Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Methods ; 20(9): 1355-1367, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37443338

RESUMEN

Joint profiling of chromatin accessibility and gene expression in individual cells provides an opportunity to decipher enhancer-driven gene regulatory networks (GRNs). Here we present a method for the inference of enhancer-driven GRNs, called SCENIC+. SCENIC+ predicts genomic enhancers along with candidate upstream transcription factors (TFs) and links these enhancers to candidate target genes. To improve both recall and precision of TF identification, we curated and clustered a motif collection with more than 30,000 motifs. We benchmarked SCENIC+ on diverse datasets from different species, including human peripheral blood mononuclear cells, ENCODE cell lines, melanoma cell states and Drosophila retinal development. Next, we exploit SCENIC+ predictions to study conserved TFs, enhancers and GRNs between human and mouse cell types in the cerebral cortex. Finally, we use SCENIC+ to study the dynamics of gene regulation along differentiation trajectories and the effect of TF perturbations on cell state. SCENIC+ is available at scenicplus.readthedocs.io .


Asunto(s)
Redes Reguladoras de Genes , Multiómica , Animales , Humanos , Ratones , Leucocitos Mononucleares , Regulación de la Expresión Génica , Cromatina/genética , Drosophila/genética , Elementos de Facilitación Genéticos
2.
Nature ; 601(7894): 630-636, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34987221

RESUMEN

The Drosophila brain is a frequently used model in neuroscience. Single-cell transcriptome analysis1-6, three-dimensional morphological classification7 and electron microscopy mapping of the connectome8,9 have revealed an immense diversity of neuronal and glial cell types that underlie an array of functional and behavioural traits in the fly. The identities of these cell types are controlled by gene regulatory networks (GRNs), involving combinations of transcription factors that bind to genomic enhancers to regulate their target genes. Here, to characterize GRNs at the cell-type level in the fly brain, we profiled the chromatin accessibility of 240,919 single cells spanning 9 developmental timepoints and integrated these data with single-cell transcriptomes. We identify more than 95,000 regulatory regions that are used in different neuronal cell types, of which 70,000 are linked to developmental trajectories involving neurogenesis, reprogramming and maturation. For 40 cell types, uniquely accessible regions were associated with their expressed transcription factors and downstream target genes through a combination of motif discovery, network inference and deep learning, creating enhancer GRNs. The enhancer architectures revealed by DeepFlyBrain lead to a better understanding of neuronal regulatory diversity and can be used to design genetic driver lines for cell types at specific timepoints, facilitating their characterization and manipulation.


Asunto(s)
Drosophila , Regulación de la Expresión Génica , Animales , Encéfalo/metabolismo , Drosophila/genética , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes/genética , Factores de Transcripción/metabolismo
3.
Nat Protoc ; 15(7): 2247-2276, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32561888

RESUMEN

This protocol explains how to perform a fast SCENIC analysis alongside standard best practices steps on single-cell RNA-sequencing data using software containers and Nextflow pipelines. SCENIC reconstructs regulons (i.e., transcription factors and their target genes) assesses the activity of these discovered regulons in individual cells and uses these cellular activity patterns to find meaningful clusters of cells. Here we present an improved version of SCENIC with several advances. SCENIC has been refactored and reimplemented in Python (pySCENIC), resulting in a tenfold increase in speed, and has been packaged into containers for ease of use. It is now also possible to use epigenomic track databases, as well as motifs, to refine regulons. In this protocol, we explain the different steps of SCENIC: the workflow starts from the count matrix depicting the gene abundances for all cells and consists of three stages. First, coexpression modules are inferred using a regression per-target approach (GRNBoost2). Next, the indirect targets are pruned from these modules using cis-regulatory motif discovery (cisTarget). Lastly, the activity of these regulons is quantified via an enrichment score for the regulon's target genes (AUCell). Nonlinear projection methods can be used to display visual groupings of cells based on the cellular activity patterns of these regulons. The results can be exported as a loom file and visualized in the SCope web application. This protocol is illustrated on two use cases: a peripheral blood mononuclear cell data set and a panel of single-cell RNA-sequencing cancer experiments. For a data set of 10,000 genes and 50,000 cells, the pipeline runs in <2 h.


Asunto(s)
Redes Reguladoras de Genes , Análisis de la Célula Individual/métodos , Flujo de Trabajo , Animales , Línea Celular Tumoral , Humanos , Ratones
4.
Mol Syst Biol ; 16(5): e9438, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32431014

RESUMEN

Single-cell technologies allow measuring chromatin accessibility and gene expression in each cell, but jointly utilizing both layers to map bona fide gene regulatory networks and enhancers remains challenging. Here, we generate independent single-cell RNA-seq and single-cell ATAC-seq atlases of the Drosophila eye-antennal disc and spatially integrate the data into a virtual latent space that mimics the organization of the 2D tissue using ScoMAP (Single-Cell Omics Mapping into spatial Axes using Pseudotime ordering). To validate spatially predicted enhancers, we use a large collection of enhancer-reporter lines and identify ~ 85% of enhancers in which chromatin accessibility and enhancer activity are coupled. Next, we infer enhancer-to-gene relationships in the virtual space, finding that genes are mostly regulated by multiple, often redundant, enhancers. Exploiting cell type-specific enhancers, we deconvolute cell type-specific effects of bulk-derived chromatin accessibility QTLs. Finally, we discover that Prospero drives neuronal differentiation through the binding of a GGG motif. In summary, we provide a comprehensive spatial characterization of gene regulation in a 2D tissue.


Asunto(s)
Cromatina/metabolismo , Drosophila/genética , Elementos de Facilitación Genéticos , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/genética , Análisis de la Célula Individual/métodos , Animales , Animales Modificados Genéticamente , Antenas de Artrópodos/metabolismo , Diferenciación Celular/genética , Cromatina/genética , Secuenciación de Inmunoprecipitación de Cromatina , Bases de Datos Genéticas , Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Epigenómica , Ojo/crecimiento & desarrollo , Ojo/metabolismo , Ontología de Genes , Redes Reguladoras de Genes , Genómica , Inmunohistoquímica , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Células Fotorreceptoras/metabolismo , Regiones Promotoras Genéticas , Sitios de Carácter Cuantitativo , Análisis Espacio-Temporal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/genética
5.
Nat Methods ; 16(5): 397-400, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962623

RESUMEN

We present cisTopic, a probabilistic framework used to simultaneously discover coaccessible enhancers and stable cell states from sparse single-cell epigenomics data ( http://github.com/aertslab/cistopic ). Using a compendium of single-cell ATAC-seq datasets from differentiating hematopoietic cells, brain and transcription factor perturbations, we demonstrate that topic modeling can be exploited for robust identification of cell types, enhancers and relevant transcription factors. cisTopic provides insight into the mechanisms underlying regulatory heterogeneity in cell populations.


Asunto(s)
Epigenómica/métodos , Perfilación de la Expresión Génica/métodos , Modelos Teóricos , Análisis de la Célula Individual/métodos , Animales , Células Sanguíneas/metabolismo , Encéfalo/metabolismo , Células Cultivadas , Análisis por Conglomerados , Redes Reguladoras de Genes/genética , Humanos , Ratones , Secuencias Reguladoras de Ácidos Nucleicos/genética , Análisis de Secuencia de ARN , Flujo de Trabajo
6.
Cell ; 174(4): 843-855.e19, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30017245

RESUMEN

Many patients with advanced cancers achieve dramatic responses to a panoply of therapeutics yet retain minimal residual disease (MRD), which ultimately results in relapse. To gain insights into the biology of MRD, we applied single-cell RNA sequencing to malignant cells isolated from BRAF mutant patient-derived xenograft melanoma cohorts exposed to concurrent RAF/MEK-inhibition. We identified distinct drug-tolerant transcriptional states, varying combinations of which co-occurred within MRDs from PDXs and biopsies of patients on treatment. One of these exhibited a neural crest stem cell (NCSC) transcriptional program largely driven by the nuclear receptor RXRG. An RXR antagonist mitigated accumulation of NCSCs in MRD and delayed the development of resistance. These data identify NCSCs as key drivers of resistance and illustrate the therapeutic potential of MRD-directed therapy. They also highlight how gene regulatory network architecture reprogramming may be therapeutically exploited to limit cellular heterogeneity, a key driver of disease progression and therapy resistance.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma/tratamiento farmacológico , Neoplasia Residual/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Receptor gamma X Retinoide/antagonistas & inhibidores , Animales , Biomarcadores de Tumor , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 1/genética , Masculino , Melanoma/metabolismo , Melanoma/patología , Ratones SCID , Mutación , Neoplasia Residual/metabolismo , Neoplasia Residual/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Nat Med ; 24(8): 1277-1289, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29988129

RESUMEN

Cancer cells are embedded in the tumor microenvironment (TME), a complex ecosystem of stromal cells. Here, we present a 52,698-cell catalog of the TME transcriptome in human lung tumors at single-cell resolution, validated in independent samples where 40,250 additional cells were sequenced. By comparing with matching non-malignant lung samples, we reveal a highly complex TME that profoundly molds stromal cells. We identify 52 stromal cell subtypes, including novel subpopulations in cell types hitherto considered to be homogeneous, as well as transcription factors underlying their heterogeneity. For instance, we discover fibroblasts expressing different collagen sets, endothelial cells downregulating immune cell homing and genes coregulated with established immune checkpoint transcripts and correlating with T-cell activity. By assessing marker genes for these cell subtypes in bulk RNA-sequencing data from 1,572 patients, we illustrate how these correlate with survival, while immunohistochemistry for selected markers validates them as separate cellular entities in an independent series of lung tumors. Hence, in providing a comprehensive catalog of stromal cells types and by characterizing their phenotype and co-optive behavior, this resource provides deeper insights into lung cancer biology that will be helpful in advancing lung cancer diagnosis and therapy.


Asunto(s)
Neoplasias Pulmonares/patología , Microambiente Tumoral , Linfocitos B/patología , Biomarcadores de Tumor/metabolismo , Regulación hacia Abajo , Células Endoteliales/patología , Fibroblastos/patología , Humanos , Pulmón/patología , Células Mieloides/patología , Neoplasias/inmunología , Neoplasias/patología , Fenotipo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Células del Estroma/patología , Análisis de Supervivencia , Linfocitos T/patología
8.
Nat Genet ; 50(7): 1011-1020, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29867222

RESUMEN

Transcriptional enhancers function as docking platforms for combinations of transcription factors (TFs) to control gene expression. How enhancer sequences determine nucleosome occupancy, TF recruitment and transcriptional activation in vivo remains unclear. Using ATAC-seq across a panel of Drosophila inbred strains, we found that SNPs affecting binding sites of the TF Grainy head (Grh) causally determine the accessibility of epithelial enhancers. We show that deletion and ectopic expression of Grh cause loss and gain of DNA accessibility, respectively. However, although Grh binding is necessary for enhancer accessibility, it is insufficient to activate enhancers. Finally, we show that human Grh homologs-GRHL1, GRHL2 and GRHL3-function similarly. We conclude that Grh binding is necessary and sufficient for the opening of epithelial enhancers but not for their activation. Our data support a model positing that complex spatiotemporal expression patterns are controlled by regulatory hierarchies in which pioneer factors, such as Grh, establish tissue-specific accessible chromatin landscapes upon which other factors can act.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Drosophila/genética , Nucleosomas/genética , Factores de Transcripción/genética , Animales , Animales Modificados Genéticamente , Sitios de Unión , Línea Celular Tumoral , Cromatina/genética , Drosophila melanogaster/genética , Elementos de Facilitación Genéticos , Células Epiteliales , Regulación del Desarrollo de la Expresión Génica , Humanos , Células MCF-7 , Polimorfismo de Nucleótido Simple , Activación Transcripcional
9.
Cell ; 174(4): 982-998.e20, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29909982

RESUMEN

The diversity of cell types and regulatory states in the brain, and how these change during aging, remains largely unknown. We present a single-cell transcriptome atlas of the entire adult Drosophila melanogaster brain sampled across its lifespan. Cell clustering identified 87 initial cell clusters that are further subclustered and validated by targeted cell-sorting. Our data show high granularity and identify a wide range of cell types. Gene network analyses using SCENIC revealed regulatory heterogeneity linked to energy consumption. During aging, RNA content declines exponentially without affecting neuronal identity in old brains. This single-cell brain atlas covers nearly all cells in the normal brain and provides the tools to study cellular diversity alongside other Drosophila and mammalian single-cell datasets in our unique single-cell analysis platform: SCope (http://scope.aertslab.org). These results, together with SCope, allow comprehensive exploration of all transcriptional states of an entire aging brain.


Asunto(s)
Envejecimiento , Encéfalo/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Redes Reguladoras de Genes , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Drosophila melanogaster/fisiología , Femenino , Perfilación de la Expresión Génica , Masculino
10.
Brief Funct Genomics ; 17(4): 246-254, 2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29342231

RESUMEN

Single-cell techniques are advancing rapidly and are yielding unprecedented insight into cellular heterogeneity. Mapping the gene regulatory networks (GRNs) underlying cell states provides attractive opportunities to mechanistically understand this heterogeneity. In this review, we discuss recently emerging methods to map GRNs from single-cell transcriptomics data, tackling the challenge of increased noise levels and data sparsity compared with bulk data, alongside increasing data volumes. Next, we discuss how new techniques for single-cell epigenomics, such as single-cell ATAC-seq and single-cell DNA methylation profiling, can be used to decipher gene regulatory programmes. We finally look forward to the application of single-cell multi-omics and perturbation techniques that will likely play important roles for GRN inference in the future.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Epigenómica/métodos
11.
Nat Methods ; 14(11): 1083-1086, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28991892

RESUMEN

We present SCENIC, a computational method for simultaneous gene regulatory network reconstruction and cell-state identification from single-cell RNA-seq data (http://scenic.aertslab.org). On a compendium of single-cell data from tumors and brain, we demonstrate that cis-regulatory analysis can be exploited to guide the identification of transcription factors and cell states. SCENIC provides critical biological insights into the mechanisms driving cellular heterogeneity.


Asunto(s)
Redes Reguladoras de Genes , Análisis de la Célula Individual , Algoritmos , Animales , Encéfalo/metabolismo , Análisis por Conglomerados , Perfilación de la Expresión Génica , Humanos , Ratones
12.
J Clin Invest ; 127(6): 2310-2325, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28463229

RESUMEN

Identification and functional validation of oncogenic drivers are essential steps toward advancing cancer precision medicine. Here, we have presented a comprehensive analysis of the somatic genomic landscape of the widely used BRAFV600E- and NRASQ61K-driven mouse models of melanoma. By integrating the data with publically available genomic, epigenomic, and transcriptomic information from human clinical samples, we confirmed the importance of several genes and pathways previously implicated in human melanoma, including the tumor-suppressor genes phosphatase and tensin homolog (PTEN), cyclin dependent kinase inhibitor 2A (CDKN2A), LKB1, and others. Importantly, this approach also identified additional putative melanoma drivers with prognostic and therapeutic relevance. Surprisingly, one of these genes encodes the tyrosine kinase FES. Whereas FES is highly expressed in normal human melanocytes, FES expression is strongly decreased in over 30% of human melanomas. This downregulation correlates with poor overall survival. Correspondingly, engineered deletion of Fes accelerated tumor progression in a BRAFV600E-driven mouse model of melanoma. Together, these data implicate FES as a driver of melanoma progression and demonstrate the potential of cross-species oncogenomic approaches combined with mouse modeling to uncover impactful mutations and oncogenic driver alleles with clinical importance in the treatment of human cancer.


Asunto(s)
Melanoma/genética , Proteínas Proto-Oncogénicas c-fes/genética , Neoplasias Cutáneas/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Variaciones en el Número de Copia de ADN , Genes Supresores de Tumor , Genómica , Humanos , Melanoma/metabolismo , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias , Oncogenes , Proteínas Proto-Oncogénicas c-fes/metabolismo , Neoplasias Cutáneas/metabolismo , Vía de Señalización Wnt
13.
Nat Commun ; 7: 12846, 2016 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-27667448

RESUMEN

Gene expression data are accumulating exponentially in public repositories. Reanalysis and integration of themed collections from these studies may provide new insights, but requires further human curation. Here we report a crowdsourcing project to annotate and reanalyse a large number of gene expression profiles from Gene Expression Omnibus (GEO). Through a massive open online course on Coursera, over 70 participants from over 25 countries identify and annotate 2,460 single-gene perturbation signatures, 839 disease versus normal signatures, and 906 drug perturbation signatures. All these signatures are unique and are manually validated for quality. Global analysis of these signatures confirms known associations and identifies novel associations between genes, diseases and drugs. The manually curated signatures are used as a training set to develop classifiers for extracting similar signatures from the entire GEO repository. We develop a web portal to serve these signatures for query, download and visualization.

14.
Dev Biol ; 415(1): 157-167, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27178668

RESUMEN

Drosophila eye development is a complex process that involves many transcription factors (TFs) and interactions with their cofactors and targets. The TF Sine oculis (So) and its cofactor Eyes absent (Eya) are highly conserved and are both necessary and sufficient for eye development. Despite their many important roles during development, the direct targets of So are still largely unknown. Therefore the So-dependent regulatory network governing eye determination and differentiation is poorly understood. In this study, we intersected gene expression profiles of so or eya mutant eye tissue prepared from three different developmental stages and identified 1731 differentially expressed genes across the Drosophila genome. A combination of co-expression analyses and motif discovery identified a set of twelve putative direct So targets, including three known and nine novel targets. We also used our previous So ChIP-seq data to assess motif predictions for So and identified a canonical So binding motif. Finally, we performed in vivo enhancer reporter assays to test predicted enhancers from six candidate target genes and find that at least one enhancer from each gene is expressed in the developing eye disc and that their expression patterns overlap with that of So. We furthermore confirmed that the expression level of predicted direct So targets, for which antibodies are available, are reduced in so or eya post-mitotic knockout eye discs. In summary, we expand the set of putative So targets and show for the first time that the combined use of expression profiling of so with its cofactor eya is an effective method to identify novel So targets. Moreover, since So is highly conserved throughout the metazoa, our results provide the basis for future functional studies in a wide variety of organisms.


Asunto(s)
Ojo Compuesto de los Artrópodos/crecimiento & desarrollo , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Proteínas del Ojo/fisiología , Regulación del Desarrollo de la Expresión Génica , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Inmunoprecipitación de Cromatina , Ojo Compuesto de los Artrópodos/ultraestructura , Secuencia de Consenso , Drosophila melanogaster/crecimiento & desarrollo , Ontología de Genes , Estudios de Asociación Genética , Discos Imaginales/metabolismo , Larva , Pupa , ARN Mensajero/genética , Transcripción Genética , Transcriptoma
15.
BMC Bioinformatics ; 17(Suppl 15): 432, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-28185568

RESUMEN

BACKGROUND: In the study of complex diseases using genome-wide expression data from clinical samples, a difficult case is the identification and mapping of the gene signatures associated to the stages that occur in the progression of a disease. The stages usually correspond to different subtypes or classes of the disease, and the difficulty to identify them often comes from patient heterogeneity and sample variability that can hide the biomedical relevant changes that characterize each stage, making standard differential analysis inadequate or inefficient. RESULTS: We propose a methodology to study diseases or disease stages ordered in a sequential manner (e.g. from early stages with good prognosis to more acute or serious stages associated to poor prognosis). The methodology is applied to diseases that have been studied obtaining genome-wide expression profiling of cohorts of patients at different stages. The approach allows searching for consistent expression patterns along the progression of the disease through two major steps: (i) identifying genes with increasing or decreasing trends in the progression of the disease; (ii) clustering the increasing/decreasing gene expression patterns using an unsupervised approach to reveal whether there are consistent patterns and find genes altered at specific disease stages. The first step is carried out using Gamma rank correlation to identify genes whose expression correlates with a categorical variable that represents the stages of the disease. The second step is done using a Self Organizing Map (SOM) to cluster the genes according to their progressive profiles and identify specific patterns. Both steps are done after normalization of the genomic data to allow the integration of multiple independent datasets. In order to validate the results and evaluate their consistency and biological relevance, the methodology is applied to datasets of three different diseases: myelodysplastic syndrome, colorectal cancer and Alzheimer's disease. A software script written in R, named genediseasePatterns, is provided to allow the use and application of the methodology. CONCLUSION: The method presented allows the analysis of the progression of complex and heterogeneous diseases that can be divided in pathological stages. It identifies gene groups whose expression patterns change along the advance of the disease, and it can be applied to different types of genomic data studying cohorts of patients in different states.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Transcriptoma , Algoritmos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Análisis por Conglomerados , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Bases de Datos Genéticas , Progresión de la Enfermedad , Humanos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Estadificación de Neoplasias , Análisis de Secuencia de ARN , Índice de Severidad de la Enfermedad
16.
BMC Genomics ; 16 Suppl 5: S3, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26040557

RESUMEN

BACKGROUND: Despite the large increase of transcriptomic studies that look for gene signatures on diseases, there is still a need for integrative approaches that obtain separation of multiple pathological states providing robust selection of gene markers for each disease subtype and information about the possible links or relations between those genes. RESULTS: We present a network-oriented and data-driven bioinformatic approach that searches for association of genes and diseases based on the analysis of genome-wide expression data derived from microarrays or RNA-Seq studies. The approach aims to (i) identify gene sets associated to different pathological states analysed together; (ii) identify a minimum subset within these genes that unequivocally differentiates and classifies the compared disease subtypes; (iii) provide a measurement of the discriminant power of these genes and (iv) identify links between the genes that characterise each of the disease subtypes. This bioinformatic approach is implemented in an R package, named geNetClassifier, available as an open access tool in Bioconductor. To illustrate the performance of the tool, we applied it to two independent datasets: 250 samples from patients with four major leukemia subtypes analysed using expression arrays; another leukemia dataset analysed with RNA-Seq that includes a subtype also present in the previous set. The results show the selection of key deregulated genes recently reported in the literature and assigned to the leukemia subtypes studied. We also show, using these independent datasets, the selection of similar genes in a network built for the same disease subtype. CONCLUSIONS: The construction of gene networks related to specific disease subtypes that include parameters such as gene-to-gene association, gene disease specificity and gene discriminant power can be very useful to draw gene-disease maps and to unravel the molecular features that characterize specific pathological states. The application of the bioinformatic tool here presented shows a neat way to achieve such molecular characterization of the diseases using genome-wide expression data.


Asunto(s)
Biomarcadores de Tumor/genética , Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Marcadores Genéticos/genética , Leucemia/genética , Secuencia de Bases , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Predisposición Genética a la Enfermedad , Humanos , Leucemia/clasificación , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ARN
17.
Bioinformatics ; 31(10): 1686-8, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25600944

RESUMEN

Functional Gene Networks (FGNet) is an R/Bioconductor package that generates gene networks derived from the results of functional enrichment analysis (FEA) and annotation clustering. The sets of genes enriched with specific biological terms (obtained from a FEA platform) are transformed into a network by establishing links between genes based on common functional annotations and common clusters. The network provides a new view of FEA results revealing gene modules with similar functions and genes that are related to multiple functions. In addition to building the functional network, FGNet analyses the similarity between the groups of genes and provides a distance heatmap and a bipartite network of functionally overlapping genes. The application includes an interface to directly perform FEA queries using different external tools: DAVID, GeneTerm Linker, TopGO or GAGE; and a graphical interface to facilitate the use.


Asunto(s)
Redes Reguladoras de Genes , Programas Informáticos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Análisis por Conglomerados , Corteza Entorrinal/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Neuronas/metabolismo
18.
BMC Genomics ; 13 Suppl 5: S5, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23095915

RESUMEN

BACKGROUND: Analysis of DNA copy number alterations and gene expression changes in human samples have been used to find potential target genes in complex diseases. Recent studies have combined these two types of data using different strategies, but focusing on finding gene-based relationships. However, it has been proposed that these data can be used to identify key genomic regions, which may enclose causal genes under the assumption that disease-associated gene expression changes are caused by genomic alterations.


Asunto(s)
Algoritmos , Dosificación de Gen/genética , Genoma Humano/genética , Genómica/métodos , Glioblastoma/genética , Modelos Genéticos , Transcriptoma , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...