Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Genet ; 54(7): 1037-1050, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35789323

RESUMEN

Zebrafish, a popular organism for studying embryonic development and for modeling human diseases, has so far lacked a systematic functional annotation program akin to those in other animal models. To address this, we formed the international DANIO-CODE consortium and created a central repository to store and process zebrafish developmental functional genomic data. Our data coordination center ( https://danio-code.zfin.org ) combines a total of 1,802 sets of unpublished and re-analyzed published genomic data, which we used to improve existing annotations and show its utility in experimental design. We identified over 140,000 cis-regulatory elements throughout development, including classes with distinct features dependent on their activity in time and space. We delineated the distinct distance topology and chromatin features between regulatory elements active during zygotic genome activation and those active during organogenesis. Finally, we matched regulatory elements and epigenomic landscapes between zebrafish and mouse and predicted functional relationships between them beyond sequence similarity, thus extending the utility of zebrafish developmental genomics to mammals.


Asunto(s)
Bases de Datos Genéticas , Regulación del Desarrollo de la Expresión Génica , Genoma , Genómica , Secuencias Reguladoras de Ácidos Nucleicos , Proteínas de Pez Cebra , Pez Cebra , Animales , Cromatina/genética , Genoma/genética , Humanos , Ratones , Anotación de Secuencia Molecular , Organogénesis/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
2.
Sci Rep ; 11(1): 22717, 2021 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-34811400

RESUMEN

Retinoic acid (RA) is a key signal for the specification of the pancreas. Still, the gene regulatory cascade triggered by RA in the endoderm remains poorly characterized. In this study, we investigated this regulatory network in zebrafish by combining RNA-seq, RAR ChIP-seq and ATAC-seq assays. By analysing the effect of RA and of the RA receptor (RAR) inverse-agonist BMS493 on the transcriptome and on the chromatin accessibility of endodermal cells, we identified a large set of genes and regulatory regions regulated by RA signalling. RAR ChIP-seq further defined the direct RAR target genes in zebrafish, including hox genes as well as several pancreatic regulators like mnx1, insm1b, hnf1ba and gata6. Comparison of zebrafish and murine RAR ChIP-seq data highlighted the conserved direct target genes and revealed that some RAR sites are under strong evolutionary constraints. Among them, a novel highly conserved RAR-induced enhancer was identified downstream of the HoxB locus and driving expression in the nervous system and in the gut in a RA-dependent manner. Finally, ATAC-seq data unveiled the role of the RAR-direct targets Hnf1ba and Gata6 in opening chromatin at many regulatory loci upon RA treatment.


Asunto(s)
Genómica , Páncreas/efectos de los fármacos , Receptores de Ácido Retinoico/agonistas , Transcriptoma , Tretinoina/farmacología , Pez Cebra/genética , Animales , Animales Modificados Genéticamente , Ensamble y Desensamble de Cromatina , Secuenciación de Inmunoprecipitación de Cromatina , Factores de Transcripción GATA/genética , Factores de Transcripción GATA/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Páncreas/embriología , Páncreas/metabolismo , RNA-Seq , Receptores de Ácido Retinoico/genética , Receptores de Ácido Retinoico/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Dev Cell ; 56(5): 641-656.e5, 2021 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-33651978

RESUMEN

In many animal models, primordial germ cell (PGC) development depends on maternally deposited germ plasm, which prevents somatic cell fate. Here, we show that PGCs respond to regulatory information from the germ plasm in two distinct phases using two distinct mechanisms in zebrafish. We demonstrate that PGCs commence zygotic genome activation together with the somatic blastocysts with no demonstrable differences in transcriptional and chromatin opening. Unexpectedly, both PGC and somatic blastocysts activate germ-cell-specific genes, which are only stabilized in PGCs by cytoplasmic germ plasm determinants. Disaggregated perinuclear relocalization of germ plasm during PGC migration is regulated by the germ plasm determinant Tdrd7 and is coupled to dramatic divergence between PGC and somatic transcriptomes. This transcriptional divergence relies on PGC-specific cis-regulatory elements characterized by promoter-proximal distribution. We show that Tdrd7-dependent reconfiguration of chromatin accessibility is required for elaboration of PGC fate but not for PGC migration.


Asunto(s)
Diferenciación Celular , Cromatina/genética , Células Germinativas/citología , Ribonucleoproteínas/metabolismo , Transcriptoma , Proteínas de Pez Cebra/metabolismo , Pez Cebra/crecimiento & desarrollo , Animales , Movimiento Celular , Cromatina/química , Epigénesis Genética , Genoma , Células Germinativas/metabolismo , Ribonucleoproteínas/genética , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
4.
Nature ; 564(7734): 64-70, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30464347

RESUMEN

Vertebrates have greatly elaborated the basic chordate body plan and evolved highly distinctive genomes that have been sculpted by two whole-genome duplications. Here we sequence the genome of the Mediterranean amphioxus (Branchiostoma lanceolatum) and characterize DNA methylation, chromatin accessibility, histone modifications and transcriptomes across multiple developmental stages and adult tissues to investigate the evolution of the regulation of the chordate genome. Comparisons with vertebrates identify an intermediate stage in the evolution of differentially methylated enhancers, and a high conservation of gene expression and its cis-regulatory logic between amphioxus and vertebrates that occurs maximally at an earlier mid-embryonic phylotypic period. We analyse regulatory evolution after whole-genome duplications, and find that-in vertebrates-over 80% of broadly expressed gene families with multiple paralogues derived from whole-genome duplications have members that restricted their ancestral expression, and underwent specialization rather than subfunctionalization. Counter-intuitively, paralogues that restricted their expression increased the complexity of their regulatory landscapes. These data pave the way for a better understanding of the regulatory principles that underlie key vertebrate innovations.


Asunto(s)
Regulación de la Expresión Génica , Genómica , Anfioxos/genética , Vertebrados/genética , Animales , Tipificación del Cuerpo/genética , Metilación de ADN , Humanos , Anfioxos/embriología , Anotación de Secuencia Molecular , Regiones Promotoras Genéticas , Transcriptoma/genética
5.
Nucleic Acids Res ; 42(14): 9313-26, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25030899

RESUMEN

The findings that microRNAs (miRNAs) are essential for early development in many species and that embryonic miRNAs can reprogram somatic cells into induced pluripotent stem cells suggest that these miRNAs act directly on transcriptional and chromatin regulators of pluripotency. To elucidate the transcription regulatory networks immediately downstream of embryonic miRNAs, we extended the motif activity response analysis approach that infers the regulatory impact of both transcription factors (TFs) and miRNAs from genome-wide expression states. Applying this approach to multiple experimental data sets generated from mouse embryonic stem cells (ESCs) that did or did not express miRNAs of the ESC-specific miR-290-295 cluster, we identified multiple TFs that are direct miRNA targets, some of which are known to be active during cell differentiation. Our results provide new insights into the transcription regulatory network downstream of ESC-specific miRNAs, indicating that these miRNAs act on cell cycle and chromatin regulators at several levels and downregulate TFs that are involved in the innate immune response.


Asunto(s)
Células Madre Embrionarias/metabolismo , Redes Reguladoras de Genes , MicroARNs/metabolismo , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Epigénesis Genética , Factor 2 Regulador del Interferón/metabolismo , Ratones , Células Madre Pluripotentes/metabolismo , Factor de Transcripción ReIA/metabolismo
6.
Blood ; 123(17): e68-78, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24671953

RESUMEN

CD4(+)CD25(+)FOXP3(+) human regulatory T cells (Tregs) are essential for self-tolerance and immune homeostasis. Here, we describe the promoterome of CD4(+)CD25(high)CD45RA(+) naïve and CD4(+)CD25(high)CD45RA(-) memory Tregs and their CD25(-) conventional T-cell (Tconv) counterparts both before and after in vitro expansion by cap analysis of gene expression (CAGE) adapted to single-molecule sequencing (HeliScopeCAGE). We performed comprehensive comparative digital gene expression analyses and revealed novel transcription start sites, of which several were validated as alternative promoters of known genes. For all in vitro expanded subsets, we additionally generated global maps of poised and active enhancer elements marked by histone H3 lysine 4 monomethylation and histone H3 lysine 27 acetylation, describe their cell type-specific motif signatures, and evaluate the role of candidate transcription factors STAT5, FOXP3, RUNX1, and ETS1 in both Treg- and Tconv-specific enhancer architectures. Network analyses of gene expression data revealed additional candidate transcription factors contributing to cell type specificity and a transcription factor network in Tregs that is dominated by FOXP3 interaction partners and targets. In summary, we provide a comprehensive and easily accessible resource of gene expression and gene regulation in human Treg and Tconv subpopulations.


Asunto(s)
Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Subgrupos de Linfocitos T/metabolismo , Secuencia de Bases , Sitios de Unión , Linfocitos T CD4-Positivos/citología , Bases de Datos Factuales , Epigénesis Genética , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genes Reporteros , Histonas/metabolismo , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Células Jurkat , Lisina/metabolismo , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , ARN/metabolismo , Análisis de Secuencia de ADN , Linfocitos T Reguladores/metabolismo , Factores de Transcripción/metabolismo , Transfección
7.
Blood ; 123(17): e90-9, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24671955

RESUMEN

Human blood monocytes comprise at least 3 subpopulations that differ in phenotype and function. Here, we present the first in-depth regulome analysis of human classical (CD14(++)CD16(-)), intermediate (CD14(+)CD16(+)), and nonclassical (CD14(dim)CD16(+)) monocytes. Cap analysis of gene expression adapted to Helicos single-molecule sequencing was used to map transcription start sites throughout the genome in all 3 subsets. In addition, global maps of H3K4me1 and H3K27ac deposition were generated for classical and nonclassical monocytes defining enhanceosomes of the 2 major subsets. We identified differential regulatory elements (including promoters and putative enhancers) that were associated with subset-specific motif signatures corresponding to different transcription factor activities and exemplarily validated novel downstream enhancer elements at the CD14 locus. In addition to known subset-specific features, pathway analysis revealed marked differences in metabolic gene signatures. Whereas classical monocytes expressed higher levels of genes involved in carbohydrate metabolism, priming them for anaerobic energy production, nonclassical monocytes expressed higher levels of oxidative pathway components and showed a higher mitochondrial routine activity. Our findings describe promoter/enhancer landscapes and provide novel insights into the specific biology of human monocyte subsets.


Asunto(s)
Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica , Monocitos/citología , Monocitos/metabolismo , Transcripción Genética , Secuencias de Aminoácidos , Metabolismo de los Hidratos de Carbono , Separación Celular , Citrato (si)-Sintasa/metabolismo , Epigénesis Genética , Citometría de Flujo , Regulación de la Expresión Génica , Humanos , Receptores de Lipopolisacáridos/metabolismo , Estrés Oxidativo , Regiones Promotoras Genéticas , Receptores de IgG/metabolismo , Análisis de Secuencia de ADN
8.
Nature ; 507(7493): 462-70, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24670764

RESUMEN

Regulated transcription controls the diversity, developmental pathways and spatial organization of the hundreds of cell types that make up a mammal. Using single-molecule cDNA sequencing, we mapped transcription start sites (TSSs) and their usage in human and mouse primary cells, cell lines and tissues to produce a comprehensive overview of mammalian gene expression across the human body. We find that few genes are truly 'housekeeping', whereas many mammalian promoters are composite entities composed of several closely separated TSSs, with independent cell-type-specific expression profiles. TSSs specific to different cell types evolve at different rates, whereas promoters of broadly expressed genes are the most conserved. Promoter-based expression analysis reveals key transcription factors defining cell states and links them to binding-site motifs. The functions of identified novel transcripts can be predicted by coexpression and sample ontology enrichment analyses. The functional annotation of the mammalian genome 5 (FANTOM5) project provides comprehensive expression profiles and functional annotation of mammalian cell-type-specific transcriptomes with wide applications in biomedical research.


Asunto(s)
Atlas como Asunto , Anotación de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Transcriptoma/genética , Animales , Línea Celular , Células Cultivadas , Análisis por Conglomerados , Secuencia Conservada/genética , Regulación de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Genes Esenciales/genética , Genoma/genética , Humanos , Ratones , Sistemas de Lectura Abierta/genética , Especificidad de Órganos , ARN Mensajero/análisis , ARN Mensajero/genética , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción , Transcripción Genética/genética
9.
Genome Res ; 24(5): 869-84, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24515121

RESUMEN

Accurate reconstruction of the regulatory networks that control gene expression is one of the key current challenges in molecular biology. Although gene expression and chromatin state dynamics are ultimately encoded by constellations of binding sites recognized by regulators such as transcriptions factors (TFs) and microRNAs (miRNAs), our understanding of this regulatory code and its context-dependent read-out remains very limited. Given that there are thousands of potential regulators in mammals, it is not practical to use direct experimentation to identify which of these play a key role for a particular system of interest. We developed a methodology that models gene expression or chromatin modifications in terms of genome-wide predictions of regulatory sites and completely automated it into a web-based tool called ISMARA (Integrated System for Motif Activity Response Analysis). Given only gene expression or chromatin state data across a set of samples as input, ISMARA identifies the key TFs and miRNAs driving expression/chromatin changes and makes detailed predictions regarding their regulatory roles. These include predicted activities of the regulators across the samples, their genome-wide targets, enriched gene categories among the targets, and direct interactions between the regulators. Applying ISMARA to data sets from well-studied systems, we show that it consistently identifies known key regulators ab initio. We also present a number of novel predictions including regulatory interactions in innate immunity, a master regulator of mucociliary differentiation, TFs consistently disregulated in cancer, and TFs that mediate specific chromatin modifications.


Asunto(s)
Genoma Humano , Modelos Genéticos , Motivos de Nucleótidos , Secuencias Reguladoras de Ácidos Nucleicos , Análisis de Secuencia de ADN/métodos , Algoritmos , Animales , Ensamble y Desensamble de Cromatina , Humanos , Ratones
10.
Cancer Cell ; 23(6): 768-83, 2013 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-23764001

RESUMEN

Gene expression profiling has uncovered the transcription factor Sox4 with upregulated activity during TGF-ß-induced epithelial-mesenchymal transition (EMT) in normal and cancerous breast epithelial cells. Sox4 is indispensable for EMT and cell survival in vitro and for primary tumor growth and metastasis in vivo. Among several EMT-relevant genes, Sox4 directly regulates the expression of Ezh2, encoding the Polycomb group histone methyltransferase that trimethylates histone 3 lysine 27 (H3K27me3) for gene repression. Ablation of Ezh2 expression prevents EMT, whereas forced expression of Ezh2 restores EMT in Sox4-deficient cells. Ezh2-mediated H3K27me3 marks associate with key EMT genes, representing an epigenetic EMT signature that predicts patient survival. Our results identify Sox4 as a master regulator of EMT by governing the expression of the epigenetic modifier Ezh2.


Asunto(s)
Epigénesis Genética , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Experimentales/genética , Complejo Represivo Polycomb 2/genética , Factores de Transcripción SOXC/fisiología , Animales , Línea Celular , Movimiento Celular/genética , Supervivencia Celular/genética , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , Histonas/metabolismo , Humanos , Células MCF-7 , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Metilación , Ratones , Metástasis de la Neoplasia/genética , Complejo Represivo Polycomb 2/metabolismo , Complejo Represivo Polycomb 2/fisiología , Regiones Promotoras Genéticas , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Transcripción Genética
11.
Breast Cancer Res ; 15(2): R36, 2013 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-23621987

RESUMEN

INTRODUCTION: Early pregnancy has a strong protective effect against breast cancer in humans and rodents, but the underlying mechanism is unknown. Because breast cancers are thought to arise from specific cell subpopulations of mammary epithelia, we studied the effect of parity on the transcriptome and the differentiation/proliferation potential of specific luminal and basal mammary cells in mice. METHODS: Mammary epithelial cell subpopulations (luminal Sca1-, luminal Sca1+, basal stem/progenitor, and basal myoepithelial cells) were isolated by flow cytometry from parous and age-matched virgin mice and examined by using a combination of unbiased genomics, bioinformatics, in vitro colony formation, and in vivo limiting dilution transplantation assays. Specific findings were further investigated with immunohistochemistry in entire glands of parous and age-matched virgin mice. RESULTS: Transcriptome analysis revealed an upregulation of differentiation genes and a marked decrease in the Wnt/Notch signaling ratio in basal stem/progenitor cells of parous mice. Separate bioinformatics analyses showed reduced activity for the canonical Wnt transcription factor LEF1/TCF7 and increased activity for the Wnt repressor TCF3. This finding was specific for basal stem/progenitor cells and was associated with downregulation of potentially carcinogenic pathways and a reduction in the proliferation potential of this cell subpopulation in vitro and in vivo. As a possible mechanism for decreased Wnt signaling in basal stem/progenitor cells, we found a more than threefold reduction in the expression of the secreted Wnt ligand Wnt4 in total mammary cells from parous mice, which corresponded to a similar decrease in the proportion of Wnt4-secreting and estrogen/progesterone receptor-positive cells. Because recombinant Wnt4 rescued the proliferation defect of basal stem/progenitor cells in vitro, reduced Wnt4 secretion appears to be causally related to parity-induced alterations of basal stem/progenitor cell properties in mice. CONCLUSIONS: By revealing that parity induces differentiation and downregulates the Wnt/Notch signaling ratio and the in vitro and in vivo proliferation potential of basal stem/progenitor cells in mice, our study sheds light on the long-term consequences of an early pregnancy. Furthermore, it opens the door to future studies assessing whether inhibitors of the Wnt pathway may be used to mimic the parity-induced protective effect against breast cancer.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Epitelio/patología , Glándulas Mamarias Animales/citología , Receptores Notch/metabolismo , Células Madre/citología , Proteínas Wnt/metabolismo , Animales , Antígenos Ly , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Epitelio/metabolismo , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Técnicas para Inmunoenzimas , Glándulas Mamarias Animales/metabolismo , Proteínas de la Membrana , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Paridad , Embarazo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Células Madre/metabolismo , Proteínas Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
12.
Genome Res ; 23(1): 60-73, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22964890

RESUMEN

Although changes in chromatin are integral to transcriptional reprogramming during cellular differentiation, it is currently unclear how chromatin modifications are targeted to specific loci. To systematically identify transcription factors (TFs) that can direct chromatin changes during cell fate decisions, we model the relationship between genome-wide dynamics of chromatin marks and the local occurrence of computationally predicted TF binding sites. By applying this computational approach to a time course of Polycomb-mediated H3K27me3 marks during neuronal differentiation of murine stem cells, we identify several motifs that likely regulate the dynamics of this chromatin mark. Among these, the sites bound by REST and by the SNAIL family of TFs are predicted to transiently recruit H3K27me3 in neuronal progenitors. We validate these predictions experimentally and show that absence of REST indeed causes loss of H3K27me3 at target promoters in trans, specifically at the neuronal progenitor state. Moreover, using targeted transgenic insertion, we show that promoter fragments containing REST or SNAIL binding sites are sufficient to recruit H3K27me3 in cis, while deletion of these sites results in loss of H3K27me3. These findings illustrate that the occurrence of TF binding sites can determine chromatin dynamics. Local determination of Polycomb activity by REST and SNAIL motifs exemplifies such TF based regulation of chromatin. Furthermore, our results show that key TFs can be identified ab initio through computational modeling of epigenome data sets using a modeling approach that we make readily accessible.


Asunto(s)
Ensamble y Desensamble de Cromatina , Epigénesis Genética , Modelos Genéticos , Proteínas del Grupo Polycomb/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Bovinos , Diferenciación Celular , Cromatina/metabolismo , Perros , Genoma , Histonas/metabolismo , Caballos , Humanos , Macaca , Ratones , Neuronas/citología , Zarigüeyas , Regiones Promotoras Genéticas , Factores de Transcripción de la Familia Snail , Células Madre/citología , Transgenes
13.
Nucleic Acids Res ; 41(Database issue): D214-20, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23180783

RESUMEN

Identification of genomic regulatory elements is essential for understanding the dynamics of cellular processes. This task has been substantially facilitated by the availability of genome sequences for many species and high-throughput data of transcripts and transcription factor (TF) binding. However, rigorous computational methods are necessary to derive accurate genome-wide annotations of regulatory sites from such data. SwissRegulon (http://swissregulon.unibas.ch) is a database containing genome-wide annotations of regulatory motifs, promoters and TF binding sites (TFBSs) in promoter regions across model organisms. Its binding site predictions were obtained with rigorous Bayesian probabilistic methods that operate on orthologous regions from related genomes, and use explicit evolutionary models to assess the evidence of purifying selection on each site. New in the current version of SwissRegulon is a curated collection of 190 mammalian regulatory motifs associated with ∼340 TFs, and TFBS annotations across a curated set of ∼35 000 promoters in both human and mouse. Predictions of TFBSs for Saccharomyces cerevisiae have also been significantly extended and now cover 158 of yeast's ∼180 TFs. All data are accessible through both an easily navigable genome browser with search functions, and as flat files that can be downloaded for further analysis.


Asunto(s)
Bases de Datos Genéticas , Anotación de Secuencia Molecular , Elementos Reguladores de la Transcripción , Factores de Transcripción/metabolismo , Algoritmos , Animales , Sitios de Unión , Genómica , Humanos , Internet , Ratones , Regiones Promotoras Genéticas , Regulón , Saccharomyces cerevisiae/genética , Sitio de Iniciación de la Transcripción , Interfaz Usuario-Computador
14.
Mol Cell Biol ; 32(24): 4913-24, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23028049

RESUMEN

Skeletal muscle exhibits a high plasticity and accordingly can quickly adapt to different physiological and pathological stimuli by changing its phenotype largely through diverse epigenetic mechanisms. The nuclear receptor corepressor 1 (NCoR1) has the ability to mediate gene repression; however, its role in regulating biological programs in skeletal muscle is still poorly understood. We therefore studied the mechanistic and functional aspects of NCoR1 function in this tissue. NCoR1 muscle-specific knockout mice exhibited a 7.2% higher peak oxygen consumption (VO(2peak)), a 11% reduction in maximal isometric force, and increased ex vivo fatigue resistance during maximal stimulation. Interestingly, global gene expression analysis revealed a high overlap between the effects of NCoR1 deletion and peroxisome proliferator-activated receptor gamma (PPARγ) coactivator 1α (PGC-1α) overexpression on oxidative metabolism in muscle. Importantly, PPARß/δ and estrogen-related receptor α (ERRα) were identified as common targets of NCoR1 and PGC-1α with opposing effects on the transcriptional activity of these nuclear receptors. In fact, the repressive effect of NCoR1 on oxidative phosphorylation gene expression specifically antagonizes PGC-1α-mediated coactivation of ERRα. We therefore delineated the molecular mechanism by which a transcriptional network controlled by corepressor and coactivator proteins determines the metabolic properties of skeletal muscle, thus representing a potential therapeutic target for metabolic diseases.


Asunto(s)
Músculo Esquelético/fisiología , Co-Represor 1 de Receptor Nuclear/metabolismo , Receptores de Estrógenos/metabolismo , Transactivadores/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Contracción Muscular/genética , Contracción Muscular/fisiología , Músculo Esquelético/metabolismo , Co-Represor 1 de Receptor Nuclear/deficiencia , Co-Represor 1 de Receptor Nuclear/genética , Fosforilación Oxidativa , Consumo de Oxígeno , PPAR delta/metabolismo , PPAR-beta/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Estrógenos/genética , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Factores de Transcripción , Receptor Relacionado con Estrógeno ERRalfa
15.
Diabetes ; 61(8): 1986-93, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22688341

RESUMEN

In obesity, white adipose tissue (WAT) inflammation is linked to insulin resistance. Increased adipocyte chemokine (C-C motif) ligand 2 (CCL2) secretion may initiate adipose inflammation by attracting the migration of inflammatory cells into the tissue. Using an unbiased approach, we identified adipose microRNAs (miRNAs) that are dysregulated in human obesity and assessed their possible role in controlling CCL2 production. In subcutaneous WAT obtained from 56 subjects, 11 miRNAs were present in all subjects and downregulated in obesity. Of these, 10 affected adipocyte CCL2 secretion in vitro and for 2 miRNAs (miR-126 and miR-193b), regulatory circuits were defined. While miR-126 bound directly to the 3'-untranslated region of CCL2 mRNA, miR-193b regulated CCL2 production indirectly through a network of transcription factors, many of which have been identified in other inflammatory conditions. In addition, overexpression of miR-193b and miR-126 in a human monocyte/macrophage cell line attenuated CCL2 production. The levels of the two miRNAs in subcutaneous WAT were significantly associated with CCL2 secretion (miR-193b) and expression of integrin, α-X, an inflammatory macrophage marker (miR-193b and miR-126). Taken together, our data suggest that miRNAs may be important regulators of adipose inflammation through their effects on CCL2 release from human adipocytes and macrophages.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Quimiocina CCL2/biosíntesis , MicroARNs/metabolismo , Obesidad/metabolismo , Adipocitos/metabolismo , Línea Celular , Femenino , Humanos , Inflamación/metabolismo , Resistencia a la Insulina/fisiología , Macrófagos/metabolismo
16.
Nat Med ; 18(4): 529-37, 2012 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-22388088

RESUMEN

New cancer therapies are likely to arise from an in-depth understanding of the signaling networks influencing tumor initiation, progression and metastasis. We show a fundamental role for Src-homology 2 domain-containing phosphatase 2 (SHP2) in these processes in human epidermal growth factor receptor 2 (HER2)-positive and triple-negative breast cancers. Knockdown of SHP2 eradicated breast tumor-initiating cells in xenograft models, and SHP2 depletion also prevented invasion in three-dimensional cultures and in a transductal invasion assay in vivo. Notably, SHP2 knockdown in established breast tumors blocked their growth and reduced metastasis. Mechanistically, SHP2 activated stemness-associated transcription factors, including v-myc myelocytomatosis viral oncogene homolog (c-Myc) and zinc finger E-box binding homeobox 1 (ZEB1), which resulted in the repression of let-7 microRNA and the expression of a set of 'SHP2 signature' genes. We found these genes to be simultaneously activated in a large subset of human primary breast tumors that are associated with invasive behavior and poor prognosis. These results provide new insights into the signaling cascades influencing tumor-initiating cells as well as a rationale for targeting SHP2 in breast cancer.


Asunto(s)
Neoplasias de la Mama/patología , Transformación Celular Neoplásica/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Autoantígenos/metabolismo , Caspasa 3/metabolismo , Moléculas de Adhesión Celular/metabolismo , Polaridad Celular/fisiología , Proliferación Celular , Biología Computacional , Progresión de la Enfermedad , Doxiciclina/farmacología , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones SCID , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/metabolismo , Receptor ErbB-2/metabolismo , Factores de Tiempo , Factores de Transcripción/genética , Células Tumorales Cultivadas , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , Dominios Homologos src/fisiología , Kalinina
17.
Hum Mol Genet ; 19(7): 1153-64, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20053671

RESUMEN

The loss of HBII-52 and related C/D box small nucleolar RNA (snoRNA) expression units have been implicated as a cause for the Prader-Willi syndrome (PWS). We recently found that the C/D box snoRNA HBII-52 changes the alternative splicing of the serotonin receptor 2C pre-mRNA, which is different from the traditional C/D box snoRNA function in non-mRNA methylation. Using bioinformatic predictions and experimental verification, we identified five pre-mRNAs (DPM2, TAF1, RALGPS1, PBRM1 and CRHR1) containing alternative exons that are regulated by MBII-52, the mouse homolog of HBII-52. Analysis of a single member of the MBII-52 cluster of snoRNAs by RNase protection and northern blot analysis shows that the MBII-52 expressing unit generates shorter RNAs that originate from the full-length MBII-52 snoRNA through additional processing steps. These novel RNAs associate with hnRNPs and not with proteins associated with canonical C/D box snoRNAs. Our data indicate that not a traditional C/D box snoRNA MBII-52, but a processed version lacking the snoRNA stem is the predominant MBII-52 RNA missing in PWS. This processed snoRNA functions in alternative splice-site selection. Its substitution could be a therapeutic principle for PWS.


Asunto(s)
Empalme Alternativo , Síndrome de Prader-Willi/genética , ARN Nucleolar Pequeño , Receptor de Serotonina 5-HT2C/genética , Animales , Regulación de la Expresión Génica , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Ratones , Edición de ARN , Precursores del ARN
18.
PLoS Pathog ; 5(8): e1000547, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19680535

RESUMEN

Small nucleolar RNAs (snoRNAs) are localized within the nucleolus, a sub-nuclear compartment, in which they guide ribosomal or spliceosomal RNA modifications, respectively. Up until now, snoRNAs have only been identified in eukaryal and archaeal genomes, but are notably absent in bacteria. By screening B lymphocytes for expression of non-coding RNAs (ncRNAs) induced by the Epstein-Barr virus (EBV), we here report, for the first time, the identification of a snoRNA gene within a viral genome, designated as v-snoRNA1. This genetic element displays all hallmark sequence motifs of a canonical C/D box snoRNA, namely C/C'- as well as D/D'-boxes. The nucleolar localization of v-snoRNA1 was verified by in situ hybridisation of EBV-infected cells. We also confirmed binding of the three canonical snoRNA proteins, fibrillarin, Nop56 and Nop58, to v-snoRNA1. The C-box motif of v-snoRNA1 was shown to be crucial for the stability of the viral snoRNA; its selective deletion in the viral genome led to a complete down-regulation of v-snoRNA1 expression levels within EBV-infected B cells. We further provide evidence that v-snoRNA1 might serve as a miRNA-like precursor, which is processed into 24 nt sized RNA species, designated as v-snoRNA1(24pp). A potential target site of v-snoRNA1(24pp) was identified within the 3'-UTR of BALF5 mRNA which encodes the viral DNA polymerase. V-snoRNA1 was found to be expressed in all investigated EBV-positive cell lines, including lymphoblastoid cell lines (LCL). Interestingly, induction of the lytic cycle markedly up-regulated expression levels of v-snoRNA1 up to 30-fold. By a computational approach, we identified a v-snoRNA1 homolog in the rhesus lymphocryptovirus genome. This evolutionary conservation suggests an important role of v-snoRNA1 during gamma-herpesvirus infection.


Asunto(s)
Regulación Viral de la Expresión Génica , Genoma Viral , Herpesvirus Humano 4/genética , ARN Nucleolar Pequeño/genética , Animales , Linfocitos B/virología , Secuencia de Bases , Línea Celular Tumoral , Humanos , Hibridación Fluorescente in Situ , Macaca mulatta , Datos de Secuencia Molecular , ARN Viral/genética
19.
Genome Biol ; 10(7): R79, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19624849

RESUMEN

With the advent of ultra high-throughput sequencing technologies, increasingly researchers are turning to deep sequencing for gene expression studies. Here we present a set of rigorous methods for normalization, quantification of noise, and co-expression analysis of deep sequencing data. Using these methods on 122 cap analysis of gene expression (CAGE) samples of transcription start sites, we construct genome-wide 'promoteromes' in human and mouse consisting of a three-tiered hierarchy of transcription start sites, transcription start clusters, and transcription start regions.


Asunto(s)
Perfilación de la Expresión Génica/estadística & datos numéricos , Análisis de Secuencia por Matrices de Oligonucleótidos/estadística & datos numéricos , Regiones Promotoras Genéticas/genética , Análisis de Secuencia de ADN/estadística & datos numéricos , Sitio de Iniciación de la Transcripción , Algoritmos , Animales , Composición de Base , Línea Celular , Análisis por Conglomerados , Biología Computacional/métodos , Islas de CpG/genética , Perfilación de la Expresión Génica/métodos , Estudio de Asociación del Genoma Completo/métodos , Humanos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN/métodos
20.
Genome Biol ; 10(4): R39, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19374773

RESUMEN

EdgeExpressDB is a novel database and set of interfaces for interpreting biological networks and comparing large high-throughput expression datasets that requires minimal development for new data types and search patterns. The FANTOM4 EdgeExpress database http://fantom.gsc.riken.jp/4/edgeexpress summarizes gene expression patterns in the context of alternative promoter structures and regulatory transcription factors and microRNAs using intuitive gene-centric and sub-network views. This is an important resource for gene regulation in acute myeloid leukemia, monocyte/macrophage differentiation and human transcriptional networks.


Asunto(s)
Bases de Datos Genéticas , Perfilación de la Expresión Génica/estadística & datos numéricos , Redes Reguladoras de Genes/genética , Enfermedad Aguda , Diferenciación Celular/genética , Biología Computacional/métodos , Biología Computacional/estadística & datos numéricos , Perfilación de la Expresión Génica/métodos , Genómica/métodos , Genómica/estadística & datos numéricos , Humanos , Internet , Leucemia Mieloide/genética , MicroARNs/genética , Regiones Promotoras Genéticas/genética , Programas Informáticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...