Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep Methods ; 2(11): 100340, 2022 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-36452860

RESUMEN

Tumor heterogeneity is a major challenge for oncology drug discovery and development. Understanding of the spatial tumor landscape is key to identifying new targets and impactful model systems. Here, we test the utility of spatial transcriptomics (ST) for oncology discovery by profiling 40 tissue sections and 80,024 capture spots across a diverse set of tissue types, sample formats, and RNA capture chemistries. We verify the accuracy and fidelity of ST by leveraging matched pathology analysis, which provides a ground truth for tissue section composition. We then use spatial data to demonstrate the capture of key tumor depth features, identifying hypoxia, necrosis, vasculature, and extracellular matrix variation. We also leverage spatial context to identify relative cell-type locations showing the anti-correlation of tumor and immune cells in syngeneic cancer models. Lastly, we demonstrate target identification approaches in clinical pancreatic adenocarcinoma samples, highlighting tumor intrinsic biomarkers and paracrine signaling.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Transcriptoma/genética , Neoplasias Pancreáticas/diagnóstico , Oncología Médica , Perfilación de la Expresión Génica , Biomarcadores de Tumor/genética
2.
Nat Commun ; 11(1): 6315, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33298926

RESUMEN

Despite the increasing interest in targeting stromal elements of the tumor microenvironment, we still face tremendous challenges in developing adequate therapeutics to modify the tumor stromal landscape. A major obstacle to this is our poor understanding of the phenotypic and functional heterogeneity of stromal cells in tumors. Herein, we perform an unbiased interrogation of tumor mesenchymal cells, delineating the co-existence of distinct subsets of cancer-associated fibroblasts (CAFs) in the microenvironment of murine carcinomas, each endowed with unique phenotypic features and functions. Furthermore, our study shows that neutralization of TGFß in vivo leads to remodeling of CAF dynamics, greatly reducing the frequency and activity of the myofibroblast subset, while promoting the formation of a fibroblast population characterized by strong response to interferon and heightened immunomodulatory properties. These changes correlate with the development of productive anti-tumor immunity and greater efficacy of PD1 immunotherapy. Along with providing the scientific rationale for the evaluation of TGFß and PD1 co-blockade in the clinical setting, this study also supports the concept of plasticity of the stromal cell landscape in tumors, laying the foundation for future investigations aimed at defining pathways and molecules to program CAF composition for cancer therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Fibroblastos Asociados al Cáncer/inmunología , Carcinoma/tratamiento farmacológico , Interferón beta/inmunología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Carcinoma/inmunología , Carcinoma/patología , Línea Celular Tumoral/trasplante , Plasticidad de la Célula/efectos de los fármacos , Plasticidad de la Célula/inmunología , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Ratones , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Células del Estroma/efectos de los fármacos , Células del Estroma/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
4.
Nat Med ; 25(1): 95-102, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30559422

RESUMEN

Interferons (IFNs) are cytokines that play a critical role in limiting infectious and malignant diseases 1-4 . Emerging data suggest that the strength and duration of IFN signaling can differentially impact cancer therapies, including immune checkpoint blockade 5-7 . Here, we characterize the output of IFN signaling, specifically IFN-stimulated gene (ISG) signatures, in primary tumors from The Cancer Genome Atlas. While immune infiltration correlates with the ISG signature in some primary tumors, the existence of ISG signature-positive tumors without evident infiltration of IFN-producing immune cells suggests that cancer cells per se can be a source of IFN production. Consistent with this hypothesis, analysis of patient-derived tumor xenografts propagated in immune-deficient mice shows evidence of ISG-positive tumors that correlates with expression of human type I and III IFNs derived from the cancer cells. Mechanistic studies using cell line models from the Cancer Cell Line Encyclopedia that harbor ISG signatures demonstrate that this is a by-product of a STING-dependent pathway resulting in chronic tumor-derived IFN production. This imposes a transcriptional state on the tumor, poising it to respond to the aberrant accumulation of double-stranded RNA (dsRNA) due to increased sensor levels (MDA5, RIG-I and PKR). By interrogating our functional short-hairpin RNA screen dataset across 398 cancer cell lines, we show that this ISG transcriptional state creates a novel genetic vulnerability. ISG signature-positive cancer cells are sensitive to the loss of ADAR, a dsRNA-editing enzyme that is also an ISG. A genome-wide CRISPR genetic suppressor screen reveals that the entire type I IFN pathway and the dsRNA-activated kinase, PKR, are required for the lethality induced by ADAR depletion. Therefore, tumor-derived IFN resulting in chronic signaling creates a cellular state primed to respond to dsRNA accumulation, rendering ISG-positive tumors susceptible to ADAR loss.


Asunto(s)
Adenosina Desaminasa/metabolismo , Interferones/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Proteínas de la Membrana/metabolismo , Ratones Desnudos , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Supresión Genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nat Commun ; 9(1): 4181, 2018 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-30327465

RESUMEN

Racial/ethnic disparities in breast cancer mortality continue to widen but genomic studies rarely interrogate breast cancer in diverse populations. Through genome, exome, and RNA sequencing, we examined the molecular features of breast cancers using 194 patients from Nigeria and 1037 patients from The Cancer Genome Atlas (TCGA). Relative to Black and White cohorts in TCGA, Nigerian HR + /HER2 - tumors are characterized by increased homologous recombination deficiency signature, pervasive TP53 mutations, and greater structural variation-indicating aggressive biology. GATA3 mutations are also more frequent in Nigerians regardless of subtype. Higher proportions of APOBEC-mediated substitutions strongly associate with PIK3CA and CDH1 mutations, which are underrepresented in Nigerians and Blacks. PLK2, KDM6A, and B2M are also identified as previously unreported significantly mutated genes in breast cancer. This dataset provides novel insights into potential molecular mechanisms underlying outcome disparities and lay a foundation for deployment of precision therapeutics in underserved populations.


Asunto(s)
Neoplasias de la Mama/genética , Recombinación Homóloga , Mutación , Desaminasas APOBEC/genética , Negro o Afroamericano/genética , Antígenos CD/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Cadherinas/genética , Fosfatidilinositol 3-Quinasa Clase I/genética , Exoma , Femenino , Humanos , Nigeria , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Proteína p53 Supresora de Tumor/genética , Población Blanca/genética , Secuenciación Completa del Genoma
6.
Cancer Immunol Res ; 6(12): 1472-1485, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30266714

RESUMEN

Cancer-associated fibroblasts (CAFs) are generally associated with poor clinical outcome. CAFs support tumor growth in a variety of ways and can suppress antitumor immunity and response to immunotherapy. However, a precise understanding of CAF contributions to tumor growth and therapeutic response is lacking. Discrepancies in this field of study may stem from heterogeneity in the composition and function of fibroblasts in the tumor microenvironment. Furthermore, it remains unclear whether CAFs directly interact with and suppress T cells. Here, mouse and human breast tumors were used to examine stromal cells expressing fibroblast activation protein (FAP), a surface marker for CAFs. Two discrete populations of FAP+ mesenchymal cells were identified on the basis of podoplanin (PDPN) expression: a FAP+PDPN+ population of CAFs and a FAP+PDPN- population of cancer-associated pericytes (CAPs). Although both subsets expressed extracellular matrix molecules, the CAF transcriptome was enriched in genes associated with TGFß signaling and fibrosis compared with CAPs. In addition, CAFs were enriched at the outer edge of the tumor, in close contact with T cells, whereas CAPs were localized around vessels. Finally, FAP+PDPN+ CAFs suppressed the proliferation of T cells in a nitric oxide-dependent manner, whereas FAP+PDPN- pericytes were not immunosuppressive. Collectively, these findings demonstrate that breast tumors contain multiple populations of FAP-expressing stromal cells of dichotomous function, phenotype, and location.


Asunto(s)
Neoplasias de la Mama/patología , Gelatinasas/metabolismo , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Células del Estroma/metabolismo , Microambiente Tumoral/inmunología , Animales , Neoplasias de la Mama/inmunología , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/patología , Proliferación Celular , Endopeptidasas , Femenino , Regulación de la Expresión Génica , Humanos , Glicoproteínas de Membrana/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Óxido Nítrico/metabolismo , Pericitos/metabolismo , Pericitos/patología , Células del Estroma/patología , Linfocitos T/patología
7.
Mol Cancer Res ; 15(12): 1722-1732, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28851814

RESUMEN

Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV) associated cancer characterized by a poor prognosis and a high level of lymphocyte infiltrate. Genetic hallmarks of NPC are not completely known but include deletion of the p16 (CDKN2A) locus and mutations in NF-κB pathway components, with a relatively low total mutational load. To better understand the genetic landscape, an integrated genomic analysis was performed using a large clinical cohort of treatment-naïve NPC tumor specimens. This genomic analysis was generally concordant with previous studies; however, three subtypes of NPC were identified by differences in immune cell gene expression, prognosis, tumor cell morphology, and genetic characteristics. A gene expression signature of proliferation was poorly prognostic and associated with either higher mutation load or specific EBV gene expression patterns in a subtype-specific manner. Finally, higher levels of stromal tumor-infiltrating lymphocytes associated with good prognosis and lower expression of a WNT and TGFß pathway activation signature.Implications: This study represents the first integrated analysis of mutation, copy number, and gene expression data in NPC and suggests how tumor genetics and EBV infection influence the tumor microenvironment in this disease. These insights should be considered for guiding immunotherapy treatment strategies in this disease. Mol Cancer Res; 15(12); 1722-32. ©2017 AACR.


Asunto(s)
Carcinoma/genética , Genoma Humano/genética , Neoplasias Nasofaríngeas/genética , Pronóstico , Microambiente Tumoral/genética , Adulto , Anciano , Carcinoma/patología , Carcinoma/virología , Proliferación Celular/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Genómica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidad , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Masculino , Persona de Mediana Edad , Mutación , FN-kappa B/genética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/virología , Factor de Crecimiento Transformador beta/genética , Vía de Señalización Wnt/genética
8.
Cell Rep ; 17(12): 3206-3218, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28009290

RESUMEN

Recent studies have elucidated the molecular mechanism of RORγT transcriptional regulation of Th17 differentiation and function. RORγT was initially identified as a transcription factor required for thymopoiesis by maintaining survival of CD4+CD8+ (DP) thymocytes. While RORγ antagonists are currently being developed to treat autoimmunity, it remains unclear how RORγT inhibition may impact thymocyte development. In this study, we show that in addition to regulating DP thymocytes survival, RORγT also controls genes that regulate thymocyte migration, proliferation, and T cell receptor (TCR)α selection. Strikingly, pharmacological inhibition of RORγ skews TCRα gene rearrangement, limits T cell repertoire diversity, and inhibits development of autoimmune encephalomyelitis. Thus, targeting RORγT not only inhibits Th17 cell development and function but also fundamentally alters thymic-emigrant recognition of self and foreign antigens. The analysis of RORγ inhibitors has allowed us to gain a broader perspective of the diverse function of RORγT and its impact on T cell biology.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timocitos/inmunología , Animales , Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/genética , Encefalomielitis Autoinmune Experimental/genética , Encefalomielitis Autoinmune Experimental/terapia , Regulación de la Expresión Génica/inmunología , Reordenamiento Génico/genética , Humanos , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Células Th17/efectos de los fármacos , Células Th17/inmunología
9.
PLoS One ; 10(2): e0118286, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25706956

RESUMEN

OBJECTIVES: Human airway epithelial cells are the principal target of human rhinovirus (HRV), a common cold pathogen that triggers the majority of asthma exacerbations. The objectives of this study were 1) to evaluate an in vitro air liquid interface cultured human airway epithelial cell model for HRV infection, and 2) to identify gene expression patterns associated with asthma intrinsically and/or after HRV infection using this model. METHODS: Air-liquid interface (ALI) human airway epithelial cell cultures were prepared from 6 asthmatic and 6 non-asthmatic donors. The effects of rhinovirus RV-A16 on ALI cultures were compared. Genome-wide gene expression changes in ALI cultures following HRV infection at 24 hours post exposure were further analyzed using RNA-seq technology. Cellular gene expression and cytokine/chemokine secretion were further evaluated by qPCR and a Luminex-based protein assay, respectively. MAIN RESULTS: ALI cultures were readily infected by HRV. RNA-seq analysis of HRV infected ALI cultures identified sets of genes associated with asthma specific viral responses. These genes are related to inflammatory pathways, epithelial structure and remodeling and cilium assembly and function, including those described previously (e.g. CCL5, CXCL10 and CX3CL1, MUC5AC, CDHR3), and novel ones that were identified for the first time in this study (e.g. CCRL1). CONCLUSIONS: ALI-cultured human airway epithelial cells challenged with HRV are a useful translational model for the study of HRV-induced responses in airway epithelial cells, given that gene expression profile using this model largely recapitulates some important patterns of gene responses in patients during clinical HRV infection. Furthermore, our data emphasize that both abnormal airway epithelial structure and inflammatory signaling are two important asthma signatures, which can be further exacerbated by HRV infection.


Asunto(s)
Asma/genética , Asma/virología , Diferenciación Celular/genética , Células Epiteliales/virología , Infecciones por Picornaviridae/genética , Sistema Respiratorio/virología , Adolescente , Adulto , Células Cultivadas , Quimiocinas/genética , Niño , Femenino , Expresión Génica/genética , Humanos , Inflamación/genética , Inflamación/virología , Masculino , Persona de Mediana Edad , Infecciones por Picornaviridae/virología , Rhinovirus , Transducción de Señal/genética
10.
PLoS One ; 9(12): e113937, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25504080

RESUMEN

UNLABELLED: Approximately 30% of rheumatoid arthritis patients achieve inadequate response to anti-TNF biologics. Attempts to identify molecular biomarkers predicting response have met with mixed success. This may be attributable, in part, to the variable and subjective disease assessment endpoints with large placebo effects typically used to classify patient response. Sixty-one patients with active RA despite methotrexate treatment, and with MRI-documented synovitis, were randomized to receive infliximab or placebo. Blood was collected at baseline and genome-wide transcription in whole blood was measured using microarrays. The primary endpoint in this study was determined by measuring the transfer rate constant (Ktrans) of a gadolinium-based contrast agent from plasma to synovium using MRI. Secondary endpoints included repeated clinical assessments with DAS28(CRP), and assessments of osteitis and synovitis by the RAMRIS method. Infliximab showed greater decrease from baseline in DCE-MRI Ktrans of wrist and MCP at all visits compared with placebo (P<0.001). Statistical analysis was performed to identify genes associated with treatment-specific 14-week change in Ktrans. The 256 genes identified were used to derive a gene signature score by averaging their log expression within each patient. The resulting score correlated with improvement of Ktrans in infliximab-treated patients and with deterioration of Ktrans in placebo-treated subjects. Poor responders showed high expression of activated B-cell genes whereas good responders exhibited a gene expression pattern consistent with mobilization of neutrophils and monocytes and high levels of reticulated platelets. This gene signature was significantly associated with clinical response in two previously published whole blood gene expression studies using anti-TNF therapies. These data provide support for the hypothesis that anti-TNF inadequate responders comprise a distinct molecular subtype of RA characterized by differences in pre-treatment blood mRNA expression. They also highlight the importance of placebo controls and robust, objective endpoints in biomarker discovery. TRIAL REGISTRATION: ClinicalTrials.gov NCT01313520.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antirreumáticos/administración & dosificación , Artritis Reumatoide/sangre , Artritis Reumatoide/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Muñeca/patología , Adulto , Anticuerpos Monoclonales/farmacología , Antirreumáticos/farmacología , Artritis Reumatoide/patología , Femenino , Perfilación de la Expresión Génica , Humanos , Infliximab , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Análisis de Secuencia de ARN , Resultado del Tratamiento
11.
Drug Discov Today ; 19(2): 133-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24126144

RESUMEN

The high failure rate of new therapeutic mechanisms tested in clinical development has spurred an upsurge in research dedicated to discovering biomarker readouts that can improve decision-making. Increasingly, systems biology and genomic technologies, such as transcriptional profiling, are being leveraged to aid in the discovery of biomarker readouts. For inflammatory and immunological diseases, such as rheumatoid arthritis (RA) and asthma, progress has been made in developing biomarkers to monitor disease activity, prediction of response to therapy, and pharmacodynamic (PD) measurements. In this review, we discuss recent successes and challenges in these endeavors, highlighting the importance of human clinical studies of standard-of-care treatments in control subjects and patients with disease as the most direct path toward identifying useful translational biomarkers for clinical development.


Asunto(s)
Genómica/métodos , Enfermedades del Sistema Inmune/tratamiento farmacológico , Biología de Sistemas/métodos , Animales , Biomarcadores/metabolismo , Diseño de Fármacos , Humanos , Enfermedades del Sistema Inmune/fisiopatología , Inflamación/tratamiento farmacológico , Inflamación/fisiopatología , Investigación Biomédica Traslacional/métodos
12.
Genome Res ; 22(6): 1173-83, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22454233

RESUMEN

We developed PolyA-seq, a strand-specific and quantitative method for high-throughput sequencing of 3' ends of polyadenylated transcripts, and used it to globally map polyadenylation (polyA) sites in 24 matched tissues in human, rhesus, dog, mouse, and rat. We show that PolyA-seq is as accurate as existing RNA sequencing (RNA-seq) approaches for digital gene expression (DGE), enabling simultaneous mapping of polyA sites and quantitative measurement of their usage. In human, we confirmed 158,533 known sites and discovered 280,857 novel sites (FDR < 2.5%). On average 10% of novel human sites were also detected in matched tissues in other species. Most novel sites represent uncharacterized alternative polyA events and extensions of known transcripts in human and mouse, but primarily delineate novel transcripts in the other three species. A total of 69.1% of known human genes that we detected have multiple polyA sites in their 3'UTRs, with 49.3% having three or more. We also detected polyadenylation of noncoding and antisense transcripts, including constitutive and tissue-specific primary microRNAs. The canonical polyA signal was strongly enriched and positionally conserved in all species. In general, usage of polyA sites is more similar within the same tissues across different species than within a species. These quantitative maps of polyA usage in evolutionarily and functionally related samples constitute a resource for understanding the regulatory mechanisms underlying alternative polyadenylation.


Asunto(s)
Mamíferos/genética , Poli A/genética , Poliadenilación/genética , Regiones no Traducidas 3' , Animales , Embrión de Pollo , Perros , Evolución Molecular , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Macaca mulatta/genética , Ratones , MicroARNs/genética , Modelos Genéticos , ARN no Traducido , Ratas , Transcriptoma
13.
Methods Mol Biol ; 674: 179-93, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20827592

RESUMEN

Chromatin immunoprecipitation (ChIP) experiments allow the location of transcription factors to be determined across the genome. Subsequent analysis of the sequences of the identified regions allows binding to be localized at a higher resolution than can be achieved by current high-throughput experiments without sequence analysis and may provide important insight into the regulatory programs enacted by the protein of interest. In this chapter we review the tools, workflow, and common pitfalls of such analyses and recommend strategies for effective motif discovery from these data.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Análisis de Secuencia de ADN/métodos , Factores de Transcripción/metabolismo , Sitios de Unión , Mapeo Cromosómico , ADN/genética , ADN/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Especificidad por Sustrato
14.
PLoS Comput Biol ; 6(4): e1000773, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20442865

RESUMEN

Understanding the mechanistic basis of transcriptional regulation has been a central focus of molecular biology since its inception. New high-throughput chromatin immunoprecipitation experiments have revealed that most regulatory proteins bind thousands of sites in mammalian genomes. However, the functional significance of these binding sites remains unclear. We present a quantitative model of transcriptional regulation that suggests the contribution of each binding site to tissue-specific gene expression depends strongly on its position relative to the transcription start site. For three cell types, we show that, by considering binding position, it is possible to predict relative expression levels between cell types with an accuracy approaching the level of agreement between different experimental platforms. Our model suggests that, for the transcription factors profiled in these cell types, a regulatory site's influence on expression falls off almost linearly with distance from the transcription start site in a 10 kilobase range. Binding to both evolutionarily conserved and non-conserved sequences contributes significantly to transcriptional regulation. Our approach also reveals the quantitative, tissue-specific role of individual proteins in activating or repressing transcription. These results suggest that regulator binding position plays a previously unappreciated role in influencing expression and blurs the classical distinction between proximal promoter and distal binding events.


Asunto(s)
Regulación de la Expresión Génica , Modelos Genéticos , Células 3T3-L1 , Algoritmos , Animales , Sitios de Unión , Proteína de Unión a CREB/metabolismo , Cerebelo/metabolismo , Secuencia Conservada , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Elementos Reguladores de la Transcripción , Transcripción Genética
15.
Development ; 137(7): 1189-203, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20215353

RESUMEN

The Wilms' tumor suppressor 1 (WT1) gene encodes a DNA- and RNA-binding protein that plays an essential role in nephron progenitor differentiation during renal development. To identify WT1 target genes that might regulate nephron progenitor differentiation in vivo, we performed chromatin immunoprecipitation (ChIP) coupled to mouse promoter microarray (ChIP-chip) using chromatin prepared from embryonic mouse kidney tissue. We identified 1663 genes bound by WT1, 86% of which contain a previously identified, conserved, high-affinity WT1 binding site. To investigate functional interactions between WT1 and candidate target genes in nephron progenitors, we used a novel, modified WT1 morpholino loss-of-function model in embryonic mouse kidney explants to knock down WT1 expression in nephron progenitors ex vivo. Low doses of WT1 morpholino resulted in reduced WT1 target gene expression specifically in nephron progenitors, whereas high doses of WT1 morpholino arrested kidney explant development and were associated with increased nephron progenitor cell apoptosis, reminiscent of the phenotype observed in Wt1(-/-) embryos. Collectively, our results provide a comprehensive description of endogenous WT1 target genes in nephron progenitor cells in vivo, as well as insights into the transcriptional signaling networks controlled by WT1 that might direct nephron progenitor fate during renal development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Riñón/citología , Riñón/embriología , Nefronas/citología , Células Madre/fisiología , Proteínas WT1/metabolismo , Animales , Apoptosis/fisiología , Secuencia de Bases , Inmunoprecipitación de Cromatina , Bases de Datos Factuales , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Femenino , Hibridación in Situ , Riñón/metabolismo , Ratones , Análisis por Micromatrices , Nefronas/embriología , Nefronas/metabolismo , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Embarazo , Células Madre/citología , Técnicas de Cultivo de Tejidos , Proteínas WT1/genética
16.
Nat Genet ; 39(6): 730-2, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17529977

RESUMEN

We demonstrate that the binding sites for highly conserved transcription factors vary extensively between human and mouse. We mapped the binding of four tissue-specific transcription factors (FOXA2, HNF1A, HNF4A and HNF6) to 4,000 orthologous gene pairs in hepatocytes purified from human and mouse livers. Despite the conserved function of these factors, from 41% to 89% of their binding events seem to be species specific. When the same protein binds the promoters of orthologous genes, approximately two-thirds of the binding sites do not align.


Asunto(s)
Secuencia Conservada/genética , Regulación de la Expresión Génica , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/genética , Transcripción Genética , Animales , Variación Genética , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 6 del Hepatocito/genética , Humanos , Ratones , Homología de Secuencia
17.
Nature ; 445(7130): 931-5, 2007 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-17237765

RESUMEN

Foxp3+CD4+CD25+ regulatory T (T(reg)) cells are essential for the prevention of autoimmunity. T(reg) cells have an attenuated cytokine response to T-cell receptor stimulation, and can suppress the proliferation and effector function of neighbouring T cells. The forkhead transcription factor Foxp3 (forkhead box P3) is selectively expressed in T(reg) cells, is required for T(reg) development and function, and is sufficient to induce a T(reg) phenotype in conventional CD4+CD25- T cells. Mutations in Foxp3 cause severe, multi-organ autoimmunity in both human and mouse. FOXP3 can cooperate in a DNA-binding complex with NFAT (nuclear factor of activated T cells) to regulate the transcription of several known target genes. However, the global set of genes regulated directly by Foxp3 is not known and consequently, how this transcription factor controls the gene expression programme for T(reg) function is not understood. Here we identify Foxp3 target genes and report that many of these are key modulators of T-cell activation and function. Remarkably, the predominant, although not exclusive, effect of Foxp3 occupancy is to suppress the activation of target genes on T-cell stimulation. Foxp3 suppression of its targets appears to be crucial for the normal function of T(reg) cells, because overactive variants of some target genes are known to be associated with autoimmune disease.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/genética , Activación de Linfocitos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Línea Celular , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Ratones , Modelos Inmunológicos , Fenotipo , Linfocitos T Reguladores/citología , Transcripción Genética/genética
18.
Nat Biotechnol ; 24(8): 963-70, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16900145

RESUMEN

Direct physical information that describes where transcription factors, nucleosomes, modified histones, RNA polymerase II and other key proteins interact with the genome provides an invaluable mechanistic foundation for understanding complex programs of gene regulation. We present a method, joint binding deconvolution (JBD), which uses additional easily obtainable experimental data about chromatin immunoprecipitation (ChIP) to improve the spatial resolution of the transcription factor binding locations inferred from ChIP followed by DNA microarray hybridization (ChIP-Chip) data. Based on this probabilistic model of binding data, we further pursue improved spatial resolution by using sequence information. We produce positional priors that link ChIP-Chip data to sequence data by guiding motif discovery to inferred protein-DNA binding sites. We present results on the yeast transcription factors Gcn4 and Mig2 to demonstrate JBD's spatial resolution capabilities and show that positional priors allow computational discovery of the Mig2 motif when a standard approach fails.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Proteínas de Unión al ADN/química , ADN/química , Modelos Químicos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Análisis de Secuencia de ADN/métodos , Factores de Transcripción/química , Secuencia de Bases , Simulación por Computador , Modelos Genéticos , Modelos Moleculares , Datos de Secuencia Molecular
20.
BMC Bioinformatics ; 7: 113, 2006 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-16522208

RESUMEN

BACKGROUND: The regulatory map of a genome consists of the binding sites for proteins that determine the transcription of nearby genes. An initial regulatory map for S. cerevisiae was recently published using six motif discovery programs to analyze genome-wide chromatin immunoprecipitation data for 203 transcription factors. The programs were used to identify sequence motifs that were likely to correspond to the DNA-binding specificity of the immunoprecipitated proteins. We report improved versions of two conservation-based motif discovery algorithms, PhyloCon and Converge. Using these programs, we create a refined regulatory map for S. cerevisiae by reanalyzing the same chromatin immunoprecipitation data. RESULTS: Applying the same conservative criteria that were applied in the original study, we find that PhyloCon and Converge each separately discover more known specificities than the combination of all six programs in the previous study. Combining the results of PhyloCon and Converge, we discover significant sequence motifs for 36 transcription factors that were previously missed. The new set of motifs identifies 636 more regulatory interactions than the previous one. The new network contains 28% more regulatory interactions among transcription factors, evidence of greater cross-talk between regulators. CONCLUSION: Combining two complementary computational strategies for conservation-based motif discovery improves the ability to identify the specificity of transcriptional regulators from genome-wide chromatin immunoprecipitation data. The increased sensitivity of these methods significantly expands the map of yeast regulatory sites without the need to alter any of the thresholds for statistical significance. The new map of regulatory sites reveals a more elaborate and complex view of the yeast genetic regulatory network than was observed previously.


Asunto(s)
Algoritmos , Mapeo Cromosómico/métodos , Regulación Fúngica de la Expresión Génica/genética , Elementos Reguladores de la Transcripción/genética , Saccharomyces cerevisiae/genética , Análisis de Secuencia de ADN/métodos , Factores de Transcripción/genética , Secuencia Conservada/genética , Filogenia , Activación Transcripcional/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA