Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Bioinformatics ; 32(17): i421-i429, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27587658

RESUMEN

MOTIVATION: A central task of bioinformatics is to develop sensitive and specific means of providing medical prognoses from biomarker patterns. Common methods to predict phenotypes in RNA-Seq datasets utilize machine learning algorithms trained via gene expression. Isoforms, however, generated from alternative splicing, may provide a novel and complementary set of transcripts for phenotype prediction. In contrast to gene expression, the number of isoforms increases significantly due to numerous alternative splicing patterns, resulting in a prioritization problem for many machine learning algorithms. This study identifies the empirically optimal methods of transcript quantification, feature engineering and filtering steps using phenotype prediction accuracy as a metric. At the same time, the complementary nature of gene and isoform data is analyzed and the feasibility of identifying isoforms as biomarker candidates is examined. RESULTS: Isoform features are complementary to gene features, providing non-redundant information and enhanced predictive power when prioritized and filtered. A univariate filtering algorithm, which selects up to the N highest ranking features for phenotype prediction is described and evaluated in this study. An empirical comparison of pipelines for isoform quantification is reported by performing cross-validation prediction tests with datasets from human non-small cell lung cancer (NSCLC) patients, human patients with chronic obstructive pulmonary disease (COPD) and amyotrophic lateral sclerosis (ALS) transgenic mice, each including samples of diseased and non-diseased phenotypes. AVAILABILITY AND IMPLEMENTATION: https://github.com/clabuzze/Phenotype-Prediction-Pipeline.git CONTACT: clabuzze@iastate.edu, antoniom@bc.edu, watsondk@musc.edu, andersonpe2@cofc.edu.


Asunto(s)
Algoritmos , Empalme Alternativo , Aprendizaje Automático , Fenotipo , Esclerosis Amiotrófica Lateral , Animales , Carcinoma de Pulmón de Células no Pequeñas , Humanos , Neoplasias Pulmonares , Ratones Transgénicos , Enfermedad Pulmonar Obstructiva Crónica
2.
EMBO Rep ; 16(5): 654-69, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25802403

RESUMEN

The ETS factor ETV2 (aka ER71) is essential for the generation of the blood and vascular system, as ETV2 deficiency leads to a complete block in blood and endothelial cell formation and embryonic lethality in the mouse. However, the ETV2-mediated gene regulatory network and signaling governing hematopoietic and endothelial cell development are poorly understood. Here, we map ETV2 global binding sites and carry out in vitro differentiation of embryonic stem cells, and germ line and conditional knockout mouse studies to uncover mechanisms involved in the hemangiogenic fate commitment from mesoderm. We show that ETV2 binds to enhancers that specify hematopoietic and endothelial cell lineages. We find that the hemangiogenic progenitor population in the developing embryo can be identified as FLK1(high)PDGFRα(-). Notably, these hemangiogenic progenitors are exclusively sensitive to ETV2-dependent FLK1 signaling. Importantly, ETV2 turns on other Ets genes, thereby establishing an ETS hierarchy. Consequently, the hematopoietic and endothelial cell program initiated by ETV2 is maintained partly by other ETS factors through an ETS switching mechanism. These findings highlight the critical role that transient ETV2 expression plays in the regulation of hematopoietic and endothelial cell lineage specification and stability.


Asunto(s)
Células Sanguíneas/citología , Células Sanguíneas/metabolismo , Diferenciación Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Factores de Transcripción/metabolismo , Animales , Sitios de Unión , Diferenciación Celular/genética , Linaje de la Célula/genética , Inmunoprecipitación de Cromatina , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunofenotipificación , Masculino , Ratones , Ratones Noqueados , Motivos de Nucleótidos , Especificidad de Órganos/genética , Posición Específica de Matrices de Puntuación , Unión Proteica , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
3.
BMC Genomics ; 17: 263, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27029813

RESUMEN

BACKGROUND: It has recently been shown that significant and accurate single nucleotide variants (SNVs) can be reliably called from RNA-Seq data. These may provide another source of features for multivariate predictive modeling of disease phenotype for the prioritization of candidate biomarkers. The continuous nature of SNV allele fraction features allows the concurrent investigation of several genomic phenomena, including allele specific expression, clonal expansion and/or deletion, and copy number variation. RESULTS: The proposed software pipeline and package, SNV Discriminant Analysis (SNV-DA), was applied on two RNA-Seq datasets with varying sample sizes sequenced at different depths: a dataset containing primary tumors from twenty patients with different disease outcomes in lung adenocarcinoma and a larger dataset of primary tumors representing two major breast cancer subtypes, estrogen receptor positive and triple negative. Predictive models were generated using the machine learning algorithm, sparse projections to latent structures discriminant analysis. Training sets composed of RNA-Seq SNV features limited to genomic regions of origin (e.g. exonic or intronic) and/or RNA-editing sites were shown to produce models with accurate predictive performances, were discriminant towards true label groupings, and were able to produce SNV rankings significantly different from than univariate tests. Furthermore, the utility of the proposed methodology is supported by its comparable performance to traditional models as well as the enrichment of selected SNVs located in genes previously associated with cancer and genes showing allele-specific expression. As proof of concept, we highlight the discovery of a previously unannotated intergenic locus that is associated with epigenetic regulatory marks in cancer and whose significant allele-specific expression is correlated with ER+ status; hereafter named ER+ associated hotspot (ERPAHS). CONCLUSION: The use of models from RNA-Seq SNVs to identify and prioritize candidate molecular targets for biomarker discovery is supported by the ability of the proposed method to produce significantly accurate predictive models that are discriminant towards true label groupings. Importantly, the proposed methodology allows investigation of mutations outside of exonic regions and identification of interesting expressed loci not included in traditional gene annotations. An implementation of the proposed methodology is provided that allows the user to specify SNV filtering criteria and cross-validation design during model creation and evaluation.


Asunto(s)
Modelos Genéticos , Polimorfismo de Nucleótido Simple , Análisis de Secuencia de ARN , Programas Informáticos , Regiones no Traducidas 3' , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Algoritmos , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , ADN Intergénico/genética , Análisis Discriminante , Exones , Femenino , Humanos , Intrones , Neoplasias Pulmonares/genética , Edición de ARN
4.
Circ Res ; 114(11): 1690-9, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24727028

RESUMEN

RATIONALE: Cardiovascular health depends on proper development and integrity of blood vessels. Ets variant 2 (Etv2), a member of the E26 transforming-specific family of transcription factors, is essential to initiate a transcriptional program leading to vascular morphogenesis in early mouse embryos. However, endothelial expression of the Etv2 gene ceases at midgestation; therefore, vascular development past this stage must continue independent of Etv2. OBJECTIVE: To identify molecular mechanisms underlying transcriptional regulation of vascular morphogenesis and homeostasis in the absence of Etv2. METHODS AND RESULTS: Using loss- and gain-of-function strategies and a series of molecular techniques, we identify Friend leukemia integration 1 (Fli1), another E26 transforming-specific family transcription factor, as a downstream target of Etv2. We demonstrate that Etv2 binds to conserved Ets-binding sites within the promoter region of the Fli1 gene and governs Fli1 expression. Importantly, in the absence of Etv2 at midgestation, binding of Etv2 at Ets-binding sites in the Fli1 promoter is replaced by Fli1 protein itself, sustaining expression of Fli1 as well as selective Etv2-regulated endothelial genes to promote endothelial cell survival and vascular integrity. Consistent with this, we report that Fli1 binds to the conserved Ets-binding sites within promoter and enhancer regions of other Etv2-regulated endothelial genes, including Tie2, to control their expression at and beyond midgestation. CONCLUSIONS: We have identified a novel positive feed-forward regulatory loop in which Etv2 activates expression of genes involved in vasculogenesis, including Fli1. Once the program is activated in early embryos, Fli1 then takes over to sustain the process in the absence of Etv2.


Asunto(s)
Endotelio Vascular/citología , Homeostasis/fisiología , Neovascularización Fisiológica/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , Factores de Transcripción/fisiología , Animales , Supervivencia Celular/fisiología , Desarrollo Embrionario/fisiología , Endotelio Vascular/fisiología , Femenino , Hemorragia/etiología , Hemorragia/fisiopatología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Morfogénesis/fisiología , Proteína Proto-Oncogénica c-fli-1/deficiencia , Proteína Proto-Oncogénica c-fli-1/genética
5.
Mol Carcinog ; 53 Suppl 1: E130-9, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23996472

RESUMEN

Recently, a reciprocal relationship between calcitriol and epithelial-to-mesenchymal transition has been described. Therefore, we hypothesized that calcitriol (1α,25-dihydroxyvitamin D3) would enhance radiation sensitivity in colorectal cancer regulated by epithelial mesenchymal transition. Vitamin-D receptor, E-cadherin and vimentin protein as well as E-cadherin, Snail and Slug mRNA levels were assessed in a panel of human colorectal cancer cell lines at baseline and in response calcitriol. We defined cell lines as calcitriol sensitive based on demonstrating an enhanced epithelial phenotype with increased E-cadherin, reduced vimentin and decreased expression of Snail and Slug as well as decreased cellular migration in response to calcitriol. In calcitriol sensitive cells, including DLD-1 and HCT116, 24 h calcitriol pre-treatment enhanced the radiation sensitivity by 2.3- and 2.6-fold, respectively, at 4 Gy (P < 0.05). In contrast, SW620 cells with high baseline mesenchymal features including high Slug and vimentin expression with low E-cadherin expression demonstrated no significant radiation sensitizing response to calcitriol treatment. Similarly, transfection of Slug in the calcitriol sensitive colon cancer cell lines, DLD-1 and HCT 116, completely inhibited the radiation sensitizing effect of calcitriol. Collectively, we demonstrate that calcitriol can enhance the therapeutic effects of radiation in colon cancer cells and Slug expression mitigates this observed effect potentially representing an effective biomarker for calcitriol therapy.


Asunto(s)
Calcitriol/farmacología , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Factores de Transcripción/genética , Western Blotting , Cadherinas/genética , Cadherinas/metabolismo , Agonistas de los Canales de Calcio/farmacología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/radioterapia , Transición Epitelial-Mesenquimal/efectos de la radiación , Técnica del Anticuerpo Fluorescente , Rayos gamma , Humanos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Vimentina/genética , Vimentina/metabolismo
6.
J Virol ; 87(1): 621-35, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23097457

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS), and KSHV activation of mitogen-activated protein kinases (MAPKs) initiates a number of key pathogenic determinants of KS. Direct inhibition of signal transduction as a therapeutic approach presents several challenges, and a better understanding of KSHV-induced mechanisms regulating MAPK activation may facilitate the development of new treatment or prevention strategies for KS. MAPK phosphatases, including dual-specificity phosphatase-1 (DUSP1), negatively regulate signal transduction and cytokine activation through MAPK dephosphorylation or interference with effector molecule binding to MAPKs, including the extracellular signal-regulated kinase (ERK). We found that ERK-dependent latent viral gene expression, the induction of promigratory factors, and cell invasiveness following de novo infection of primary human endothelial cells are in part dependent on KSHV suppression of DUSP1 expression during de novo infection. KSHV-encoded miR-K12-11 upregulates the expression of xCT (an amino acid transporter and KSHV fusion/entry receptor), and existing data indicate a role for xCT in the regulation of 14-3-3ß, a transcriptional repressor of DUSP1. We found that miR-K12-11 induces endothelial cell secretion of promigratory factors and cell invasiveness through upregulation of xCT-dependent, 14-3-3ß-mediated suppression of DUSP1. Finally, proof-of-principle experiments revealed that pharmacologic upregulation of DUSP1 inhibits the induction of promigratory factors and cell invasiveness during de novo KSHV infection. These data reveal an indirect role for miR-K12-11 in the regulation of DUSP1 and downstream pathogenesis.


Asunto(s)
Fosfatasa 1 de Especificidad Dual/antagonistas & inhibidores , Herpesvirus Humano 8/patogenicidad , Interacciones Huésped-Patógeno , MicroARNs/metabolismo , Línea Celular , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , ARN Viral/metabolismo
7.
Immunology ; 139(3): 318-27, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23320737

RESUMEN

Fli-1 belongs to the Ets transcription factor family and is expressed in haematopoietic cells, including most of the cells that are active in immunity. The mononuclear phagocytes, i.e. monocytes, macrophages and dendritic cells, originate in haematopoietic stem cells and play an important role in immunity. To assess the role of Fli-1 in mononuclear phagocyte development in vivo, we generated mice that express a truncated Fli-1 protein, lacking the C-terminal transcriptional activation domain (Fli-1(Δ) (CTA) ). Fli-1(Δ) (CTA) (/Δ) (CTA) mice had significantly increased populations of haematopoietic stem cells and common dendritic cell precursors in bone marrow compared with wild-type littermates. Significantly increased classical dendritic cells, plasmacytoid dendritic cells, and macrophage populations were found in spleens from Fli-1(∆) (CTA) (/∆) (CTA) mice compared with wild-type littermates. Fli-1(Δ) (CTA) (/Δ) (CTA) mice also had increased pre-classical dendritic cell and monocyte populations in peripheral blood mononuclear cells. Furthermore, bone marrow reconstitution studies demonstrated that expression of Fli-1 in both haematopoietic cells and stromal cells affected mononuclear phagocyte development in mice. Expression of Fms-like tyrosine kinase 3 ligand (Flt3L), a haematopoietic growth factor, in multipotent progenitors was statistically significantly increased from Fli-1(∆) (CTA) (/∆) (CTA) mice compared with wild-type littermates. Fli-1 protein binds directly to the promoter region of the Flt3L gene. Hence, Fli-1 plays an important role in the mononuclear phagocyte development, and the C-terminal transcriptional activation domain of Fli-1 negatively modulates mononuclear phagocyte development.


Asunto(s)
Diferenciación Celular , Células Dendríticas/citología , Macrófagos/citología , Monocitos/citología , Proteína Proto-Oncogénica c-fli-1/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Animales , Médula Ósea/inmunología , Línea Celular , Células Cultivadas , Células Dendríticas/inmunología , Femenino , Macrófagos/inmunología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Proteína Proto-Oncogénica c-fli-1/química , Proteína Proto-Oncogénica c-fli-1/genética , Bazo/inmunología
8.
Blood ; 118(20): 5604-12, 2011 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-21917756

RESUMEN

Friend leukemia virus integration 1 (FLI1), an Ets transcription factor family member, is linked to acute myelogenous leukemia (AML) by chromosomal events at the FLI1 locus, but the biologic impact of FLI1 expression on AML is unknown. FLI1 protein expression was measured in 511 newly diagnosed AML patients. Expression was similar in peripheral blood (PB) and BM and higher at diagnosis than at relapse (P = .02). Compared with normal CD34(+) cells, expression in AML was above or below normal in 32% and 5% of patients, respectively. Levels were negatively correlated with an antecedent hematologic disorder (P = .002) but not with age or cytogenetics. Mutated NPM1 (P = .0007) or FLT3-ITD (P < .02) had higher expression. FLI1 levels were negatively correlated with 10 of 195 proteins associated with proliferation and stromal interaction, and positively correlated (R > 0.3) with 19 others. The FLI1 level was not predictive of remission attainment, but patients with low or high FLI1 expression had shorter remission duration (22.6 and 40.3 vs 51.1 weeks, respectively; P = .01) and overall survival (45.2 and 35.4 vs 59.4 weeks, respectively; P = .03). High FLI1 levels were adverse in univariate and multivariate analysis. FLI1 expression is frequently abnormal and prognostically adverse in AML. FLI1 and/or its response genes may be therapeutically targetable to interfere with AML cell biology.


Asunto(s)
Regulación Leucémica de la Expresión Génica/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Proteína Proto-Oncogénica c-fli-1/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Nucleofosmina , Pronóstico , Proteómica , Factores de Riesgo , Adulto Joven , Tirosina Quinasa 3 Similar a fms/genética
9.
Clin Immunol ; 145(3): 201-8, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23108091

RESUMEN

Expression of transcription factor Fli-1 is implicated in the development of glomerulonephritis. Fli-1 heterozygous knockout (Fli1(+/-)) NZM2410 mice, a murine model of lupus, had significantly improved survival and reduced glomerulonephritis. In this study, we found that infiltrated inflammatory cells were significantly decreased in the kidneys from Fli-1(+/-) NZM2410 mice. The expression of monocyte chemoattractant protein-1 (MCP-1) was significantly decreased in kidneys from Fli-1(+/-) NZM2410 mice. The primary endothelial cells isolated from the kidneys of Fli-1(+/-) NZM2410 mice produced significantly less MCP-1. In endothelial cells transfected with specific Fli-1 siRNA the production of MCP-1 was significantly reduced compared to cells transfected with negative control siRNA. By Chromatin Immunoprecipitation (ChIP) assay, we further demonstrated that Fli-1 directly binds to the promoter of the MCP-1 gene. Our data indicate that Fli-1 impacts glomerulonephritis development by regulating expression of inflammatory chemokine MCP-1 and inflammatory cell infiltration in the kidneys in the NZM2410 mice.


Asunto(s)
Quimiocina CCL2/metabolismo , Glomerulonefritis/etiología , Glomerulonefritis/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Animales , Secuencia de Bases , Sitios de Unión/genética , Quimiocina CCL2/genética , ADN/genética , ADN/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Expresión Génica , Glomerulonefritis/genética , Glomerulonefritis/patología , Heterocigoto , Humanos , Riñón/metabolismo , Riñón/patología , Nefritis Lúpica/etiología , Nefritis Lúpica/genética , Nefritis Lúpica/metabolismo , Nefritis Lúpica/patología , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Proteína Proto-Oncogénica c-fli-1/antagonistas & inhibidores , Proteína Proto-Oncogénica c-fli-1/deficiencia , Proteína Proto-Oncogénica c-fli-1/genética , ARN Interferente Pequeño/genética
10.
J Surg Res ; 176(2): 359-66, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22221605

RESUMEN

BACKGROUND: The development of novel therapeutics for pancreatic cancer has been hindered by a lack of relevant preclinical models. The purpose of this study was to evaluate the clinical relevancy of two pancreatic cancer models using standard-of-care therapeutic agent gemcitabine. MATERIALS AND METHODS: Murine Panc02 cells were injected directly into the spleen or pancreas of C57BL/6 mice to respectively create models of metastatic and locally advanced pancreatic cancer. Beginning 7 d post-Panc02 injection, treated mice received 20 mg/kg gemcitabine i.p. every 3 d. Animals were sacrificed when the untreated mice became moribund and tumor/liver weight used to assess tumor burden. RESULTS: Untreated mice became moribund 22 d after pancreatic Panc02 injection. Gross analysis revealed localized pancreatic tumors weighing 1.063 g. Intrasplenic Panc02 injection produced extensive liver metastasis by d 15 when the untreated mice first became moribund. Liver weights at this time averaged 3.6 g compared with the average non-tumor-bearing weight of 1.23 g. Gemcitabine therapy resulted in a 54% decrease in localized pancreatic tumor weight and 62.5% decrease in metastatic liver weight. Additionally, gemcitabine therapy extended animal survival to 20.5 d compared with 18.0 d average for the untreated mice. CONCLUSIONS: We describe two models depicting both locally advanced and metastatic pancreatic cancer in immunocompetent mice. In efforts to establish baseline therapeutic efficacy, we determined that gemcitabine reduces tumor burden in both models and enhances survival in the metastatic model. These clinically relevant models provide valuable tools to evaluate novel therapeutics in pancreatic cancer.


Asunto(s)
Modelos Animales de Enfermedad , Inmunocompetencia/inmunología , Ratones , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/secundario , Animales , Antimetabolitos Antineoplásicos/farmacología , División Celular/efectos de los fármacos , Línea Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Ratones Endogámicos C57BL , Trasplante de Neoplasias/métodos , Páncreas/inmunología , Neoplasias Pancreáticas/tratamiento farmacológico , Índice de Severidad de la Enfermedad , Bazo/inmunología , Tasa de Supervivencia , Gemcitabina
11.
Proc Natl Acad Sci U S A ; 106(33): 13814-9, 2009 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-19666492

RESUMEN

Fli-1 and Erg are closely related members of the Ets family of transcription factors. Both genes are translocated in human cancers, including Ewing's sarcoma, leukemia, and in the case of Erg, more than half of all prostate cancers. Although evidence from mice and humans suggests that Fli-1 is required for megakaryopoiesis, and that Erg is required for normal adult hematopoietic stem cell (HSC) regulation, their precise physiological roles remain to be defined. To elucidate the relationship between Fli-1 and Erg in hematopoiesis, we conducted an analysis of mice carrying mutations in both genes. Our results demonstrate that there is a profound genetic interaction between Fli-1 and Erg. Double heterozygotes displayed phenotypes more dramatic than single heterozygotes: severe thrombocytopenia, with a significant deficit in megakaryocyte numbers and evidence of megakaryocyte dysmorphogenesis, and loss of HSCs accompanied by a reduction in the number of committed hematopoietic progenitor cells. These results illustrate an indispensable requirement for both Fli-1 and Erg in normal HSC and megakaryocyte homeostasis, and suggest these transcription factors may coregulate common target genes.


Asunto(s)
Regulación de la Expresión Génica , Megacariocitos/citología , Proteínas Oncogénicas/química , Proteína Proto-Oncogénica c-fli-1/química , Animales , Plaquetas/metabolismo , Linaje de la Célula , Cruzamientos Genéticos , Heterocigoto , Humanos , Ratones , Ratones Transgénicos , Modelos Genéticos , Mutación , Trombopoyetina/metabolismo , Factores de Transcripción , Regulador Transcripcional ERG
12.
J Biol Chem ; 285(15): 11258-69, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20139077

RESUMEN

The Ets family of transcription factors control a myriad of cellular processes and contribute to the underlying genetic loss of cellular homeostasis resulting in cancer. PDEF (prostate-derived Ets factor) has been under investigation for its role in tumor development and progression. However, the role of PDEF in cancer development has been controversial. Some reports link PDEF to tumor promoter, and others show tumor-suppressing functions in various systems under different conditions. So far, there has been no conclusive evidence from in vivo experiments to prove the role of PDEF. We have used both in vitro and in vivo systems to provide a conclusive role of PDEF in the progression process. PDEF-expressing cells block the cell growth rate, and this retardation was reversible when PDEF expression was silenced with PDEF-specific small interfering RNA. When these PDEF-expressing cells were orthotopically implanted into the mouse mammary gland, tumor incidence and growth rate were significantly retarded. Cell cycle analysis revealed that PDEF expression partially blocked cell cycle progression at G(1)/S without an effect on apoptosis. PDEF overexpression resulted in an increase in p21/CIP1 at both the mRNA and protein levels, resulting in decreased Cdk2 activity. Promoter deletion analysis, electrophoresis mobility shift assays, and chromatin immunoprecipitation studies identified the functional Ets DNA binding site at -2118 bp of the p21/CIP1 gene promoter. This site is capable of binding and responding to PDEF. Furthermore, we silenced p21/CIP1 expression in PDEF-overexpressing cells by small interfering RNA. p21-silenced PDEF cells exhibited significantly increased cell growth in vitro and in vivo, demonstrating the p21 regulation by PDEF as a key player. These experiments identified PDEF as a new transcription factor that directly regulates p21/CIP1 expression under non-stressed conditions. This study conclusively proves that PDEF is a breast tumor suppressor for the first time using both in vitro and in vivo systems. PDEF can be further developed as a target for designing therapeutic intervention of breast cancer.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Mamarias Animales/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Animales , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Silenciador del Gen , Humanos , Ratones , Trasplante de Neoplasias
13.
Prostate ; 71(16): 1723-35, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21446014

RESUMEN

BACKGROUND: Ets is a large family of transcriptional regulators with functions in most biological processes. While the Ets family gene, prostate-derived epithelial factor (PDEF), is expressed in epithelial tissues, PDEF protein expression has been found to be reduced or lost during cancer progression. The goal of this study was to examine the mechanism for and biologic impact of altered PDEF expression in prostate cancer. METHODS: PDEF protein expression of prostate specimens was examined by immunohistochemistry. RNA and protein expression in cell lines were measured by q-PCR and Western blot, respectively. Cellular growth was determined by quantifying viable and apoptotic cells over time. Cell cycle was measured by flow cytometry. Migration and invasion were determined by transwell assays. PDEF promoter occupancy was determined by chromatin immunoprecipitation (ChIP). RESULTS: While normal prostate epithelium expresses PDEF mRNA and protein, tumors show no or decreased PDEF protein expression. Re-expression of PDEF in prostate cancer cells inhibits cell growth. PDEF expression is inversely correlated with survivin, urokinase plasminogen activator (uPA) and slug expression and ChIP studies identify survivin and uPA as direct transcriptional targets of PDEF. This study also shows that PDEF expression is regulated via a functional microRNA-204 (miR-204) binding site within the 3'UTR. Furthermore, we demonstrate the biologic significance of miR-204 expression and that miR-204 is over-expressed in human prostate cancer specimens. CONCLUSIONS: Collectively, the reported studies demonstrate that PDEF is a negative regulator of tumor progression and that the miR-204-PDEF regulatory axis contributes to PDEF protein loss and resultant cancer progression.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , MicroARNs/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/fisiopatología , Proteínas Proto-Oncogénicas c-ets/genética , Ciclo Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Núcleo Celular/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Humanos , Masculino , Invasividad Neoplásica/fisiopatología , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo
14.
Am J Pathol ; 176(4): 1983-98, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20228226

RESUMEN

Systemic sclerosis or scleroderma (SSc) is a complex autoimmune connective tissue disease characterized by obliterative vasculopathy and tissue fibrosis. The molecular mechanisms underlying SSc vasculopathy are largely unknown. Friend leukemia integration factor 1 (Fli1), an important regulator of immune function and collagen fibrillogenesis, is expressed at reduced levels in endothelial cells in affected skin of patients with SSc. To develop a disease model and to investigate the function of Fli1 in the vasculature, we generated mice with a conditional deletion of Fli1 in endothelial cells (Fli1 CKO). Fli1 CKO mice showed a disorganized dermal vascular network with greatly compromised vessel integrity and markedly increased vessel permeability. We show that Fli1 regulates expression of genes involved in maintaining vascular homeostasis including VE-cadherin, platelet endothelial cell adhesion molecule 1, type IV collagen, matrix metalloproteinase 9, platelet-derived growth factor B, and S1P(1) receptor. Accordingly, Fli1 CKO mice are characterized by down-regulation of VE-cadherin and platelet endothelial cell adhesion molecule 1, impaired development of basement membrane, and a decreased presence of alpha-smooth muscle actin-positive cells in dermal microvessels. This phenotype is consistent with a role of Fli1 as a regulator of vessel maturation and stabilization. Importantly, vascular characteristics of Fli1 CKO mice are recapitulated by SSc microvasculature. Thus, persistently reduced levels of Fli1 in endothelial cells may play a critical role in the development of SSc vasculopathy.


Asunto(s)
Proteína Proto-Oncogénica c-fli-1/genética , Esclerodermia Sistémica/metabolismo , Enfermedades Vasculares/patología , Actinas/metabolismo , Alelos , Animales , Humanos , Inmunohistoquímica/métodos , Ratones , Ratones Noqueados , Microcirculación , Permeabilidad , Fenotipo , Proteína Proto-Oncogénica c-fli-1/fisiología , Piel/patología
15.
J Surg Res ; 170(1): 56-63, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21470622

RESUMEN

BACKGROUND: Epithelial-to-mesenchymal transition (EMT) is a series of molecular changes allowing epithelial cancer cells to acquire properties of mesenchymal cells: increased motility, invasion, and protection from apoptosis. Transcriptional regulators such as Slug mediate EMT, working in part to repress E-cadherin transcription. We report a novel, noninvasive in vivo rectal cancer model to explore the role of Slug in colorectal cancer (CRC) tumor development. METHODS: For the generation of DLD-1 cells overexpressing Slug (Slug DLD-1), a Slug or empty (Empty DLD-1) pCMV-3Tag-1 (kanamycin-resistant) vector was used for transfection. Cells were evaluated for Slug and E-cadherin expression, and cell migration and invasion. For the in vivo study, colon cancer cells (parental DLD-1, Slug DLD-1, empty DLD-1, and HCT-116) were submucosally injected into the posterior rectum of nude mice using endoscopic guidance. After 28 d, tumors were harvested and tissue was analyzed. RESULTS: Slug expression in our panel of colon cancer cell lines was inversely correlated with E-cadherin expression and enhanced migration/invasion. Slug DLD-1 cells demonstrated a 21-fold increased Slug and 19-fold decreased E-cadherin expression compared with empty DLD-1. Similarly, the Slug DLD-1 cells had significantly enhanced cellular migration and invasion. In the orthotopic rectal cancer model, Slug DLD-1 cells formed rectal tumors in 9/10 (90%) of the mice (mean volume = 458 mm(3)) compared with only 1/10 (10%) with empty DLD-1 cells. CONCLUSION: Slug mediates EMT with enhanced in vivo rectal tumor formation. Our noninvasive in vivo model enables researchers to explore the molecular consequences of altered genes in a clinically relevant rectal cancer in an effort to develop novel therapeutic approaches for patients with rectal cancer.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias del Recto/etiología , Factores de Transcripción/fisiología , Animales , Cadherinas/análisis , Línea Celular Tumoral , Movimiento Celular , Humanos , Ratones , Neoplasias del Recto/patología , Factores de Transcripción de la Familia Snail , Factores de Transcripción/análisis
16.
Prostaglandins Other Lipid Mediat ; 96(1-4): 41-4, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21983220

RESUMEN

We have previously found increased expression of thromboxane synthase (TXAS) and thromboxane receptor (TP) beta isoform in the tissues of patients with bladder cancer. Studies in cell lines and mice have indicated a potential significant role of the thromboxane signaling pathway in the pathogenesis of human bladder cancer. This study was designed to determine if the changes observed in the tissues of patients with bladder cancer were mirrored by changes in the urine of these patients. We found increased levels of thromboxane B(2) (TXB(2)) the major metabolite of TXAS and increased levels of the TPß receptor. These results raised the possibility that patients with bladder cancer may be followed for progression or remission of their disease by quantitation of these substances in their urine.


Asunto(s)
Biomarcadores de Tumor/orina , Isoformas de Proteínas/orina , Receptores de Tromboxanos/biosíntesis , Transducción de Señal/genética , Tromboxano B2/orina , Tromboxano-A Sintasa/orina , Neoplasias de la Vejiga Urinaria/orina , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Isoformas de Proteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Tromboxanos/genética , Tromboxano-A Sintasa/genética , Estados Unidos , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/patología
17.
Mol Cell Biol ; 27(9): 3353-66, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17339335

RESUMEN

Ets1 is a member of the Ets transcription factor family. Alternative splicing of exon VII results in two naturally occurring protein isoforms: full-length Ets1 (p51-Ets1) and Ets1(DeltaVII) (p42-Ets1). These isoforms bear key distinctions regarding protein-protein interactions, DNA binding kinetics, and transcriptional target specificity. Disruption of both Ets1 isoforms in mice results in the loss of detectable NK and NKT cell activity and defects in B and T lymphocytes. We generated mice that express only the Ets1(DeltaVII) isoform. Ets1(DeltaVII) homozygous mice express no p51-Ets1 and elevated levels of the p42-Ets1 protein relative to the wild type and display increased perinatal lethality, thymomegaly, and peripheral lymphopenia. Proliferation was increased in both the thymus and the spleen, while apoptosis was decreased in the thymus and increased in the spleen of homozygotes. Significant elevations of CD8(+) and CD8(+)CD4(+) thymocytes were observed. Lymphoid cell (CD19(+), CD4(+), and CD8(+)) reductions were predominantly responsible for diminished spleen cellularity, with fewer memory cells and a failure of homeostatic proliferation to maintain peripheral lymphocytes. Collectively, the Ets1(DeltaVII) mutants demonstrate lymphocyte maturation defects associated with misregulation of p16(Ink4a), p27(Kip1), and CD44. Thus, a balance in the differential regulation of Ets1 isoforms represents a potential mechanism in the control of lymphoid maturation and homeostasis.


Asunto(s)
Homeostasis , Linfocitos/citología , Linfocitos/metabolismo , Proteína Proto-Oncogénica c-ets-1/deficiencia , Proteína Proto-Oncogénica c-ets-1/metabolismo , Bazo/metabolismo , Timo/metabolismo , Animales , Secuencia de Bases , Proliferación Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación de la Expresión Génica , Heterocigoto , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Ratones , Datos de Secuencia Molecular , Fenotipo , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína Proto-Oncogénica c-ets-1/genética , Bazo/citología , Timo/citología , Transcripción Genética/genética
18.
J Immunol ; 181(3): 1644-54, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18641300

RESUMEN

Fli-1 belongs to the Ets transcription factor family and is expressed primarily in hematopoietic cells, including most cells active in immunity. To assess the role of Fli-1 in lymphocyte development in vivo, we generated mice that express a truncated Fli-1 protein, lacking the C-terminal transcriptional activation domain (Fli-1(DeltaCTA)). Fli-1(DeltaCTA)/Fli-1(DeltaCTA) mice had significantly fewer splenic follicular B cells, and an increased number of transitional and marginal zone B cells, compared with wild-type controls. Bone marrow reconstitution studies demonstrated that this phenotype is the result of lymphocyte intrinsic effects. Expression of Igalpha and other genes implicated in B cell development, including Pax-5, E2A, and Egr-1, are reduced, while Id1 and Id2 are increased in Fli-1(DeltaCTA)/Fli-1(DeltaCTA) mice. Proliferation of B cells from Fli-1(DeltaCTA)/Fli-1(DeltaCTA) mice was diminished, although intracellular Ca(2+) flux in B cells from Fli-1(DeltaCTA)/Fli-1(DeltaCTA) mice was similar to that of wild-type controls after anti-IgM stimulation. Immune responses and in vitro class switch recombination were also altered in Fli-1(DeltaCTA)/Fli-1(DeltaCTA) mice. Thus, Fli-1 modulates B cell development both centrally and peripherally, resulting in a significant impact on the in vivo immune response.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Diferenciación Celular/inmunología , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Proteína Proto-Oncogénica c-fli-1/metabolismo , Animales , Linfocitos B/citología , Médula Ósea/inmunología , Médula Ósea/metabolismo , Proliferación Celular , Células Cultivadas , Regulación de la Expresión Génica , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulina G/inmunología , Tejido Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Proto-Oncogénica c-fli-1/deficiencia , Proteína Proto-Oncogénica c-fli-1/genética , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología
19.
J Cell Biochem ; 108(6): 1389-98, 2009 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-19830706

RESUMEN

ETS is a family of transcriptional regulators with functions in most biological processes. Dysregulated ETS factor function leads to altered expression of multiple genes that play critical roles in many of the processes required for cancer progression. While the Ets family gene, prostate-derived ETS factor (PDEF), is expressed in epithelial tissues including prostate, breast, and colon, PDEF protein expression has been found to be reduced or lost during prostate and breast cancer progression. The goal of this study was to examine the expression and biologic impact of altered PDEF expression in colon cancer. PDEF mRNA and protein are not detectable in several colon-cancer-derived cell lines. Re-expression of PDEF in colon cancer cells inhibits growth and migration. Growth affects are due to altered cellular proliferation, indicated by increased altered cell population in G(1) and S phases of the cell cycle, as well as increased apoptosis. Relevant to its modulation of growth and migration phenotypes, PDEF expression resulted in altered expression of genes with established roles in cell cycle, motility, and invasion. Furthermore, chromatin immunoprecipitation studies show that p21 and urokinase plasminogen activator (uPA) are direct PDEF transcriptional targets. While non-tumor colon epithelium expresses PDEF mRNA and protein, the majority of tumors showed decreased mRNA and/or protein expression. In human tumor tissue samples, PDEF expression was inversely correlated with the expression levels of uPA. Collectively, the data support the model that PDEF is a negative regulator of tumor progression by modulating the expression of growth and migration promoting genes.


Asunto(s)
Movimiento Celular , Neoplasias del Colon/metabolismo , Proteínas Proto-Oncogénicas c-ets/genética , Células CACO-2/metabolismo , Procesos de Crecimiento Celular , Línea Celular Tumoral , Neoplasias del Colon/genética , Humanos , Proteínas Proto-Oncogénicas c-ets/metabolismo , ARN Mensajero/metabolismo , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
20.
Cancer Res ; 67(4): 1618-25, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17308102

RESUMEN

Cell migration and invasion are critical events during the progression to metastasis. Without motile function, cancer cells are unable to leave the primary tumor site, invade through the basement membrane, and form secondary tumors. Expression of the epithelial-specific ETS factor prostate-derived ETS factor (PDEF) is reduced in human invasive breast tissue and lost in invasive breast cancer cell lines. Gain-of-function studies that examine different aspects of cell migration show that constitutive or inducible PDEF reexpression inhibits migration and invasion in multiple breast cancer cell lines, and loss-of-function studies show a stimulation of migration in noninvasive breast cancer cells. Furthermore, the introduction of PDEF into invasive breast cancer cells led to a remodeling of the actin cytoskeleton and altered focal adhesion localization and adherence levels. Cells expressing PDEF no longer form the defined morphologic polarity required for efficient, directional migration. Collectively, these data indicate that PDEF down-regulation in invasive breast cancer may promote actin-mediated cell migration through the extracellular matrix.


Asunto(s)
Neoplasias de la Mama/patología , Movimiento Celular/fisiología , Proteínas Proto-Oncogénicas c-ets/biosíntesis , Actinas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Humanos , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-ets/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA