Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 621(7978): 389-395, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37648852

RESUMEN

Insulin resistance is the primary pathophysiology underlying metabolic syndrome and type 2 diabetes1,2. Previous metagenomic studies have described the characteristics of gut microbiota and their roles in metabolizing major nutrients in insulin resistance3-9. In particular, carbohydrate metabolism of commensals has been proposed to contribute up to 10% of the host's overall energy extraction10, thereby playing a role in the pathogenesis of obesity and prediabetes3,4,6. Nevertheless, the underlying mechanism remains unclear. Here we investigate this relationship using a comprehensive multi-omics strategy in humans. We combine unbiased faecal metabolomics with metagenomics, host metabolomics and transcriptomics data to profile the involvement of the microbiome in insulin resistance. These data reveal that faecal carbohydrates, particularly host-accessible monosaccharides, are increased in individuals with insulin resistance and are associated with microbial carbohydrate metabolisms and host inflammatory cytokines. We identify gut bacteria associated with insulin resistance and insulin sensitivity that show a distinct pattern of carbohydrate metabolism, and demonstrate that insulin-sensitivity-associated bacteria ameliorate host phenotypes of insulin resistance in a mouse model. Our study, which provides a comprehensive view of the host-microorganism relationships in insulin resistance, reveals the impact of carbohydrate metabolism by microbiota, suggesting a potential therapeutic target for ameliorating insulin resistance.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Microbioma Gastrointestinal , Resistencia a la Insulina , Animales , Humanos , Ratones , Diabetes Mellitus Tipo 2/metabolismo , Microbioma Gastrointestinal/fisiología , Resistencia a la Insulina/fisiología , Monosacáridos/metabolismo , Insulina/metabolismo , Síndrome Metabólico/metabolismo , Heces/química , Heces/microbiología , Metabolómica
2.
BMC Bioinformatics ; 24(1): 14, 2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36631751

RESUMEN

BACKGROUND: Elucidating the Transcription Factors (TFs) that drive the gene expression changes in a given experiment is a common question asked by researchers. The existing methods rely on the predicted Transcription Factor Binding Site (TFBS) to model the changes in the motif activity. Such methods only work for TFs that have a motif and assume the TF binding profile is the same in all cell types. RESULTS: Given the wealth of the ChIP-seq data available for a wide range of the TFs in various cell types, we propose that gene expression modeling can be done using ChIP-seq "signatures" directly, effectively skipping the motif finding and TFBS prediction steps. We present xcore, an R package that allows TF activity modeling based on ChIP-seq signatures and the user's gene expression data. We also provide xcoredata a companion data package that provides a collection of preprocessed ChIP-seq signatures. We demonstrate that xcore leads to biologically relevant predictions using transforming growth factor beta induced epithelial-mesenchymal transition time-courses, rinderpest infection time-courses, and embryonic stem cells differentiated to cardiomyocytes time-course profiled with Cap Analysis Gene Expression. CONCLUSIONS: xcore provides a simple analytical framework for gene expression modeling using linear models that can be easily incorporated into differential expression analysis pipelines. Taking advantage of public ChIP-seq databases, xcore can identify meaningful molecular signatures and relevant ChIP-seq experiments.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina , Factores de Transcripción , Animales , Inmunoprecipitación de Cromatina/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Unión Proteica , Expresión Génica , Sitios de Unión
3.
Genome Res ; 30(7): 951-961, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32718981

RESUMEN

Gene expression profiles in homologous tissues have been observed to be different between species, which may be due to differences between species in the gene expression program in each cell type, but may also reflect differences in cell type composition of each tissue in different species. Here, we compare expression profiles in matching primary cells in human, mouse, rat, dog, and chicken using Cap Analysis Gene Expression (CAGE) and short RNA (sRNA) sequencing data from FANTOM5. While we find that expression profiles of orthologous genes in different species are highly correlated across cell types, in each cell type many genes were differentially expressed between species. Expression of genes with products involved in transcription, RNA processing, and transcriptional regulation was more likely to be conserved, while expression of genes encoding proteins involved in intercellular communication was more likely to have diverged during evolution. Conservation of expression correlated positively with the evolutionary age of genes, suggesting that divergence in expression levels of genes critical for cell function was restricted during evolution. Motif activity analysis showed that both promoters and enhancers are activated by the same transcription factors in different species. An analysis of expression levels of mature miRNAs and of primary miRNAs identified by CAGE revealed that evolutionary old miRNAs are more likely to have conserved expression patterns than young miRNAs. We conclude that key aspects of the regulatory network are conserved, while differential expression of genes involved in cell-to-cell communication may contribute greatly to phenotypic differences between species.


Asunto(s)
Evolución Molecular , Transcriptoma , Animales , Pollos/genética , Perros , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Ratones , MicroARNs/metabolismo , Motivos de Nucleótidos , Análisis de Componente Principal , Regiones Promotoras Genéticas , Ratas , Especificidad de la Especie , Factores de Transcripción/metabolismo
4.
Genome Res ; 30(7): 1060-1072, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32718982

RESUMEN

Long noncoding RNAs (lncRNAs) constitute the majority of transcripts in the mammalian genomes, and yet, their functions remain largely unknown. As part of the FANTOM6 project, we systematically knocked down the expression of 285 lncRNAs in human dermal fibroblasts and quantified cellular growth, morphological changes, and transcriptomic responses using Capped Analysis of Gene Expression (CAGE). Antisense oligonucleotides targeting the same lncRNAs exhibited global concordance, and the molecular phenotype, measured by CAGE, recapitulated the observed cellular phenotypes while providing additional insights on the affected genes and pathways. Here, we disseminate the largest-to-date lncRNA knockdown data set with molecular phenotyping (over 1000 CAGE deep-sequencing libraries) for further exploration and highlight functional roles for ZNF213-AS1 and lnc-KHDC3L-2.


Asunto(s)
ARN Largo no Codificante/fisiología , Procesos de Crecimiento Celular/genética , Movimiento Celular/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Canales de Potasio KCNQ/metabolismo , Anotación de Secuencia Molecular , Oligonucleótidos Antisentido , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/metabolismo , ARN Interferente Pequeño
5.
Nature ; 543(7644): 199-204, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28241135

RESUMEN

Long non-coding RNAs (lncRNAs) are largely heterogeneous and functionally uncharacterized. Here, using FANTOM5 cap analysis of gene expression (CAGE) data, we integrate multiple transcript collections to generate a comprehensive atlas of 27,919 human lncRNA genes with high-confidence 5' ends and expression profiles across 1,829 samples from the major human primary cell types and tissues. Genomic and epigenomic classification of these lncRNAs reveals that most intergenic lncRNAs originate from enhancers rather than from promoters. Incorporating genetic and expression data, we show that lncRNAs overlapping trait-associated single nucleotide polymorphisms are specifically expressed in cell types relevant to the traits, implicating these lncRNAs in multiple diseases. We further demonstrate that lncRNAs overlapping expression quantitative trait loci (eQTL)-associated single nucleotide polymorphisms of messenger RNAs are co-expressed with the corresponding messenger RNAs, suggesting their potential roles in transcriptional regulation. Combining these findings with conservation data, we identify 19,175 potentially functional lncRNAs in the human genome.


Asunto(s)
Bases de Datos Genéticas , ARN Largo no Codificante/química , ARN Largo no Codificante/genética , Transcriptoma/genética , Células Cultivadas , Secuencia Conservada/genética , Conjuntos de Datos como Asunto , Elementos de Facilitación Genéticos/genética , Epigénesis Genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Genoma Humano/genética , Estudio de Asociación del Genoma Completo , Genómica , Humanos , Internet , Anotación de Secuencia Molecular , Especificidad de Órganos/genética , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas/genética , Sitios de Carácter Cuantitativo/genética , Estabilidad del ARN , ARN Mensajero/genética
6.
Int J Obes (Lond) ; 46(6): 1196-1203, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35228658

RESUMEN

BACKGROUND/OBJECTIVE: The development of overweight/obesity associates with alterations in white adipose tissue (WAT) cellularity (fat cell size/number) and lipid metabolism, in particular lipolysis. If these changes differ between early/juvenile (EOO < 18 years of age) or late onset overweight/obesity (LOO) is unknown and was presently examined. SUBJECTS/METHODS: We included 439 subjects with validated information on body mass index (BMI) at 18 years of age. Using this information and current BMI, subjects were divided into never overweight/obese (BMI < 25 kg/m2), EOO and LOO. Adipocyte size, number, morphology (size in relation to body fat) and lipolysis were determined in subcutaneous abdominal WAT. Body composition and WAT distribution was assessed by dual-X-ray absorptiometry. RESULTS: Compared with never overweight/obese, EOO and LOO displayed larger WAT amounts in all examined depots, which in subcutaneous WAT was explained by a combination of increased size and number of fat cells in EOO and LOO. EOO had 40% larger subcutaneous fat mass than LOO (p < 0.0001). Visceral WAT mass, WAT morphology and lipolysis did not differ between EOO and LOO except for minor differences in men between the two obesity groups. On average, the increase in BMI per year was 57% higher in subjects with EOO compared to LOO (p < 0.0001). CONCLUSION: Early onset overweight/obesity causes a more rapid and pronounced accumulation of subcutaneous WAT than adult onset. However, fat mass expansion measures including WAT cellularity, morphology and fat cell lipolysis do not differ in an important way suggesting that similar mechanisms of WAT growth operate in EOO and LOO.


Asunto(s)
Sobrepeso , Grasa Subcutánea , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adulto , Humanos , Masculino , Obesidad/metabolismo , Sobrepeso/metabolismo , Grasa Subcutánea/metabolismo
7.
PLoS Comput Biol ; 17(10): e1009465, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34610009

RESUMEN

Drug treatment induces cell type specific transcriptional programs, and as the number of combinations of drugs and cell types grows, the cost for exhaustive screens measuring the transcriptional drug response becomes intractable. We developed DeepCellState, a deep learning autoencoder-based framework, for predicting the induced transcriptional state in a cell type after drug treatment, based on the drug response in another cell type. Training the method on a large collection of transcriptional drug perturbation profiles, prediction accuracy improves significantly over baseline and alternative deep learning approaches when applying the method to two cell types, with improved accuracy when generalizing the framework to additional cell types. Treatments with drugs or whole drug families not seen during training are predicted with similar accuracy, and the same framework can be used for predicting the results from other interventions, such as gene knock-downs. Finally, analysis of the trained model shows that the internal representation is able to learn regulatory relationships between genes in a fully data-driven manner.


Asunto(s)
Biología Computacional/métodos , Aprendizaje Profundo , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Antineoplásicos/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Células PC-3 , Aprendizaje Automático no Supervisado
8.
PLoS Comput Biol ; 17(9): e1009376, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34491989

RESUMEN

Regulatory elements control gene expression through transcription initiation (promoters) and by enhancing transcription at distant regions (enhancers). Accurate identification of regulatory elements is fundamental for annotating genomes and understanding gene expression patterns. While there are many attempts to develop computational promoter and enhancer identification methods, reliable tools to analyze long genomic sequences are still lacking. Prediction methods often perform poorly on the genome-wide scale because the number of negatives is much higher than that in the training sets. To address this issue, we propose a dynamic negative set updating scheme with a two-model approach, using one model for scanning the genome and the other one for testing candidate positions. The developed method achieves good genome-level performance and maintains robust performance when applied to other vertebrate species, without re-training. Moreover, the unannotated predicted regulatory regions made on the human genome are enriched for disease-associated variants, suggesting them to be potentially true regulatory elements rather than false positives. We validated high scoring "false positive" predictions using reporter assay and all tested candidates were successfully validated, demonstrating the ability of our method to discover novel human regulatory regions.


Asunto(s)
Aprendizaje Profundo , Modelos Genéticos , Secuencias Reguladoras de Ácidos Nucleicos , Iniciación de la Transcripción Genética , Biología Computacional , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Genes Reporteros , Genoma Humano , Estudio de Asociación del Genoma Completo/estadística & datos numéricos , Humanos , Anotación de Secuencia Molecular , Mutación , Regiones Promotoras Genéticas
9.
PLoS Biol ; 15(9): e2002887, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28873399

RESUMEN

Cap Analysis of Gene Expression (CAGE) in combination with single-molecule sequencing technology allows precision mapping of transcription start sites (TSSs) and genome-wide capture of promoter activities in differentiated and steady state cell populations. Much less is known about whether TSS profiling can characterize diverse and non-steady state cell populations, such as the approximately 400 transitory and heterogeneous cell types that arise during ontogeny of vertebrate animals. To gain such insight, we used the chick model and performed CAGE-based TSS analysis on embryonic samples covering the full 3-week developmental period. In total, 31,863 robust TSS peaks (>1 tag per million [TPM]) were mapped to the latest chicken genome assembly, of which 34% to 46% were active in any given developmental stage. ZENBU, a web-based, open-source platform, was used for interactive data exploration. TSSs of genes critical for lineage differentiation could be precisely mapped and their activities tracked throughout development, suggesting that non-steady state and heterogeneous cell populations are amenable to CAGE-based transcriptional analysis. Our study also uncovered a large set of extremely stable housekeeping TSSs and many novel stage-specific ones. We furthermore demonstrated that TSS mapping could expedite motif-based promoter analysis for regulatory modules associated with stage-specific and housekeeping genes. Finally, using Brachyury as an example, we provide evidence that precise TSS mapping in combination with Clustered Regularly Interspaced Short Palindromic Repeat (CRISPR)-on technology enables us, for the first time, to efficiently target endogenous avian genes for transcriptional activation. Taken together, our results represent the first report of genome-wide TSS mapping in birds and the first systematic developmental TSS analysis in any amniote species (birds and mammals). By facilitating promoter-based molecular analysis and genetic manipulation, our work also underscores the value of avian models in unravelling the complex regulatory mechanism of cell lineage specification during amniote development.


Asunto(s)
Desarrollo Embrionario , Estudio de Asociación del Genoma Completo , Sitio de Iniciación de la Transcripción , Animales , Evolución Biológica , Embrión de Pollo , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas
10.
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
11.
Nature ; 507(7493): 455-461, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24670763

RESUMEN

Enhancers control the correct temporal and cell-type-specific activation of gene expression in multicellular eukaryotes. Knowing their properties, regulatory activity and targets is crucial to understand the regulation of differentiation and homeostasis. Here we use the FANTOM5 panel of samples, covering the majority of human tissues and cell types, to produce an atlas of active, in vivo-transcribed enhancers. We show that enhancers share properties with CpG-poor messenger RNA promoters but produce bidirectional, exosome-sensitive, relatively short unspliced RNAs, the generation of which is strongly related to enhancer activity. The atlas is used to compare regulatory programs between different cells at unprecedented depth, to identify disease-associated regulatory single nucleotide polymorphisms, and to classify cell-type-specific and ubiquitous enhancers. We further explore the utility of enhancer redundancy, which explains gene expression strength rather than expression patterns. The online FANTOM5 enhancer atlas represents a unique resource for studies on cell-type-specific enhancers and gene regulation.


Asunto(s)
Atlas como Asunto , Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica/genética , Anotación de Secuencia Molecular , Especificidad de Órganos , Línea Celular , Células Cultivadas , Análisis por Conglomerados , Predisposición Genética a la Enfermedad/genética , Células HeLa , Humanos , Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Sitio de Iniciación de la Transcripción , Iniciación de la Transcripción Genética
12.
PLoS Genet ; 13(3): e1006641, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28263993

RESUMEN

The FANTOM5 consortium utilised cap analysis of gene expression (CAGE) to provide an unprecedented insight into transcriptional regulation in human cells and tissues. In the current study, we have used CAGE-based transcriptional profiling on an extended dense time course of the response of human monocyte-derived macrophages grown in macrophage colony-stimulating factor (CSF1) to bacterial lipopolysaccharide (LPS). We propose that this system provides a model for the differentiation and adaptation of monocytes entering the intestinal lamina propria. The response to LPS is shown to be a cascade of successive waves of transient gene expression extending over at least 48 hours, with hundreds of positive and negative regulatory loops. Promoter analysis using motif activity response analysis (MARA) identified some of the transcription factors likely to be responsible for the temporal profile of transcriptional activation. Each LPS-inducible locus was associated with multiple inducible enhancers, and in each case, transient eRNA transcription at multiple sites detected by CAGE preceded the appearance of promoter-associated transcripts. LPS-inducible long non-coding RNAs were commonly associated with clusters of inducible enhancers. We used these data to re-examine the hundreds of loci associated with susceptibility to inflammatory bowel disease (IBD) in genome-wide association studies. Loci associated with IBD were strongly and specifically (relative to rheumatoid arthritis and unrelated traits) enriched for promoters that were regulated in monocyte differentiation or activation. Amongst previously-identified IBD susceptibility loci, the vast majority contained at least one promoter that was regulated in CSF1-dependent monocyte-macrophage transitions and/or in response to LPS. On this basis, we concluded that IBD loci are strongly-enriched for monocyte-specific genes, and identified at least 134 additional candidate genes associated with IBD susceptibility from reanalysis of published GWA studies. We propose that dysregulation of monocyte adaptation to the environment of the gastrointestinal mucosa is the key process leading to inflammatory bowel disease.


Asunto(s)
Enfermedades Inflamatorias del Intestino/genética , Macrófagos/citología , Monocitos/citología , Transcriptoma , Secuencias de Aminoácidos , Diferenciación Celular , Citocinas/metabolismo , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Genómica , Humanos , Inflamación , Enfermedades Inflamatorias del Intestino/etiología , Mucosa Intestinal/metabolismo , Ligandos , Lipopolisacáridos/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Familia de Multigenes , Regiones Promotoras Genéticas , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcripción Genética , Activación Transcripcional
13.
BMC Genomics ; 20(1): 718, 2019 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-31533632

RESUMEN

BACKGROUND: The work of the FANTOM5 Consortium has brought forth a new level of understanding of the regulation of gene transcription and the cellular processes involved in creating diversity of cell types. In this study, we extended the analysis of the FANTOM5 Cap Analysis of Gene Expression (CAGE) transcriptome data to focus on understanding the genetic regulators involved in mouse cerebellar development. RESULTS: We used the HeliScopeCAGE library sequencing on cerebellar samples over 8 embryonic and 4 early postnatal times. This study showcases temporal expression pattern changes during cerebellar development. Through a bioinformatics analysis that focused on transcription factors, their promoters and binding sites, we identified genes that appear as strong candidates for involvement in cerebellar development. We selected several candidate transcriptional regulators for validation experiments including qRT-PCR and shRNA transcript knockdown. We observed marked and reproducible developmental defects in Atf4, Rfx3, and Scrt2 knockdown embryos, which support the role of these genes in cerebellar development. CONCLUSIONS: The successful identification of these novel gene regulators in cerebellar development demonstrates that the FANTOM5 cerebellum time series is a high-quality transcriptome database for functional investigation of gene regulatory networks in cerebellar development.


Asunto(s)
Cerebelo/crecimiento & desarrollo , Perfilación de la Expresión Génica , Motivos de Nucleótidos/genética , Transcripción Genética/genética , Factor de Transcripción Activador 4/deficiencia , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Cerebelo/embriología , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Factores de Transcripción del Factor Regulador X/deficiencia , Factores de Transcripción del Factor Regulador X/genética , Factores de Transcripción del Factor Regulador X/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Nucleic Acids Res ; 45(4): e25, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-27789687

RESUMEN

Promoters and enhancers regulate the initiation of gene expression and maintenance of expression levels in spatial and temporal manner. Recent findings stemming from the Cap Analysis of Gene Expression (CAGE) demonstrate that promoters and enhancers, based on their expression profiles after stimulus, belong to different transcription response subclasses. One of the most promising biological features that might explain the difference in transcriptional response between subclasses is the local chromatin environment. We introduce a novel computational framework, PEDAL, for distinguishing effectively transcriptional profiles of promoters and enhancers using solely histone modification marks, chromatin accessibility and binding sites of transcription factors and co-activators. A case study on data from MCF-7 cell-line reveals that PEDAL can identify successfully the transcription response subclasses of promoters and enhancers from two different stimulations. Moreover, we report subsets of input markers that discriminate with minimized classification error MCF-7 promoter and enhancer transcription response subclasses. Our work provides a general computational approach for identifying effectively cell-specific and stimulation-specific promoter and enhancer transcriptional profiles, and thus, contributes to improve our understanding of transcriptional activation in human.


Asunto(s)
Biología Computacional/métodos , Elementos de Facilitación Genéticos , Regiones Promotoras Genéticas , Transcripción Genética , Algoritmos , Cromatina/genética , Factor de Crecimiento Epidérmico/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Unión Proteica , Factores de Transcripción , Activación Transcripcional , Flujo de Trabajo
15.
BMC Genomics ; 19(1): 39, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29325522

RESUMEN

CORRECTION: The authors of the original article [1] would like to recognize the critical contribution of core members of the FANTOM5 Consortium, who played the critical role of HeliScopeCAGE sequencing experiments, quality control of tag reads and processing of the raw sequencing data.

16.
J Cell Sci ; 129(13): 2573-85, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27199372

RESUMEN

Lymphangiogenesis plays a crucial role during development, in cancer metastasis and in inflammation. Activation of VEGFR-3 (also known as FLT4) by VEGF-C is one of the main drivers of lymphangiogenesis, but the transcriptional events downstream of VEGFR-3 activation are largely unknown. Recently, we identified a wave of immediate early transcription factors that are upregulated in human lymphatic endothelial cells (LECs) within the first 30 to 80 min after VEGFR-3 activation. Expression of these transcription factors must be regulated by additional pre-existing transcription factors that are rapidly activated by VEGFR-3 signaling. Using transcription factor activity analysis, we identified the homeobox transcription factor HOXD10 to be specifically activated at early time points after VEGFR-3 stimulation, and to regulate expression of immediate early transcription factors, including NR4A1. Gain- and loss-of-function studies revealed that HOXD10 is involved in LECs migration and formation of cord-like structures. Furthermore, HOXD10 regulates expression of VE-cadherin, claudin-5 and NOS3 (also known as e-NOS), and promotes lymphatic endothelial permeability. Taken together, these results reveal an important and unanticipated role of HOXD10 in the regulation of VEGFR-3 signaling in lymphatic endothelial cells, and in the control of lymphangiogenesis and permeability.


Asunto(s)
Proteínas de Homeodominio/genética , Neoplasias/genética , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Factores de Transcripción/genética , Factor C de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Línea Celular , Permeabilidad de la Membrana Celular/genética , Movimiento Celular/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Linfangiogénesis/genética , Metástasis de la Neoplasia , Neoplasias/patología , Transducción de Señal , Factor C de Crecimiento Endotelial Vascular/biosíntesis , Receptor 3 de Factores de Crecimiento Endotelial Vascular/biosíntesis
17.
Bioinformatics ; 33(23): 3696-3700, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-28961713

RESUMEN

MOTIVATION: Drug combination therapy for treatment of cancers and other multifactorial diseases has the potential of increasing the therapeutic effect, while reducing the likelihood of drug resistance. In order to reduce time and cost spent in comprehensive screens, methods are needed which can model additive effects of possible drug combinations. RESULTS: We here show that the transcriptional response to combinatorial drug treatment at promoters, as measured by single molecule CAGE technology, is accurately described by a linear combination of the responses of the individual drugs at a genome wide scale. We also find that the same linear relationship holds for transcription at enhancer elements. We conclude that the described approach is promising for eliciting the transcriptional response to multidrug treatment at promoters and enhancers in an unbiased genome wide way, which may minimize the need for exhaustive combinatorial screens. AVAILABILITY AND IMPLEMENTATION: The CAGE sequence data used in this study is available in the DDBJ Sequence Read Archive (http://trace.ddbj.nig.ac.jp/index_e.html), accession number DRP001113. CONTACT: xin.gao@kaust.edu.sa or erik.arner@riken.jp. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Elementos de Facilitación Genéticos/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Genoma Humano , Humanos , Análisis de Regresión , Transcripción Genética/efectos de los fármacos
18.
Cerebellum ; 17(3): 308-325, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29307116

RESUMEN

Laser-capture microdissection was used to isolate external germinal layer tissue from three developmental periods of mouse cerebellar development: embryonic days 13, 15, and 18. The cerebellar granule cell-enriched mRNA library was generated with next-generation sequencing using the Helicos technology. Our objective was to discover transcriptional regulators that could be important for the development of cerebellar granule cells-the most numerous neuron in the central nervous system. Through differential expression analysis, we have identified 82 differentially expressed transcription factors (TFs) from a total of 1311 differentially expressed genes. In addition, with TF-binding sequence analysis, we have identified 46 TF candidates that could be key regulators responsible for the variation in the granule cell transcriptome between developmental stages. Altogether, we identified 125 potential TFs (82 from differential expression analysis, 46 from motif analysis with 3 overlaps in the two sets). From this gene set, 37 TFs are considered novel due to the lack of previous knowledge about their roles in cerebellar development. The results from transcriptome-wide analyses were validated with existing online databases, qRT-PCR, and in situ hybridization. This study provides an initial insight into the TFs of cerebellar granule cells that might be important for development and provide valuable information for further functional studies on these transcriptional regulators.


Asunto(s)
Cerebelo/embriología , Cerebelo/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Simulación por Computador , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Hibridación in Situ , Captura por Microdisección con Láser , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
19.
Nucleic Acids Res ; 44(7): 3233-52, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-27001520

RESUMEN

Functionality of the non-coding transcripts encoded by the human genome is the coveted goal of the modern genomics research. While commonly relied on the classical methods of forward genetics, integration of different genomics datasets in a global Systems Biology fashion presents a more productive avenue of achieving this very complex aim. Here we report application of a Systems Biology-based approach to dissect functionality of a newly identified vast class of very long intergenic non-coding (vlinc) RNAs. Using highly quantitative FANTOM5 CAGE dataset, we show that these RNAs could be grouped into 1542 novel human genes based on analysis of insulators that we show here indeed function as genomic barrier elements. We show that vlinc RNAs genes likely function in cisto activate nearby genes. This effect while most pronounced in closely spaced vlinc RNA-gene pairs can be detected over relatively large genomic distances. Furthermore, we identified 101 vlinc RNA genes likely involved in early embryogenesis based on patterns of their expression and regulation. We also found another 109 such genes potentially involved in cellular functions also happening at early stages of development such as proliferation, migration and apoptosis. Overall, we show that Systems Biology-based methods have great promise for functional annotation of non-coding RNAs.


Asunto(s)
ARN Largo no Codificante/genética , Núcleo Celular/genética , Desarrollo Embrionario/genética , Regulación de la Expresión Génica , Humanos , Elementos Aisladores , Anotación de Secuencia Molecular , Regiones Promotoras Genéticas , ARN Largo no Codificante/clasificación , ARN Largo no Codificante/metabolismo , Retroviridae/genética , Biología de Sistemas , Secuencias Repetidas Terminales , Factores de Transcripción/metabolismo
20.
Nature ; 478(7367): 110-3, 2011 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-21947005

RESUMEN

Adipose tissue mass is determined by the storage and removal of triglycerides in adipocytes. Little is known, however, about adipose lipid turnover in humans in health and pathology. To study this in vivo, here we determined lipid age by measuring (14)C derived from above ground nuclear bomb tests in adipocyte lipids. We report that during the average ten-year lifespan of human adipocytes, triglycerides are renewed six times. Lipid age is independent of adipocyte size, is very stable across a wide range of adult ages and does not differ between genders. Adipocyte lipid turnover, however, is strongly related to conditions with disturbed lipid metabolism. In obesity, triglyceride removal rate (lipolysis followed by oxidation) is decreased and the amount of triglycerides stored each year is increased. In contrast, both lipid removal and storage rates are decreased in non-obese patients diagnosed with the most common hereditary form of dyslipidaemia, familial combined hyperlipidaemia. Lipid removal rate is positively correlated with the capacity of adipocytes to break down triglycerides, as assessed through lipolysis, and is inversely related to insulin resistance. Our data support a mechanism in which adipocyte lipid storage and removal have different roles in health and pathology. High storage but low triglyceride removal promotes fat tissue accumulation and obesity. Reduction of both triglyceride storage and removal decreases lipid shunting through adipose tissue and thus promotes dyslipidaemia. We identify adipocyte lipid turnover as a novel target for prevention and treatment of metabolic disease.


Asunto(s)
Tejido Adiposo/metabolismo , Salud , Metabolismo de los Lípidos , Enfermedades Metabólicas/metabolismo , Adipocitos/química , Adipocitos/metabolismo , Tejido Adiposo/citología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Radioisótopos de Carbono/análisis , Tamaño de la Célula , Senescencia Celular , Niño , Preescolar , Estudios de Cohortes , ADN/química , Dislipidemias/metabolismo , Dislipidemias/patología , Humanos , Hiperlipidemia Familiar Combinada/genética , Hiperlipidemia Familiar Combinada/metabolismo , Hiperlipidemia Familiar Combinada/patología , Lipólisis , Persona de Mediana Edad , Armas Nucleares , Obesidad/metabolismo , Grasa Subcutánea/metabolismo , Factores de Tiempo , Triglicéridos/análisis , Triglicéridos/metabolismo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA