Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nucleic Acids Res ; 42(1): 249-63, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24097438

RESUMEN

Epstein-Barr virus (EBV) infects and transforms human primary B cells inducing indefinite proliferation. To investigate the potential participation of chromatin mechanisms during the EBV-mediated transformation of resting B cells we performed an analysis of global changes in histone modifications. We observed a remarkable decrease and redistribution of heterochromatin marks including H4K20me3, H3K27me3 and H3K9me3. Loss of H4K20me3 and H3K9me3 occurred at constitutive heterochromatin repeats. For H3K27me3 and H3K9me3, comparison of ChIP-seq data revealed a decrease in these marks in thousands of genes, including clusters of HOX and ZNF genes, respectively. Moreover, DNase-seq data comparison between resting and EBV-transformed B cells revealed increased endonuclease accessibility in thousands of genomic sites. We observed that both loss of H3K27me3 and increased accessibility are associated with transcriptional activation. These changes only occurred in B cells transformed with EBV and not in those stimulated to proliferate with CD40L/IL-4, despite their similarities in the cell pathways involved and proliferation rates. In fact, B cells infected with EBNA-2 deficient EBV, which have much lower proliferation rates, displayed similar decreases for heterochromatic histone marks. Our study describes a novel phenomenon related to transformation of B cells, and highlights its independence of the pure acquisition of proliferation.


Asunto(s)
Linfocitos B/virología , Herpesvirus Humano 4/fisiología , Heterocromatina/metabolismo , Transformación Genética , Linfocitos B/citología , Linfocitos B/metabolismo , Proliferación Celular , Histonas/metabolismo , Humanos
2.
Nucleic Acids Res ; 42(17): 11025-39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25200074

RESUMEN

MicroRNAs (miRNAs) have negative effects on gene expression and are major players in cell function in normal and pathological conditions. Epstein-Barr virus (EBV) infection of resting B lymphocytes results in their growth transformation and associates with different B cell lymphomas. EBV-mediated B cell transformation involves large changes in gene expression, including cellular miRNAs. We performed miRNA expression analysis in growth transformation of EBV-infected B cells. We observed predominant downregulation of miRNAs and upregulation of a few miRNAs. We observed similar profiles of miRNA expression in B cells stimulated with CD40L/IL-4, and those infected with EBNA-2- and LMP-1-deficient EBV particles, suggesting the implication of the NF-kB pathway, common to all four situations. In fact, the NF-kB subunit p65 associates with the transcription start site (TSS) of both upregulated and downregulated miRNAs following EBV infection This occurs together with changes at histone H3K27me3 and histone H3K4me3. Inhibition of the NF-kB pathway impairs changes in miRNA expression, NF-kB binding and changes at the above histone modifications near the TSS of these miRNA genes. Changes in expression of these miRNAs also occurred in diffuse large B cell lymphomas (DLBCL), which are strongly NF-kB dependent. Our results highlight the relevance of the NF-kB pathway in epigenetically mediated miRNA control in B cell transformation and DLBCL.


Asunto(s)
Linfocitos B/virología , Transformación Celular Viral/genética , Epigénesis Genética , Herpesvirus Humano 4/fisiología , Linfoma de Células B/virología , MicroARNs/metabolismo , FN-kappa B/metabolismo , Linfocitos B/metabolismo , Línea Celular Tumoral , Células Cultivadas , Humanos , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Transcripción Genética
3.
Genome Res ; 21(3): 422-32, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21324874

RESUMEN

Complex genomes utilize insulators and boundary elements to help define spatial and temporal gene expression patterns. We report that a genome-wide B1 SINE (Short Interspersed Nuclear Element) retrotransposon (B1-X35S) has potent intrinsic insulator activity in cultured cells and live animals. This insulation is mediated by binding of the transcription factors dioxin receptor (AHR) and SLUG (SNAI2) to consensus elements present in the SINE. Transcription of B1-X35S is required for insulation. While basal insulator activity is maintained by RNA polymerase (Pol) III transcription, AHR-induced insulation involves release of Pol III and engagement of Pol II transcription on the same strand. B1-X35S insulation is also associated with enrichment of heterochromatin marks H3K9me3 and H3K27me3 downstream of B1-X35S, an effect that varies with cell type. B1-X35S binds parylated CTCF and, consistent with a chromatin barrier activity, its positioning between two adjacent genes correlates with their differential expression in mouse tissues. Hence, B1 SINE retrotransposons represent genome-wide insulators activated by transcription factors that respond to developmental, oncogenic, or toxicological stimuli.


Asunto(s)
ARN Polimerasa III/metabolismo , ARN Polimerasa II/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Elementos de Nucleótido Esparcido Corto/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Adaptación Biológica , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Expresión Génica , Genes Reguladores , Marcadores Genéticos , Genoma , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Elementos Aisladores/genética , Ratones , Ratones Transgénicos , ARN Polimerasa II/genética , ARN Polimerasa III/genética , Receptores de Hidrocarburo de Aril/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Pez Cebra
4.
SLAS Discov ; 29(2): 100147, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38355016

RESUMEN

Pediatric brain tumors (PBTs) represent about 25 % of all pediatric cancers and are the most common solid tumors in children and adolescents. Medulloblastoma (MB) is the most frequently occurring malignant PBT, accounting for almost 10 % of all pediatric cancer deaths. MB Group 3 (MB G3) accounts for 25-30 % of all MB cases and has the worst outcome, particularly when associated with MYC amplification. However, no targeted treatments for this group have been developed so far. Here we describe a unique high throughput screening (HTS) platform specifically designed to identify new therapies for MB G3. The platform incorporates optimized and validated 2D and 3D efficacy and toxicity models, that account for tumor heterogenicity, limited efficacy and unacceptable toxicity from the very early stage of drug discovery. The platform has been validated by conducting a pilot HTS campaign with a 1280 lead-like compound library. Results showed 8 active compounds, targeting MB reported targets and several are currently approved or in clinical trials for pediatric patients with PBTs, including MB. Moreover, hits were combined to avoid tumor resistance, identifying 3 synergistic pairs, one of which is currently under clinical study for recurrent MB and other PBTs.


Asunto(s)
Neoplasias Encefálicas , Neoplasias Cerebelosas , Meduloblastoma , Humanos , Niño , Adolescente , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/genética , Meduloblastoma/patología , Ensayos Analíticos de Alto Rendimiento , Neoplasias Cerebelosas/tratamiento farmacológico , Neoplasias Cerebelosas/patología
5.
Sci Rep ; 7(1): 7594, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28790320

RESUMEN

Activation-induced cytidine deaminase (AID) triggers antibody diversification in B cells by catalysing deamination and subsequently mutating immunoglobulin (Ig) genes. Association of AID with RNA Pol II and occurrence of epigenetic changes during Ig gene diversification suggest participation of AID in epigenetic regulation. AID is mutated in hyper-IgM type 2 (HIGM2) syndrome. Here, we investigated the potential role of AID in the acquisition of epigenetic changes. We discovered that AID binding to the IgH locus promotes an increase in H4K20me3. In 293F cells, we demonstrate interaction between co-transfected AID and the three SUV4-20 histone H4K20 methyltransferases, and that SUV4-20H1.2, bound to the IgH switch (S) mu site, is replaced by SUV4-20H2 upon AID binding. Analysis of HIGM2 mutants shows that the AID truncated form W68X is impaired to interact with SUV4-20H1.2 and SUV4-20H2 and is unable to bind and target H4K20me3 to the Smu site. We finally show in mouse primary B cells undergoing class-switch recombination (CSR) that AID deficiency associates with decreased H4K20me3 levels at the Smu site. Our results provide a novel link between SUV4-20 enzymes and CSR and offer a new aspect of the interplay between AID and histone modifications in setting the epigenetic status of CSR sites.


Asunto(s)
Citidina Desaminasa/genética , Epigénesis Genética/inmunología , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Síndrome de Inmunodeficiencia con Hiper-IgM/genética , Cambio de Clase de Inmunoglobulina/genética , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/patología , Sitios de Unión , Línea Celular Tumoral , Citidina Desaminasa/inmunología , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , N-Metiltransferasa de Histona-Lisina/inmunología , Histonas/inmunología , Humanos , Síndrome de Inmunodeficiencia con Hiper-IgM/inmunología , Síndrome de Inmunodeficiencia con Hiper-IgM/patología , Inmunoglobulina G/genética , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación , Unión Proteica , ARN Polimerasa II/genética , ARN Polimerasa II/inmunología , Transducción de Señal
6.
Mol Cancer Ther ; 15(2): 287-98, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26590165

RESUMEN

Multiple myeloma is a plasma cell malignancy characterized by marked heterogeneous genomic instability including frequent genetic alterations in epigenetic enzymes. In particular, the histone methyltransferase Enhancer of Zeste Homolog 2 (EZH2) is overexpressed in multiple myeloma. EZH2 is the catalytic component of the polycomb repressive complex 2 (PRC2), a master transcriptional regulator of differentiation. EZH2 catalyzes methylation of lysine 27 on histone H3 and its deregulation in cancer has been reported to contribute to silencing of tumor suppressor genes, resulting in a more undifferentiated state, and thereby contributing to the multiple myeloma phenotype. In this study, we propose the use of EZH2 inhibitors as a new therapeutic approach for the treatment of multiple myeloma. We demonstrate that EZH2 inhibition causes a global reduction of H3K27me3 in multiple myeloma cells, promoting reexpression of EZH2-repressed tumor suppressor genes in a subset of cell lines. As a result of this transcriptional activation, multiple myeloma cells treated with EZH2 inhibitors become more adherent and less proliferative compared with untreated cells. The antitumor efficacy of EZH2 inhibitors is also confirmed in vivo in a multiple myeloma xenograft model in mice. Together, our data suggest that EZH2 inhibition may provide a new therapy for multiple myeloma treatment and a promising addition to current treatment options. Mol Cancer Ther; 15(2); 287-98. ©2015 AACR.


Asunto(s)
Inhibidores Enzimáticos/administración & dosificación , Histonas/metabolismo , Mieloma Múltiple/tratamiento farmacológico , Complejo Represivo Polycomb 2/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Línea Celular Tumoral , Proteína Potenciadora del Homólogo Zeste 2 , Inhibidores Enzimáticos/farmacología , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Mieloma Múltiple/enzimología , Mieloma Múltiple/genética , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Microambiente Tumoral/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Genome Biol ; 14(9): R99, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24028770

RESUMEN

BACKGROUND: DNA methylation is a key epigenetic mechanism for driving and stabilizing cell-fate decisions. Local deposition and removal of DNA methylation are tightly coupled with transcription factor binding, although the relationship varies with the specific differentiation process. Conversion of monocytes to osteoclasts is a unique terminal differentiation process within the hematopoietic system. This differentiation model is relevant to autoimmune disease and cancer, and there is abundant knowledge on the sets of transcription factors involved. RESULTS: Here we focused on DNA methylation changes during osteoclastogenesis. Hypermethylation and hypomethylation changes took place in several thousand genes, including all relevant osteoclast differentiation and function categories. Hypomethylation occurred in association with changes in 5-hydroxymethylcytosine, a proposed intermediate toward demethylation. Transcription factor binding motif analysis revealed an over-representation of PU.1, NF-κB, and AP-1 (Jun/Fos) binding motifs in genes undergoing DNA methylation changes. Among these, only PU.1 motifs were significantly enriched in both hypermethylated and hypomethylated genes; ChIP-seq data analysis confirmed its association to both gene sets. Moreover, PU.1 interacts with both DNMT3b and TET2, suggesting its participation in driving hypermethylation and hydroxymethylation-mediated hypomethylation. Consistent with this, siRNA-mediated PU.1 knockdown in primary monocytes impaired the acquisition of DNA methylation and expression changes, and reduced the association of TET2 and DNMT3b at PU.1 targets during osteoclast differentiation. CONCLUSIONS: The work described here identifies key changes in DNA methylation during monocyte-to-osteoclast differentiation and reveals novel roles for PU.1 in this process.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Monocitos/metabolismo , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , 5-Metilcitosina/análogos & derivados , Sitios de Unión , Diferenciación Celular , Islas de CpG , Citosina/análogos & derivados , Citosina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Proteínas de Unión al ADN/genética , Dioxigenasas , Humanos , Monocitos/citología , FN-kappa B/genética , FN-kappa B/metabolismo , Osteoclastos/citología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , ADN Metiltransferasa 3B
8.
Genome Biol ; 14(1): R3, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23320978

RESUMEN

BACKGROUND: Epstein-Barr virus (EBV) infection is a well characterized etiopathogenic factor for a variety of immune-related conditions, including lymphomas, lymphoproliferative disorders and autoimmune diseases. EBV-mediated transformation of resting B cells to proliferating lymphoblastoid cells occurs in early stages of infection and is an excellent model for investigating the mechanisms associated with acquisition of unlimited growth. RESULTS: We investigated the effects of experimental EBV infection of B cells on DNA methylation profiles by using high-throughput analysis. Remarkably, we observed hypomethylation of around 250 genes, but no hypermethylation. Hypomethylation did not occur at repetitive sequences, consistent with the absence of genomic instability in lymphoproliferative cells. Changes in methylation only occurred after cell divisions started, without the participation of the active demethylation machinery, and were concomitant with acquisition by B cells of the ability to proliferate. Gene Ontology analysis, expression profiling, and high-throughput analysis of the presence of transcription factor binding motifs and occupancy revealed that most genes undergoing hypomethylation are active and display the presence of NF-κB p65 and other B cell-specific transcription factors. Promoter hypomethylation was associated with upregulation of genes relevant for the phenotype of proliferating lymphoblasts. Interestingly, pharmacologically induced demethylation increased the efficiency of transformation of resting B cells to lymphoblastoid cells, consistent with productive cooperation between hypomethylation and lymphocyte proliferation. CONCLUSIONS: Our data provide novel clues on the role of the B cell transcription program leading to DNA methylation changes, which we find to be key to the EBV-associated conversion of resting B cells to proliferating lymphoblasts.


Asunto(s)
Linfocitos B/metabolismo , Metilación de ADN , Infecciones por Virus de Epstein-Barr/genética , Activación de Linfocitos , Transcripción Genética , Linfocitos B/inmunología , Linfocitos B/fisiología , Proliferación Celular , Infecciones por Virus de Epstein-Barr/metabolismo , Perfilación de la Expresión Génica , Humanos , Regulación hacia Arriba
9.
Eur J Cancer ; 48(14): 2270-81, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22244828

RESUMEN

Altered promoter DNA methylation, one of the most important molecular alterations in cancer, is proposed to correlate with deregulation of DNA methyltransferases, although the molecular mechanisms implicated are still poorly understood. Here we show that the de novo DNA methyltransferase DNMT3B is frequently repressed in human colorectal cancer cell lines (CCL) and primary tumours by aberrant DNA hypermethylation of its distal promoter. At the epigenome level, DNMT3B promoter hypermethylation was associated with the hypomethylation of gene promoters usually hypermethylated in the healthy colon. Forced DNMT3B overexpression in cancer cells restored the methylation levels of these promoters in the healthy colon. Our results show a new molecular mechanism of aberrant DNMT3B regulation in colon cancer and suggest that its expression is associated with the methylation of constitutively hypermethylated promoters in the healthy colon.


Asunto(s)
Neoplasias Colorrectales/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Regiones Promotoras Genéticas , Adulto , Anciano , Anciano de 80 o más Años , Azacitidina/análogos & derivados , Azacitidina/farmacología , Neoplasias Colorrectales/enzimología , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN/efectos de los fármacos , Decitabina , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Células HCT116 , Células HT29 , Humanos , Masculino , Persona de Mediana Edad , Regiones Promotoras Genéticas/efectos de los fármacos , Transfección , ADN Metiltransferasa 3B
10.
Discov Med ; 12(67): 535-45, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22204770

RESUMEN

The study of epigenetic mechanisms in the pathogenesis of autoimmune diseases is receiving unprecedented attention from clinicians and researchers in the field. Autoimmune disorders comprise a wide range of genetically complex diseases, including systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. Together they affect a significant proportion of the population and have a great economic impact on public health systems. Epigenetic mechanisms control gene expression and are influenced by external stimuli, linking environment and gene function. A variety of environmental agents, such as viral infection, hormones, certain drugs, and pollutants, have been found to influence the development of autoimmune diseases. On the other hand, there is considerable evidence of epigenetic changes, particularly DNA methylation alterations, in diseases like systemic lupus erythematosus, rheumatoid arthritis, or multiple sclerosis. However, the gap in our understanding between the specific effects of external agents and the influence on epigenetic profiles has not yet been filled. Here we review a number of studies describing epigenetic alterations in autoimmune diseases and a range of environmental factors that influence the development of autoimmune diseases. We also discuss potential mechanisms linking environment and epigenetics, consider the prospects for future epigenetic studies addressing the relationship between environment and epigenetics, and comment on the use of drugs with an epigenetic-reversing effect in the clinical management of these diseases.


Asunto(s)
Enfermedades Autoinmunes/genética , Ambiente , Epigénesis Genética , Interacción Gen-Ambiente , Animales , Autoinmunidad/genética , Investigación Biomédica , Humanos
11.
Epigenomics ; 3(6): 697-713, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22126290

RESUMEN

Proper immune function is the result of multiple cell commitment and differentiation steps, and adequate control of activation mechanisms. Deregulation of transcriptional programs in immune cells leads to the development of hematological malignancies, autoimmune diseases or immunodeficiencies. In this sense, epigenetic control of gene expression plays an essential role in the correct function of the immune system and the integrity of identity of relevant cell types. Epigenetic deregulation can result as a consequence of genetic changes in transcription factors, elements of signaling pathways or epigenetic enzymes, or as an effect of a variety of environmental factors. On top of genetic predisposition, viral infection and other external factors influence the development of immune-related diseases. In recent years, major strides have been made towards understanding the contribution of genetics in these immune disorders. Less progress has been made in dissecting the contribution of epigenetic factors in their etiology. Herein, it is presented what is currently known about epigenetic alterations in immune system associated disorders. It is also discussed how epigenomic analysis can help to understand the molecular basis of these diseases and how this information can be used in the clinical setting.


Asunto(s)
Cromatina/metabolismo , Metilación de ADN/fisiología , Epigénesis Genética/inmunología , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/fisiopatología , Modelos Inmunológicos , ADN (Citosina-5-)-Metiltransferasas/genética , Epigénesis Genética/fisiología , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Humanos , Enfermedades del Sistema Inmune/virología , Linfocitos/virología , Mutación/genética , ADN Metiltransferasa 3B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA