Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell ; 173(6): 1535-1548.e16, 2018 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-29706549

RESUMEN

Human hematopoiesis involves cellular differentiation of multipotent cells into progressively more lineage-restricted states. While the chromatin accessibility landscape of this process has been explored in defined populations, single-cell regulatory variation has been hidden by ensemble averaging. We collected single-cell chromatin accessibility profiles across 10 populations of immunophenotypically defined human hematopoietic cell types and constructed a chromatin accessibility landscape of human hematopoiesis to characterize differentiation trajectories. We find variation consistent with lineage bias toward different developmental branches in multipotent cell types. We observe heterogeneity within common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs) and develop a strategy to partition GMPs along their differentiation trajectory. Furthermore, we integrated single-cell RNA sequencing (scRNA-seq) data to associate transcription factors to chromatin accessibility changes and regulatory elements to target genes through correlations of expression and regulatory element accessibility. Overall, this work provides a framework for integrative exploration of complex regulatory dynamics in a primary human tissue at single-cell resolution.


Asunto(s)
Cromatina/química , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Análisis de la Célula Individual , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Epigénesis Genética , Epigenómica , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Humanos , Células Progenitoras Mieloides/citología , Análisis de Componente Principal , Secuencias Reguladoras de Ácidos Nucleicos , Análisis de Secuencia de ARN , Transcriptoma
2.
Cell ; 166(2): 328-342, 2016 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-27374332

RESUMEN

Metastases are the main cause of cancer deaths, but the mechanisms underlying metastatic progression remain poorly understood. We isolated pure populations of cancer cells from primary tumors and metastases from a genetically engineered mouse model of human small cell lung cancer (SCLC) to investigate the mechanisms that drive the metastatic spread of this lethal cancer. Genome-wide characterization of chromatin accessibility revealed the opening of large numbers of distal regulatory elements across the genome during metastatic progression. These changes correlate with copy number amplification of the Nfib locus, and differentially accessible sites were highly enriched for Nfib transcription factor binding sites. Nfib is necessary and sufficient to increase chromatin accessibility at a large subset of the intergenic regions. Nfib promotes pro-metastatic neuronal gene expression programs and drives the metastatic ability of SCLC cells. The identification of widespread chromatin changes during SCLC progression reveals an unexpected global reprogramming during metastatic progression.


Asunto(s)
Neoplasias Pulmonares/patología , Factores de Transcripción NFI/metabolismo , Metástasis de la Neoplasia/patología , Carcinoma Pulmonar de Células Pequeñas/patología , Secuencias de Aminoácidos , Animales , Línea Celular Tumoral , Células Cultivadas , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Factores de Transcripción NFI/genética , Regiones Promotoras Genéticas , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Regulación hacia Arriba
3.
Nat Methods ; 14(10): 975-978, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28825706

RESUMEN

Single-cell ATAC-seq (scATAC) yields sparse data that make conventional analysis challenging. We developed chromVAR (http://www.github.com/GreenleafLab/chromVAR), an R package for analyzing sparse chromatin-accessibility data by estimating gain or loss of accessibility within peaks sharing the same motif or annotation while controlling for technical biases. chromVAR enables accurate clustering of scATAC-seq profiles and characterization of known and de novo sequence motifs associated with variation in chromatin accessibility.


Asunto(s)
Epigenómica/métodos , Análisis de Secuencia de ADN/métodos , Programas Informáticos , Factores de Transcripción/metabolismo , Algoritmos , Animales , Línea Celular , Regulación de la Expresión Génica
4.
Genome Res ; 25(11): 1757-70, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26314830

RESUMEN

Transcription factors canonically bind nucleosome-free DNA, making the positioning of nucleosomes within regulatory regions crucial to the regulation of gene expression. Using the assay of transposase accessible chromatin (ATAC-seq), we observe a highly structured pattern of DNA fragment lengths and positions around nucleosomes in Saccharomyces cerevisiae, and use this distinctive two-dimensional nucleosomal "fingerprint" as the basis for a new nucleosome-positioning algorithm called NucleoATAC. We show that NucleoATAC can identify the rotational and translational positions of nucleosomes with up to base-pair resolution and provide quantitative measures of nucleosome occupancy in S. cerevisiae, Schizosaccharomyces pombe, and human cells. We demonstrate the application of NucleoATAC to a number of outstanding problems in chromatin biology, including analysis of sequence features underlying nucleosome positioning, promoter chromatin architecture across species, identification of transient changes in nucleosome occupancy and positioning during a dynamic cellular response, and integrated analysis of nucleosome occupancy and transcription factor binding.


Asunto(s)
Cromatina/química , Mapeo Cromosómico , Nucleosomas/genética , Secuencias Reguladoras de Ácidos Nucleicos , Saccharomyces cerevisiae/genética , Línea Celular , Ensamble y Desensamble de Cromatina , ADN de Hongos/genética , Bases de Datos Genéticas , Reordenamiento Génico , Humanos , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Schizosaccharomyces/genética , Análisis de Secuencia de ADN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
5.
J Bacteriol ; 199(7)2017 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-28115545

RESUMEN

The alternative sigma factor RpoS is a central regulator of many stress responses in Escherichia coli The level of functional RpoS differs depending on the stress. The effect of these differing concentrations of RpoS on global transcriptional responses remains unclear. We investigated the effect of RpoS concentration on the transcriptome during stationary phase in rich media. We found that 23% of genes in the E. coli genome are regulated by RpoS, and we identified many RpoS-transcribed genes and promoters. We observed three distinct classes of response to RpoS by genes in the regulon: genes whose expression changes linearly with increasing RpoS level, genes whose expression changes dramatically with the production of only a little RpoS ("sensitive" genes), and genes whose expression changes very little with the production of a little RpoS ("insensitive"). We show that sequences outside the core promoter region determine whether an RpoS-regulated gene is sensitive or insensitive. Moreover, we show that sensitive and insensitive genes are enriched for specific functional classes and that the sensitivity of a gene to RpoS corresponds to the timing of induction as cells enter stationary phase. Thus, promoter sensitivity to RpoS is a mechanism to coordinate specific cellular processes with growth phase and may also contribute to the diversity of stress responses directed by RpoS.IMPORTANCE The sigma factor RpoS is a global regulator that controls the response to many stresses in Escherichia coli Different stresses result in different levels of RpoS production, but the consequences of this variation are unknown. We describe how changing the level of RpoS does not influence all RpoS-regulated genes equally. The cause of this variation is likely the action of transcription factors that bind the promoters of the genes. We show that the sensitivity of a gene to RpoS levels explains the timing of expression as cells enter stationary phase and that genes with different RpoS sensitivities are enriched for specific functional groups. Thus, promoter sensitivity to RpoS is a mechanism that coordinates specific cellular processes in response to stresses.


Asunto(s)
Proteínas Bacterianas/metabolismo , Escherichia coli K12/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Estudio de Asociación del Genoma Completo , Factor sigma/metabolismo , Proteínas Bacterianas/genética , Western Blotting , Mutación , Regiones Promotoras Genéticas , Factor sigma/genética , Transcriptoma
6.
Cell Rep ; 22(3): 611-623, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29346761

RESUMEN

Adaptive survival requires the coordination of nutrient availability with expenditure of cellular resources. For example, in nutrient-limited environments, 50% of all S. cerevisiae genes synchronize and exhibit periodic bursts of expression in coordination with respiration and cell division in the yeast metabolic cycle (YMC). Despite the importance of metabolic and proliferative synchrony, the majority of YMC regulators are currently unknown. Here, we demonstrate that the INO80 chromatin-remodeling complex is required to coordinate respiration and cell division with periodic gene expression. Specifically, INO80 mutants have severe defects in oxygen consumption and promiscuous cell division that is no longer coupled with metabolic status. In mutant cells, chromatin accessibility of periodic genes, including TORC1-responsive genes, is relatively static, concomitant with severely attenuated gene expression. Collectively, these results reveal that the INO80 complex mediates metabolic signaling to chromatin to restrict proliferation to metabolically optimal states.


Asunto(s)
División Celular/genética , Ensamble y Desensamble de Cromatina/genética , Cromatina/genética , ADN Helicasas/genética , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas de Unión al ADN , Homeostasis
7.
Nat Commun ; 9(1): 4590, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389926

RESUMEN

Here we introduce Protein-indexed Assay of Transposase Accessible Chromatin with sequencing (Pi-ATAC) that combines single-cell chromatin and proteomic profiling. In conjunction with DNA transposition, the levels of multiple cell surface or intracellular protein epitopes are recorded by index flow cytometry and positions in arrayed microwells, and then subject to molecular barcoding for subsequent pooled analysis. Pi-ATAC simultaneously identifies the epigenomic and proteomic heterogeneity in individual cells. Pi-ATAC reveals a casual link between transcription factor abundance and DNA motif access, and deconvolute cell types and states in the tumor microenvironment in vivo. We identify a dominant role for hypoxia, marked by HIF1α protein, in the tumor microvenvironment for shaping the regulome in a subset of epithelial tumor cells.


Asunto(s)
ADN/genética , Ambiente , Epigenómica , Epítopos/metabolismo , Proteínas/metabolismo , Análisis de la Célula Individual , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Hipoxia de la Célula/genética , Línea Celular Tumoral , Cromatina/metabolismo , Epigénesis Genética , Molécula de Adhesión Celular Epitelial/metabolismo , Linfocitos/metabolismo , Ratones , Motivos de Nucleótidos/genética , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo , Transposasas/metabolismo
8.
Nat Med ; 24(5): 580-590, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29686426

RESUMEN

T cells create vast amounts of diversity in the genes that encode their T cell receptors (TCRs), which enables individual clones to recognize specific peptide-major histocompatibility complex (MHC) ligands. Here we combined sequencing of the TCR-encoding genes with assay for transposase-accessible chromatin with sequencing (ATAC-seq) analysis at the single-cell level to provide information on the TCR specificity and epigenomic state of individual T cells. By using this approach, termed transcript-indexed ATAC-seq (T-ATAC-seq), we identified epigenomic signatures in immortalized leukemic T cells, primary human T cells from healthy volunteers and primary leukemic T cells from patient samples. In peripheral blood CD4+ T cells from healthy individuals, we identified cis and trans regulators of naive and memory T cell states and found substantial heterogeneity in surface-marker-defined T cell populations. In patients with a leukemic form of cutaneous T cell lymphoma, T-ATAC-seq enabled identification of leukemic and nonleukemic regulatory pathways in T cells from the same individual by allowing separation of the signals that arose from the malignant clone from the background T cell noise. Thus, T-ATAC-seq is a new tool that enables analysis of epigenomic landscapes in clonal T cells and should be valuable for studies of T cell malignancy, immunity and immunotherapy.


Asunto(s)
Cromatina/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Transposasas/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Línea Celular Transformada , Células Clonales , Epigenómica , Humanos , Inmunidad , Células Jurkat , Leucemia/inmunología , Leucemia/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Análisis de la Célula Individual
9.
PLoS One ; 8(6): e66826, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23799133

RESUMEN

During embryonic development, a complex organism is formed from a single starting cell. These processes of growth and differentiation are driven by large transcriptional changes, which are following the expression and activity of transcription factors (TFs). This study sought to compare TF expression during embryonic development in a diverse group of metazoan animals: representatives of vertebrates (Danio rerio, Xenopus tropicalis), a chordate (Ciona intestinalis) and invertebrate phyla such as insects (Drosophila melanogaster, Anopheles gambiae) and nematodes (Caenorhabditis elegans) were sampled, The different species showed overall very similar TF expression patterns, with TF expression increasing during the initial stages of development. C2H2 zinc finger TFs were over-represented and Homeobox TFs were under-represented in the early stages in all species. We further clustered TFs for each species based on their quantitative temporal expression profiles. This showed very similar TF expression trends in development in vertebrate and insect species. However, analysis of the expression of orthologous pairs between more closely related species showed that expression of most individual TFs is not conserved, following the general model of duplication and diversification. The degree of similarity between TF expression between Xenopus tropicalis and Danio rerio followed the hourglass model, with the greatest similarity occuring during the early tailbud stage in Xenopus tropicalis and the late segmentation stage in Danio rerio. However, for Drosophila melanogaster and Anopheles gambiae there were two periods of high TF transcriptome similarity, one during the Arthropod phylotypic stage at 8-10 hours into Drosophila development and the other later at 16-18 hours into Drosophila development.


Asunto(s)
Desarrollo Embrionario , Factores de Transcripción/genética , Animales , Anopheles/embriología , Anopheles/genética , Anopheles/metabolismo , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Ciona intestinalis/embriología , Ciona intestinalis/genética , Ciona intestinalis/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriología , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Regulación del Desarrollo de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Transcripción/metabolismo , Transcriptoma , Xenopus/embriología , Xenopus/genética , Xenopus/metabolismo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
10.
Gene ; 486(1-2): 65-73, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21787851

RESUMEN

In humans, the enzyme telomerase (hTERT) is responsible for the synthesis of new repeat sequences at the telomeres of chromosomes. Although active in early embryogenesis, the hTERT gene is transcriptionally silenced in almost all somatic cells in the adult, but is aberrantly re-activated in over 90% of human cancers. The molecular mechanisms responsible for repression of this gene are thought to involve the transcription factor CTCF. In this study, we bioinformatically identify putative CTCF binding sites in the hTERT proximal exonic region (PER) and determine their functional relevance in mediating transcriptional silencing at this gene. Tests using a reporter gene assay in HeLa cancer cells demonstrate that a sub-region of the PER exhibits strong transcriptional repressive activity. This repression is independent of the previously identified CTCF binding site near the transcriptional start site of the hTERT gene. In addition, site directed mutagenesis of three predicted CTCF binding sites, including a previously characterized in vivo site in exon 2, does not result in a loss of the repression mediated by the PER. The results from this study indicate that expression of the hTERT gene in HeLa cells is regulated by sequences in the PER. This transcriptional control is mediated through additional regulatory molecular mechanisms, independent of CTCF binding.


Asunto(s)
Telomerasa/genética , Secuencia de Bases , Sitios de Unión/genética , Factor de Unión a CCCTC , Biología Computacional , Secuencia Conservada , Cartilla de ADN/genética , Exones , Técnicas de Silenciamiento del Gen , Redes Reguladoras de Genes , Silenciador del Gen , Células HeLa , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Plásmidos/genética , ARN Interferente Pequeño/genética , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transcripción Genética
11.
DNA Cell Biol ; 29(9): 499-508, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20438356

RESUMEN

Telomeres are regions of repeated DNA sequence that cap the ends of eukaryotic chromosomes. They act as disposable safeguards to prevent the loss of important genetic information during DNA replication due to the inability of DNA polymerase to replicate DNA to the ends of linear chromosomes. The synthesis of new telomeric repeats using an RNA molecule as a template is catalyzed by the enzyme telomerase. In embryonic stem cells, the gene encoding the catalytic protein subunit of the telomerase complex (telomere reverse transcriptase [TERT]) is transcriptionally active and critical for telomere elongation, allowing for continued cellular differentiation during development. The TERT gene is down-regulated as embryogenesis progresses to limit the proliferative capacity of cells. As a result, in normal human adult somatic cells the TERT gene is silenced. However, in over 90% of cancers, the TERT gene is reactivated, allowing cells to bypass senescence and become immortalized. In this study, we explore the molecular mechanisms that regulate transcriptional expression of the TERT gene. Bioinformatic analysis of the noncoding genomic regions around the human TERT gene identified a TERT ultra-conserved (TUC) module located 5 kb upstream of the transcription start site. This 308 bp region is over 75% conserved between distantly related mammalian species and over 91% conserved among primate species. The cis-regulatory potential of the TUC region was tested in cell-based reporter gene assays. Transient transfections into HeLa and lung fibroblast cells demonstrated that the TUC module has transcriptional enhancer activity. Further bioinformatic analysis revealed that the TUC region is highly enriched in putative transcription factor binding sites for proteins involved during hematopoiesis, indicating that the TUC module may be an enhancer for the TERT gene in specific cell lineages.


Asunto(s)
Biología Computacional , Secuencia Conservada , Elementos de Facilitación Genéticos/genética , Telomerasa/genética , Animales , Secuencia de Bases , Perros , Evolución Molecular , Regulación Enzimológica de la Expresión Génica/genética , Genes Reporteros/genética , Genoma/genética , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Primates/genética , Ratas , Transcripción Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA