Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 41(5): 2857-68, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23325854

RESUMEN

Nucleosome positioning on the chromatin strand plays a critical role in regulating accessibility of DNA to transcription factors and chromatin modifying enzymes. Hence, detailed information on nucleosome depletion or movement at cis-acting regulatory elements has the potential to identify predicted binding sites for trans-acting factors. Using a novel method based on enrichment of mononucleosomal DNA by bacterial artificial chromosome hybridization, we mapped nucleosome positions by deep sequencing across 250 kb, encompassing the cystic fibrosis transmembrane conductance regulator (CFTR) gene. CFTR shows tight tissue-specific regulation of expression, which is largely determined by cis-regulatory elements that lie outside the gene promoter. Although multiple elements are known, the repertoire of transcription factors that interact with these sites to activate or repress CFTR expression remains incomplete. Here, we show that specific nucleosome depletion corresponds to well-characterized binding sites for known trans-acting factors, including hepatocyte nuclear factor 1, Forkhead box A1 and CCCTC-binding factor. Moreover, the cell-type selective nucleosome positioning is effective in predicting binding sites for novel interacting factors, such as BAF155. Finally, we identify transcription factor binding sites that are overrepresented in regions where nucleosomes are depleted in a cell-specific manner. This approach recognizes the glucocorticoid receptor as a novel trans-acting factor that regulates CFTR expression in vivo.


Asunto(s)
Mapeo Cromosómico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Silenciador del Gen , Nucleosomas/metabolismo , Receptores de Glucocorticoides/fisiología , Sitios de Unión , Factor de Unión a CCCTC , Células CACO-2 , Inmunoprecipitación de Cromatina , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Dexametasona/farmacología , Sitios Genéticos , Glucocorticoides/farmacología , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Nucleosomas/genética , Unión Proteica , Receptores de Glucocorticoides/metabolismo , Proteínas Represoras/metabolismo , Elementos de Respuesta , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo
2.
Nucleic Acids Res ; 40(2): 625-37, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21948798

RESUMEN

Access to regulatory elements of the genome can be inhibited by nucleosome core particles arranged along the DNA strand. Hence, sites that are accessible by transcription factors may be located by using nuclease digestion to identify the relative nucleosome occupancy of a genomic region. In order to define novel cis regulatory elements in the ∼2.7-kb promoter region of the cystic fibrosis transmembrane conductance regulator (CFTR) gene, we define its nucleosome occupancy. This profile reveals the precise positions of nucleosome-free regions (NFRs), both cell-type specific and others apparently unrelated to CFTR-expression level and offer the first high-resolution map of the chromatin structure of the entire CFTR promoter in relevant cell types. Several of these NFRs are strongly bound by nuclear factors in a sequence-specific manner, and directly influence CFTR promoter activity. Sequences within the NFR1 and NFR4 elements are highly conserved in many human gene promoters. Moreover, NFR1 contributes to promoter activity of another gene, angiopoietin-like 3 (ANGPTL3), while NFR4 is constitutively nucleosome-free in promoters genome wide. Conserved motifs within NFRs of the CFTR promoter also show a high level of protection from DNase I digestion genome-wide, and likely have important roles in the positioning of nucleosome core particles more generally.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Secuencia de Bases , Células Cultivadas , Secuencia Conservada , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Factores de Transcripción/metabolismo , Transcripción Genética
3.
Biol Reprod ; 89(4): 104, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24006278

RESUMEN

The epithelium lining the epididymis has a pivotal role in ensuring a luminal environment that can support normal sperm maturation. Many of the individual genes that encode proteins involved in establishing the epididymal luminal fluid are well characterized. They include ion channels, ion exchangers, transporters, and solute carriers. However, the molecular mechanisms that coordinate expression of these genes and modulate their activities in response to biological stimuli are less well understood. To identify cis-regulatory elements for genes expressed in human epididymis epithelial cells, we generated genome-wide maps of open chromatin by DNase-seq. This analysis identified 33,542 epididymis-selective DNase I hypersensitive sites (DHS), which were not evident in five cell types of different lineages. Identification of genes with epididymis-selective DHS at their promoters revealed gene pathways that are active in immature epididymis epithelial cells. These include processes correlating with epithelial function and also others with specific roles in the epididymis, including retinol metabolism and ascorbate and aldarate metabolism. Peaks of epididymis-selective chromatin were seen in the androgen receptor gene and the cystic fibrosis transmembrane conductance regulator (CFTR) gene, which has a critical role in regulating ion transport across the epididymis epithelium. In silico prediction of transcription factor binding sites that were overrepresented in epididymis-selective DHS identified epithelial transcription factors, including ELF5 and ELF3, the androgen receptor, Pax2, and Sox9, as components of epididymis transcriptional networks. Active genes, which are targets of each transcription factor, reveal important biological processes in the epididymis epithelium.


Asunto(s)
Ensamble y Desensamble de Cromatina , Epidídimo/metabolismo , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Infertilidad Masculina/genética , Espermatogénesis , Células Cultivadas , Mapeo Cromosómico , Biología Computacional , ADN Intergénico , Epidídimo/citología , Epidídimo/crecimiento & desarrollo , Epidídimo/fisiopatología , Células Epiteliales/citología , Sistemas Especialistas , Feto/citología , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Infertilidad Masculina/metabolismo , Infertilidad Masculina/fisiopatología , Masculino , Nucleosomas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Regiones Promotoras Genéticas
4.
Thorax ; 67(5): 385-91, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22169360

RESUMEN

BACKGROUND: Distal cell-type-specific regulatory elements may be located at very large distances from the genes that they control and are often hidden within intergenic regions or in introns of other genes. The development of methods that enable mapping of regions of open chromatin genome wide has greatly advanced the identification and characterisation of these elements. METHODS: Here we use DNase I hypersensitivity mapping followed by deep sequencing (DNase-seq) to generate a map of open chromatin in primary human tracheal epithelial (HTE) cells and use bioinformatic approaches to characterise the distribution of these sites within the genome and with respect to gene promoters, intronic and intergenic regions. RESULTS: Genes with HTE-selective open chromatin at their promoters were associated with multiple pathways of epithelial function and differentiation. The data predict novel cell-type-specific regulatory elements for genes involved in HTE cell function, such as structural proteins and ion channels, and the transcription factors that may interact with them to control gene expression. Moreover, the map of open chromatin can identify the location of potentially critical regulatory elements in genome-wide association studies (GWAS) in which the strongest association is with single nucleotide polymorphisms in non-coding regions of the genome. We demonstrate its relevance to a recent GWAS that identifies modifiers of cystic fibrosis lung disease severity. CONCLUSION: Since HTE cells have many functional similarities with bronchial epithelial cells and other differentiated cells in the respiratory epithelium, these data are of direct relevance to elucidating the molecular basis of normal lung function and lung disease.


Asunto(s)
Cromatina/genética , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Pulmón/fisiología , Mucosa Respiratoria/metabolismo , Tráquea/citología , Mapeo Cromosómico/métodos , Biología Computacional , Desoxirribonucleasa I , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Pulmón/metabolismo
5.
J Biol Chem ; 285(47): 36922-32, 2010 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-20833714

RESUMEN

The mechanism(s) behind folate rescue of neural tube closure are not well understood. In this study we show that maternal intake of folate prior to conception reverses the proliferation potential of neural crest stem cells in homozygous Splotch embryos (Sp(-/-)) via epigenetic mechanisms. It is also shown that the pattern of differentiation seen in these cells is similar to wild-type (WT). Cells from open caudal neural tubes of Sp(-/-) embryos exhibit increased H3K27 methylation and decreased expression of KDM6B possibly due to up-regulation of KDM6B targeting micro-RNAs such as miR-138, miR-148a, miR-185, and miR-339-5p. In our model, folate reversed these epigenetic marks in folate-rescued Sp(-/-) embryos. Using tissue from caudal neural tubes of murine embryos we also examined H3K27me2 and KDM6B association with Hes1 and Neurog2 promoters at embryonic day E10.5, the proliferative stage, and E12.5, when neural differentiation begins. In Sp(-/-) embryos compared with WT, levels of H3K27me2 associated with the Hes1 promoter were increased at E10.5, and levels associated with the Neurog2 promoter were increased at E12.5. KDM6B association with Hes1 and Neurog2 promoters was inversely related to H3K27me2 levels. These epigenetic changes were reversed in folate-rescued Sp(-/-) embryos. Thus, one of the mechanisms by which folate may rescue the Sp(-/-) phenotype is by increasing the expression of KDM6B, which in turn decreases H3K27 methylation marks on Hes1 and Neurog2 promoters thereby affecting gene transcription.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Ácido Fólico/administración & dosificación , Proteínas de Homeodominio/genética , Proteínas del Tejido Nervioso/genética , Tubo Neural/efectos de los fármacos , Tubo Neural/embriología , Regiones Promotoras Genéticas/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Western Blotting , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sistema Nervioso Central/embriología , Ensamble y Desensamble de Cromatina/genética , Inmunoprecipitación de Cromatina , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Epigenómica , Femenino , Técnica del Anticuerpo Fluorescente , Ácido Fólico/farmacología , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Técnicas para Inmunoenzimas , Inmunoprecipitación , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Luciferasas/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , MicroARNs/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Defectos del Tubo Neural/prevención & control , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/fisiología , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción HES-1 , Complejo Vitamínico B/administración & dosificación , Complejo Vitamínico B/farmacología
6.
Sarcoma ; 2011: 732690, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21789031

RESUMEN

A limited number of reports have investigated the role of microRNAs in osteosarcoma. In this study, we performed miRNA expression profiling of osteosarcoma cell lines, tumor samples, and normal human osteoblasts. Twenty-two differentially expressed microRNAs were identified using high throughput real-time PCR analysis, and 4 (miR-135b, miR-150, miR-542-5p, and miR-652) were confirmed and validated in a different group of tumors. Both miR-135b and miR-150 have been previously shown to be important in cancer. We hypothesize that dysregulation of differentially expressed microRNAs may contribute to tumorigenesis. They might also represent molecular biomarkers or targets for drug development in osteosarcoma.

7.
Am J Respir Cell Mol Biol ; 43(3): 334-41, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19855085

RESUMEN

The cystic fibrosis transmembrane conductance regulator (CFTR) gene is driven by a promoter that cannot alone account for the temporal and tissue-specific regulation of the gene. This has led to the search for additional regulatory elements that cooperate with the basal promoter to achieve coordinated expression. We previously identified two alternative upstream exons of the gene that were mutually exclusive of the first exon, and one of which showed temporal regulation in the human and sheep lung. We now demonstrate that this alternative splice product generates a stable protein, which initiates translation at an ATG in exon 4, and thus lacks the N terminus of CFTR. The other splice variant inhibits translation of the protein. In a search for the promoter used by the upstream exons, we identified a novel element that contributes to the activity of the basal CFTR promoter in airway epithelial cells, but does not function independently. Finally, we demonstrate that, in primary airway cells, skin fibroblasts, and both airway and intestinal cell lines, the CFTR promoter is unmethylated, irrespective of CFTR expression status. Thus, methylation is not the main cause of inactivation of CFTR transcription.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Metilación de ADN , Regulación de la Expresión Génica , Regiones Promotoras Genéticas/genética , Transcripción Genética/genética , Empalme Alternativo , Secuencia de Bases , Bronquios/citología , Bronquios/metabolismo , Células CACO-2/metabolismo , Células Cultivadas , Fibrosis Quística/patología , Células Epiteliales/metabolismo , Exones/genética , Humanos , Intestino Delgado/metabolismo , Luciferasas/metabolismo , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Secuencias Reguladoras de Ácidos Nucleicos
8.
BMC Cancer ; 10: 471, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20809981

RESUMEN

BACKGROUND: Chondrosarcomas are malignant cartilage tumors that do not respond to traditional chemotherapy or radiation. The 5-year survival rate of histologic grade III chondrosarcoma is less than 30%. An animal model of chondrosarcoma has been established--namely, the Swarm Rat Chondrosarcoma (SRC)--and shown to resemble the human disease. Previous studies with this model revealed that tumor microenvironment could significantly influence chondrosarcoma malignancy. METHODS: To examine the effect of the microenvironment, SRC tumors were initiated at different transplantation sites. Pyrosequencing assays were utilized to assess the DNA methylation of the tumors, and SAGE libraries were constructed and sequenced to determine the gene expression profiles of the tumors. Based on the gene expression analysis, subsequent functional assays were designed to determine the relevancy of the specific genes in the development and progression of the SRC. RESULTS: The site of transplantation had a significant impact on the epigenetic and gene expression profiles of SRC tumors. Our analyses revealed that SRC tumors were hypomethylated compared to control tissue, and that tumors at each transplantation site had a unique expression profile. Subsequent functional analysis of differentially expressed genes, albeit preliminary, provided some insight into the role that thymosin-ß4, c-fos, and CTGF may play in chondrosarcoma development and progression. CONCLUSION: This report describes the first global molecular characterization of the SRC model, and it demonstrates that the tumor microenvironment can induce epigenetic alterations and changes in gene expression in the SRC tumors. We documented changes in gene expression that accompany changes in tumor phenotype, and these gene expression changes provide insight into the pathways that may play a role in the development and progression of chondrosarcoma. Furthermore, specific functional analysis indicates that thymosin-ß4 may have a role in chondrosarcoma metastasis.


Asunto(s)
Biomarcadores de Tumor/genética , Condrosarcoma/genética , Epigénesis Genética , Perfilación de la Expresión Génica , Neoplasias Pulmonares/etiología , Tibia/patología , Animales , Biomarcadores de Tumor/metabolismo , Western Blotting , Cartílago/metabolismo , Cartílago/patología , Condrosarcoma/metabolismo , Condrosarcoma/patología , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Metilación de ADN , Genes fos/fisiología , Humanos , Inyecciones Subcutáneas , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Desnudos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Ratas , Ratas Sprague-Dawley , Timosina/genética , Timosina/metabolismo , Tibia/metabolismo , Células Tumorales Cultivadas/trasplante
9.
Cancer Prev Res (Phila) ; 10(1): 89-97, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27789587

RESUMEN

miRNAs are noncoding RNAs with abnormal expression in breast cancer; their expression in high-risk benign breast tissue may relate to breast cancer risk. We examined miRNA profiles in contralateral unaffected breasts (CUB) of patients with breast cancer and validated resulting candidates in two additional sample sets. Expression profiles of 754 mature miRNAs were examined using TaqMan Low Density Arrays in 30 breast cancer samples [15 estrogen receptor (ER)-positive and 15 ER-negative] and paired CUBs and 15 reduction mammoplasty controls. Pairwise comparisons identified miRNAs with significantly differential expression. Seven candidate miRNAs were examined using qRT-PCR in a second CUB sample set (40 cases, 20 ER+, 20 ER-) and 20 reduction mammoplasty controls. Further validation was performed in 80 benign breast biopsy (BBB) samples; 40 from cases who subsequently developed breast cancer and 40 from controls who did not. Logistic regression, using tertiles of miRNA expression, was used to discriminate cases from controls. Seven miRNAs were differentially expressed in tumors and CUBs versus reduction mammoplasty samples. Among them, miR-18a and miR-210 were validated in the second CUB set, showing significantly higher expression in tumor and CUBs than in reduction mammoplasty controls. The expression of miR-18a and miR-210 was also significantly higher in BBB cases than in BBB controls. When both miR-18a and miR-210 were expressed in the upper tertiles in BBB, OR for subsequent cancer was 3.20, P = 0.023. miR-18a and miR-210 are expressed at higher levels in CUBs of patients with breast cancer, and in BBB prior to cancer development, and are therefore candidate breast cancer risk biomarkers. Cancer Prev Res; 10(1); 89-97. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Carcinogénesis/patología , MicroARNs/genética , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Biopsia , Mama , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/metabolismo , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Estrógenos/metabolismo
10.
Hum Mutat ; 27(6): 545-52, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16671097

RESUMEN

Pseudogenes are remnants of gene duplication (nonprocessed pseudogenes) and retrotransposition (processed pseudogenes) events. This study describes methods for identifying gene conversion candidates from predicted pseudogenes. Pseudogenes may accumulate and harbor sequence variations over time that become disease-causing mutations when transferred to genes by gene conversion. A total of 14,476 pseudogenes were identified, including 3,426 nonprocessed pseudogenes. In addition, 1,945 nonprocessed pseudogenes that are localized near their progenitor gene were evaluated for their possible role in gene conversion and disease. All 11 known, human cases of gene conversion (with deleterious effects) involving pseudogenes were successfully identified by these methods. Among the pseudogenes identified is a retinitis pigmentosa 9 (RP9) pseudogene that carries a c.509A>G mutation which produces a p.Asp170Gly substitution that is associated with the RP9 form of autosomal dominant retinitis pigmentosa (adRP). The c.509A>G mutation in RP9 is a previously unrecognized example of gene conversion between the progenitor gene and its pseudogene. Notably, two processed pseudogenes also contain mutations associated with diseases. An inosine monophosphate dehydrogenase 1 (IMPDH1) pseudogene carries a c.676G>A mutation that produces a p.Asp226Asn substitution that causes the retinitis pigmentosa 10 (RP10) form of adRP; and a phosphoglycerate kinase 1 (PGK1) pseudogene (PGK1P1) carries a c.837T>C mutation that produces a p.Ile252Thr substitution that is associated with a phosphoglycerate kinase deficiency. Ranking of nonprocessed pseudogenes as candidates for gene conversion was also performed based on the sequence characteristics of published cases of pseudogene-mediated gene conversion. All results and tools produced by this study are available for download at: http://genome.uiowa.edu/pseudogenes.


Asunto(s)
Análisis Mutacional de ADN/métodos , Conversión Génica/fisiología , Genómica/métodos , Seudogenes , Biología Computacional/métodos , Humanos , IMP Deshidrogenasa/genética , Fosfoglicerato Quinasa/deficiencia , Proteínas/genética , Factores de Empalme de ARN , Retinitis Pigmentosa/genética
11.
Epigenetics ; 6(4): 450-64, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21343701

RESUMEN

Melanoma progression requires deregulation of gene expression by currently uncharacterized epigenetic mechanisms. A mouse model based on changes in cell microenvironment was developed by our group to study melanocyte malignant transformation. Melanoma cell lines (4C11- and 4C11+) were obtained as result of 5 sequential anchorage blockades of non-tumorigenic melan-a melanocytes. Melan-a cells submitted to 4 de-adhesion cycles were also established (4C), are non-tumorigenic and represent an intermediary phase of tumor progression. The aim of this work was to identify factors contributing to epigenetic modifications in early and later phases of malignant transformation induced by anchorage impediment. Epigenetic alterations occur early in tumorigenesis; 4C cell line shows changes in global and gene-specific DNA methylation and histone marks. Many histone modifications differ between melan-a, 4C, 4C11- (non-metastatic melanoma cell line) and 4C11+ (metastatic melanoma cell line) which could be associated with changes in gene and microRNA expression. These epigenetic alterations seem to play a key role in malignant transformation since melanocytes treated with 5-Aza-2'-deoxycytidine before each anchorage blockade do not transform. Some epigenetic changes seem to be also responsible for the maintenance of malignant phenotype, since melanoma cell lines (4C11- and 4C11+) treated in vitro with 5-Aza-2'-deoxycytidine or Trichostatin A showed reduction of tumor growth in vivo. Changes in gene expression reflecting cell adaptation to new environment were also observed. We propose a model in which sustained microenvironmental stress in melanocytes results in epigenetic reprogramming. Thus, after adaptation, cells may acquire epigenetic marks that could contribute to the establishment of a malignant phenotype.


Asunto(s)
Transformación Celular Neoplásica/genética , Metilación de ADN , Epigénesis Genética , Histonas/metabolismo , Melanocitos/fisiología , Animales , Línea Celular , Transformación Celular Neoplásica/inducido químicamente , Regulación Neoplásica de la Expresión Génica , Ratones
12.
PLoS One ; 6(10): e25114, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22053178

RESUMEN

INTRODUCTION: We have examined expression of microRNAs (miRNAs) in ependymomas to identify molecular markers of value for clinical management. miRNAs are non-coding RNAs that can block mRNA translation and affect mRNA stability. Changes in the expression of miRNAs have been correlated with many human cancers. MATERIALS AND METHODS: We have utilized TaqMan Low Density Arrays to evaluate the expression of 365 miRNAs in ependymomas and normal brain tissue. We first demonstrated the similarity of expression profiles of paired frozen tissue (FT) and paraffin-embedded specimens (FFPE). We compared the miRNA expression profiles of 34 FFPE ependymoma samples with 8 microdissected normal brain tissue specimens enriched for ependymal cells. miRNA expression profiles were then correlated with tumor location, histology and other clinicopathological features. RESULTS: We have identified miRNAs that are over-expressed in ependymomas, such as miR-135a and miR-17-5p, and down-regulated, such as miR-383 and miR-485-5p. We have also uncovered associations between expression of specific miRNAs which portend a worse prognosis. For example, we have identified a cluster of miRNAs on human chromosome 14q32 that is associated with time to relapse. We also found that miR-203 is an independent marker for relapse compared to the parameters that are currently used. Additionally, we have identified three miRNAs (let-7d, miR-596 and miR-367) that strongly correlate to overall survival. CONCLUSION: We have identified miRNAs that are differentially expressed in ependymomas compared with normal ependymal tissue. We have also uncovered significant associations of miRNAs with clinical behavior. This is the first report of clinically relevant miRNAs in ependymomas.


Asunto(s)
Biomarcadores de Tumor/genética , Ependimoma/genética , MicroARNs/genética , Adolescente , Biomarcadores de Tumor/metabolismo , Estudios de Casos y Controles , Niño , Preescolar , Biología Computacional , Ependimoma/patología , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Lactante , Masculino , MicroARNs/metabolismo , Análisis Multivariante , Pronóstico , Modelos de Riesgos Proporcionales , Análisis de Regresión , Análisis de Supervivencia
13.
Clin Cancer Res ; 16(17): 4339-52, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20601445

RESUMEN

PURPOSE: To understand the changes in gene expression in polycythemia vera (PV) progenitor cells and their relationship to JAK2V617F. EXPERIMENTAL DESIGN: Messenger RNA isolated from CD34(+) cells from nine PV patients and normal controls was profiled using Affymetrix arrays. Gene expression change mediated by JAK2V617F was determined by profiling CD34(+) cells transduced with the kinase and by analysis of leukemia cell lines harboring JAK2V617F, treated with an inhibitor. RESULTS: A PV expression signature was enriched for genes involved in hematopoietic development, inflammatory responses, and cell proliferation. By quantitative reverse transcription-PCR, 23 genes were consistently deregulated in all patient samples. Several of these genes such as WT1 and KLF4 were regulated by JAK2, whereas others such as NFIB and EVI1 seemed to be deregulated in PV by a JAK2-independent mechanism. Using cell line models and comparing gene expression profiles of cell lines and PV CD34(+) PV specimens, we have identified panels of 14 JAK2-dependent genes and 12 JAK2-independent genes. These two 14- and 12-gene sets could separate not only PV from normal CD34(+) specimens, but also other MPN such as essential thrombocytosis and primary myelofibrosis from their normal counterparts. CONCLUSIONS: A subset of the aberrant gene expression in PV progenitor cells can be attributed to the action of the mutant kinase, but there remain a significant number of genes characteristic of the disease but deregulated by as yet unknown mechanisms. Genes deregulated in PV as a result of the action of JAK2V617F or independent of the kinase may represent other targets for therapy.


Asunto(s)
Perfilación de la Expresión Génica , Janus Quinasa 2/genética , Mutación , Policitemia Vera/genética , Sustitución de Aminoácidos , Antígenos CD34/genética , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Análisis por Conglomerados , Células Eritroides/citología , Células Eritroides/metabolismo , Eritropoyetina/farmacología , Humanos , Janus Quinasa 2/metabolismo , Factor 4 Similar a Kruppel , Análisis de Secuencia por Matrices de Oligonucleótidos , Policitemia Vera/sangre , Policitemia Vera/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
14.
Epigenomics ; 1(2): 387-98, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20495621

RESUMEN

UNLABELLED: We have previously shown that the microenvironment of human embryonic stem cells (hESCs) is able to change and reprogram aggressive cancer cells to a less aggressive state. Some mechanisms implicated in the phenotypic changes observed after this exposure are mainly associated with the Nodal signaling pathway, which plays a key role in tumor cell plasticity. However, several other molecular mechanisms might be related directly and/or indirectly to these changes, including microRNA (miRNA) regulation and DNA methylation. AIM: To further explore the epigenetic mechanisms potentially underlying the phenotypic changes that occur after exposing metastatic melanoma cells to a hESC microenvironment. MATERIALS & METHODS: A total of 365 miRNAs were screened using the TaqMan® Low Density Arrays. We also evaluated whether DNA methylation could be one of the factors regulating the expression of the inhibitor of Nodal, Lefty, in hESCs (where it is highly expressed) vs melanoma cells (where it is not expressed). RESULTS: Using these experimental approaches, we identified miRNAs that are up- and down-regulated in melanoma cells exposed to a hESC microenvironment, such as miR-302a and miR-27b, respectively. We also demonstrate that Notch4 is one of the targets of miR-302a, which is upstream of Nodal. Additionally, one of the mechanisms that might explain the absence of the inhibitor of Nodal, Lefty, in cancer cells is silencing by DNA methylation, which provides new insights into the unregulated expression of Nodal in melanoma. CONCLUSION: These findings suggest that epigenetic changes such as DNA methylation and regulation by microRNAs might play a significant role in tumor cell plasticity and the metastatic phenotype.


Asunto(s)
Metilación de ADN/fisiología , Células Madre Embrionarias/metabolismo , Epigénesis Genética/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Melanoma/fisiopatología , Modelos Biológicos , Metástasis de la Neoplasia/fisiopatología , Western Blotting , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Biología Computacional , Islas de CpG/genética , Epigénesis Genética/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Factores de Determinación Derecha-Izquierda/metabolismo , Luciferasas , Melanoma/genética , MicroARNs/metabolismo , Metástasis de la Neoplasia/genética , Proteína Nodal/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN/métodos
15.
J Cell Biochem ; 101(4): 908-17, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17177292

RESUMEN

Aggressive cancer cells and pluripotent stem cells converge in their capacity for self-renewal, proliferation and plasticity. Recent studies have capitalized on these similarities by demonstrating that tumors arise from specific cancer stem cell populations that, in a manner reminiscent of normal stem cells, are able to both self-renew and give rise to a heterogeneous tumor population. This stem cell like function of aggressive cancer cells is likely attributable to the ectopic expression of embryonic factors such as Nodal and Cancer Testis Specific Antigens (CTAs), which maintain a functional plasticity by promoting pluripotency and immortality. During development, the expression of these embryonic factors is tightly regulated by a dynamic array of mediators, including the spatial and temporal expression of inhibitors such as Lefty, and the epigenetic modulation of the genome. In aggressive cancer cells, particularly melanoma, this balance of regulatory mediators is disrupted, leading to the aberrant expression of pluripotency-associated genes. By exposing aggressive cancer cells to embryonic microenvironments, this balance of regulatory mediators is restored, thereby reprogramming tumor cells to a more benign phenotype. These stem cell-derived mediators, as well as the genes they regulate, provide therapeutic targets designed to specifically differentiate and eradicate aggressive cancers.


Asunto(s)
Células Madre Embrionarias/citología , Neoplasias/patología , Células Madre Neoplásicas/patología , Células Madre Pluripotentes/patología , Linaje de la Célula , Células Madre Embrionarias/metabolismo , Humanos , Factores de Determinación Derecha-Izquierda , Modelos Biológicos , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Proteína Nodal , Células Madre Pluripotentes/metabolismo , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA