq's improved screening capacity, efficiency, and sensitivity over those of existing technologies. The deep-coverage network resource we call AtTFIN-1 recapitulates one-third of previously reported interactions derived from diverse methods, expands the number of known plant transcription factor interactions by three-fold, and reveals previously unknown family-specific interaction module associations with plant reproductive development, root architecture, and circadian coordination.


Asunto(s)
Arabidopsis/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Mapeo de Interacción de Proteínas/métodos , Proteoma/metabolismo , Factores de Transcripción/metabolismo , Técnicas del Sistema de Dos Híbridos , Arabidopsis/genética , Proteoma/genética , Análisis de Secuencia de ADN , Factores de Transcripción/genética
17.
Neuron ; 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38838671

RESUMEN

Altered transcriptional and epigenetic regulation of brain cell types may contribute to cognitive changes with advanced age. Using single-nucleus multi-omic DNA methylation and transcriptome sequencing (snmCT-seq) in frontal cortex from young adult and aged donors, we found widespread age- and sex-related variation in specific neuron types. The proportion of inhibitory SST- and VIP-expressing neurons was reduced in aged donors. Excitatory neurons had more profound age-related changes in their gene expression and DNA methylation than inhibitory cells. Hundreds of genes involved in synaptic activity, including EGR1, were less expressed in aged adults. Genes located in subtelomeric regions increased their expression with age and correlated with reduced telomere length. We further mapped cell-type-specific sex differences in gene expression and X-inactivation escape genes. Multi-omic single-nucleus epigenomes and transcriptomes provide new insight into the effects of age and sex on human neurons.

18.
Cell Genom ; 3(7): 100342, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37492103

RESUMEN

Single-cell sequencing could help to solve the fundamental challenge of linking millions of cell-type-specific enhancers with their target genes. However, this task is confounded by patterns of gene co-expression in much the same way that genetic correlation due to linkage disequilibrium confounds fine-mapping in genome-wide association studies (GWAS). We developed a non-parametric permutation-based procedure to establish stringent statistical criteria to control the risk of false-positive associations in enhancer-gene association studies (EGAS). We applied our procedure to large-scale transcriptome and epigenome data from multiple tissues and species, including the mouse and human brain, to predict enhancer-gene associations genome wide. We tested the functional validity of our predictions by comparing them with chromatin conformation data and causal enhancer perturbation experiments. Our study shows how controlling for gene co-expression enables robust enhancer-gene linkage using single-cell sequencing data.

19.
bioRxiv ; 2023 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-37066152

RESUMEN

Sequence divergence of cis- regulatory elements drives species-specific traits, but how this manifests in the evolution of the neocortex at the molecular and cellular level remains to be elucidated. We investigated the gene regulatory programs in the primary motor cortex of human, macaque, marmoset, and mouse with single-cell multiomics assays, generating gene expression, chromatin accessibility, DNA methylome, and chromosomal conformation profiles from a total of over 180,000 cells. For each modality, we determined species-specific, divergent, and conserved gene expression and epigenetic features at multiple levels. We find that cell type-specific gene expression evolves more rapidly than broadly expressed genes and that epigenetic status at distal candidate cis -regulatory elements (cCREs) evolves faster than promoters. Strikingly, transposable elements (TEs) contribute to nearly 80% of the human-specific cCREs in cortical cells. Through machine learning, we develop sequence-based predictors of cCREs in different species and demonstrate that the genomic regulatory syntax is highly preserved from rodents to primates. Lastly, we show that epigenetic conservation combined with sequence similarity helps uncover functional cis -regulatory elements and enhances our ability to interpret genetic variants contributing to neurological disease and traits.

20.
Science ; 382(6667): eadf5357, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37824674

RESUMEN

Delineating the gene-regulatory programs underlying complex cell types is fundamental for understanding brain function in health and disease. Here, we comprehensively examined human brain cell epigenomes by probing DNA methylation and chromatin conformation at single-cell resolution in 517 thousand cells (399 thousand neurons and 118 thousand non-neurons) from 46 regions of three adult male brains. We identified 188 cell types and characterized their molecular signatures. Integrative analyses revealed concordant changes in DNA methylation, chromatin accessibility, chromatin organization, and gene expression across cell types, cortical areas, and basal ganglia structures. We further developed single-cell methylation barcodes that reliably predict brain cell types using the methylation status of select genomic sites. This multimodal epigenomic brain cell atlas provides new insights into the complexity of cell-type-specific gene regulation in adult human brains.


Asunto(s)
Encéfalo , Metilación de ADN , Epigénesis Genética , Adulto , Humanos , Masculino , Encéfalo/citología , Encéfalo/metabolismo , Cromatina/metabolismo , Genoma Humano , Análisis de la Célula Individual , Imagenología Tridimensional , Atlas como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA