Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Nat Genet ; 53(10): 1456-1468, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34594038

RESUMEN

Glioma intratumoral heterogeneity enables adaptation to challenging microenvironments and contributes to therapeutic resistance. We integrated 914 single-cell DNA methylomes, 55,284 single-cell transcriptomes and bulk multi-omic profiles across 11 adult IDH mutant or IDH wild-type gliomas to delineate sources of intratumoral heterogeneity. We showed that local DNA methylation disorder is associated with cell-cell DNA methylation differences, is elevated in more aggressive tumors, links with transcriptional disruption and is altered during the environmental stress response. Glioma cells under in vitro hypoxic and irradiation stress increased local DNA methylation disorder and shifted cell states. We identified a positive association between genetic and epigenetic instability that was supported in bulk longitudinally collected DNA methylation data. Increased DNA methylation disorder associated with accelerated disease progression and recurrently selected DNA methylation changes were enriched for environmental stress response pathways. Our work identified an epigenetically facilitated adaptive stress response process and highlights the importance of epigenetic heterogeneity in shaping therapeutic outcomes.


Asunto(s)
Neoplasias Encefálicas/genética , Plasticidad de la Célula/genética , Epigénesis Genética , Glioma/genética , Análisis de la Célula Individual , Estrés Fisiológico/genética , Evolución Clonal , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Genoma Humano , Humanos , Mutación/genética , Filogenia , Regiones Promotoras Genéticas/genética , Microambiente Tumoral/genética
2.
Epigenetics ; 11(3): 184-93, 2016 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-26890396

RESUMEN

Small cell prostate carcinoma (SCPC) morphology is rare at initial diagnosis but often emerges during prostate cancer progression and portends a dismal prognosis. It does not express androgen receptor (AR) or respond to hormonal therapies. Clinically applicable markers for its early detection and treatment with effective chemotherapy are needed. Our studies in patient tumor-derived xenografts (PDX) revealed that AR-negative SCPC (AR(-)SCPC) expresses neural development genes instead of the prostate luminal epithelial genes characteristic of AR-positive castration-resistant adenocarcinomas (AR(+)ADENO). We hypothesized that the differences in cellular lineage programs are reflected in distinct epigenetic profiles. To address this hypothesis, we compared the DNA methylation profiles of AR(-) and AR(+) PDX using methylated CpG island amplification and microarray (MCAM) analysis and identified a set of differentially methylated promoters, validated in PDX and corresponding donor patient samples. We used the Illumina 450K platform to examine additional regions of the genome and the correlation between the DNA methylation profiles of the PDX and their corresponding patient tumors. Struck by the low frequency of AR promoter methylation in the AR(-)SCPC, we investigated this region's specific histone modification patterns by chromatin immunoprecipitation. We found that the AR promoter was enriched in silencing histone modifications (H3K27me3 and H3K9me2) and that EZH2 inhibition with 3-deazaneplanocin A (DZNep) resulted in AR expression and growth inhibition in AR(-)SCPC cell lines. We conclude that the epigenome of AR(-) is distinct from that of AR(+) castration-resistant prostate carcinomas, and that the AR(-) phenotype can be reversed with epigenetic drugs.


Asunto(s)
Carcinoma de Células Pequeñas/genética , Metilación de ADN/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Neoplasias de la Próstata Resistentes a la Castración/genética , Receptores Androgénicos/genética , Adenosina/administración & dosificación , Adenosina/análogos & derivados , Animales , Carcinoma de Células Pequeñas/patología , Línea Celular Tumoral , Linaje de la Célula/genética , Islas de CpG/genética , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Regiones Promotoras Genéticas , Neoplasias de la Próstata Resistentes a la Castración/patología , Receptores Androgénicos/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Neurosci ; 35(34): 12002-17, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26311780

RESUMEN

Lamin B1 is a component of the nuclear lamina and plays a critical role in maintaining nuclear architecture, regulating gene expression and modulating chromatin positioning. We have previously shown that LMNB1 gene duplications cause autosomal dominant leukodystrophy (ADLD), a fatal adult onset demyelinating disease. The mechanisms by which increased LMNB1 levels cause ADLD are unclear. To address this, we used a transgenic mouse model where Lamin B1 overexpression is targeted to oligodendrocytes. These mice showed severe vacuolar degeneration of the spinal cord white matter together with marked astrogliosis, microglial infiltration, and secondary axonal damage. Oligodendrocytes in the transgenic mice revealed alterations in histone modifications favoring a transcriptionally repressed state. Chromatin changes were accompanied by reduced expression of genes involved in lipid synthesis pathways, many of which are known to play important roles in myelin regulation and are preferentially expressed in oligodendrocytes. Decreased lipogenic gene expression resulted in a significant reduction in multiple classes of lipids involved in myelin formation. Many of these gene expression changes and lipid alterations were observed even before the onset of the phenotype, suggesting a causal role. Our findings establish, for the first time, a link between LMNB1 and lipid synthesis in oligodendrocytes, and provide a mechanistic framework to explain the age dependence and white matter involvement of the disease phenotype. These results have implications for disease pathogenesis and may also shed light on the regulation of lipid synthesis pathways in myelin maintenance and turnover. SIGNIFICANCE STATEMENT: Autosomal dominant leukodystrophy (ADLD) is fatal neurological disorder caused by increased levels of the nuclear protein, Lamin B1. The disease is characterized by an age-dependent loss of myelin, the fatty sheath that covers nerve fibers. We have studied a mouse model where Lamin B1 level are increased in oligodendrocytes, the cell type that produces myelin in the CNS. We demonstrate that destruction of myelin in the spinal cord is responsible for the degenerative phenotype in our mouse model. We show that this degeneration is mediated by reduced expression of lipid synthesis genes and the subsequent reduction in myelin enriched lipids. These findings provide a mechanistic framework to explain the age dependence and tissue specificity of the ADLD disease phenotype.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Desmielinizantes/metabolismo , Lamina Tipo B/biosíntesis , Metabolismo de los Lípidos/fisiología , Envejecimiento/genética , Animales , Enfermedades Desmielinizantes/genética , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Lámina Nuclear/genética , Lámina Nuclear/metabolismo , Oligodendroglía/metabolismo
4.
Contemp Oncol (Pozn) ; 19(1A): A30-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25691819

RESUMEN

Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) requires profound alterations in the epigenetic landscape. During reprogramming, a change in chromatin structure resets the gene expression and stabilises self-renewal. Reprogramming is a highly inefficient process, in part due to multiple epigenetic barriers. Although many epigenetic factors have already been shown to affect self-renewal and pluripotency in embryonic stem cells (ESCs), only a few of them have been examined in the context of dedifferentiation. In order to improve current protocols of iPSCs generation, it is essential to identify epigenetic drivers and blockages of somatic cell reprogramming.

5.
Br J Haematol ; 169(3): 344-51, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25612675

RESUMEN

Correlative and functional studies support the involvement of the RUNX gene family in haematological malignancies. To elucidate the role of epigenetics in RUNX inactivation, we evaluated promoter DNA methylation of RUNX1, 2, and 3 in 23 leukaemia cell lines and samples from acute myeloid leukaemia (AML), acute lymphocytic leukaemia (ALL) and myelodysplatic syndromes (MDS) patients. RUNX1 and RUNX2 gene promoters were mostly unmethylated in cell lines and clinical samples. Hypermethylation of RUNX3 was frequent among cell lines (74%) and highly variable among patient samples, with clear association to cytogenetic status. High frequency of RUNX3 hypermethylation (85% of the 20 studied cases) was found in AML patients with inv(16)(p13.1q22) compared to other AML subtypes (31% of the other 49 cases). RUNX3 hypermethylation was also frequent in ALL (100% of the six cases) but low in MDS (21%). In support of a functional role, hypermethylation of RUNX3 was correlated with low levels of protein, and treatment of cell lines with the DNA demethylating agent, decitabine, resulted in mRNA re-expression. Furthermore, relapse-free survival of non-inv(16)(p13.1q22) AML patients without RUNX3 methylation was significantly better (P = 0·016) than that of methylated cases. These results suggest that RUNX3 silencing is an important event in inv(16)(p13.1q22) leukaemias.


Asunto(s)
Inversión Cromosómica , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Metilación de ADN , Leucemia Mieloide Aguda/genética , Regiones Promotoras Genéticas , Adulto , Anciano , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Decitabina , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/mortalidad , Persona de Mediana Edad , Activación Transcripcional/efectos de los fármacos
6.
Nucleic Acids Res ; 42(11): 6956-71, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24875481

RESUMEN

TET1 is a 5-methylcytosine dioxygenase and its DNA demethylating activity has been implicated in pluripotency and reprogramming. However, the precise role of TET1 in DNA methylation regulation outside of developmental reprogramming is still unclear. Here, we show that overexpression of the TET1 catalytic domain but not full length TET1 (TET1-FL) induces massive global DNA demethylation in differentiated cells. Genome-wide mapping reveals that 5-hydroxymethylcytosine production by TET1-FL is inhibited as DNA methylation increases, which can be explained by the preferential binding of TET1-FL to unmethylated CpG islands (CGIs) through its CXXC domain. TET1-FL specifically accumulates 5-hydroxymethylcytosine at the edges of hypomethylated CGIs, while knockdown of endogenous TET1 induces methylation spreading from methylated edges into hypomethylated CGIs. We also found that gene expression changes after TET1-FL overexpression are relatively small and independent of its dioxygenase function. Thus, our results identify TET1 as a maintenance DNA demethylase that does not purposely decrease methylation levels, but specifically prevents aberrant methylation spreading into CGIs in differentiated cells.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , 5-Metilcitosina/análogos & derivados , Dominio Catalítico , Diferenciación Celular/genética , Islas de CpG , Citosina/análogos & derivados , Citosina/análisis , Citosina/metabolismo , Proteínas de Unión al ADN/química , Dioxigenasas/química , Células HEK293 , Humanos , Oxigenasas de Función Mixta , Proteínas Proto-Oncogénicas/química , Transcripción Genética
7.
Cancer Res ; 74(5): 1311-8, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24385213

RESUMEN

Fusobacterium species are part of the gut microbiome in humans. Recent studies have identified overrepresentation of Fusobacterium in colorectal cancer tissues, but it is not yet clear whether this is pathogenic or simply an epiphenomenon. In this study, we evaluated the relationship between Fusobacterium status and molecular features in colorectal cancers through quantitative real-time PCR in 149 colorectal cancer tissues, 89 adjacent normal appearing mucosae and 72 colonic mucosae from cancer-free individuals. Results were correlated with CpG island methylator phenotype (CIMP) status, microsatellite instability (MSI), and mutations in BRAF, KRAS, TP53, CHD7, and CHD8. Whole-exome capture sequencing data were also available in 11 cases. Fusobacterium was detectable in 111 of 149 (74%) colorectal cancer tissues and heavily enriched in 9% (14/149) of the cases. As expected, Fusobacterium was also detected in normal appearing mucosae from both cancer and cancer-free individuals, but the amount of bacteria was much lower compared with colorectal cancer tissues (a mean of 250-fold lower for Pan-fusobacterium). We found the Fusobacterium-high colorectal cancer group (FB-high) to be associated with CIMP positivity (P = 0.001), TP53 wild-type (P = 0.015), hMLH1 methylation positivity (P = 0.0028), MSI (P = 0.018), and CHD7/8 mutation positivity (P = 0.002). Among the 11 cases where whole-exome sequencing data were available, two that were FB-high cases also had the highest number of somatic mutations (a mean of 736 per case in FB-high vs. 225 per case in all others). Taken together, our findings show that Fusobacterium enrichment is associated with specific molecular subsets of colorectal cancers, offering support for a pathogenic role in colorectal cancer for this gut microbiome component.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Fusobacterium/genética , Anciano , Islas de CpG/genética , ADN Helicasas/genética , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Infecciones por Fusobacterium/genética , Infecciones por Fusobacterium/microbiología , Infecciones por Fusobacterium/patología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Inestabilidad de Microsatélites , Persona de Mediana Edad , Membrana Mucosa/microbiología , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Proteínas ras/genética
8.
Genome Res ; 24(4): 580-91, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24414704

RESUMEN

The myelodysplastic syndrome (MDS) is a clonal hematologic disorder that frequently evolves to acute myeloid leukemia (AML). Its pathogenesis remains unclear, but mutations in epigenetic modifiers are common and the disease often responds to DNA methylation inhibitors. We analyzed DNA methylation in the bone marrow and spleen in two mouse models of MDS/AML, the NUP98-HOXD13 (NHD13) mouse and the RUNX1 mutant mouse model. Methylation array analysis showed an average of 512/3445 (14.9%) genes hypermethylated in NHD13 MDS, and 331 (9.6%) genes hypermethylated in RUNX1 MDS. Thirty-two percent of genes in common between the two models (2/3 NHD13 mice and 2/3 RUNX1 mice) were also hypermethylated in at least two of 19 human MDS samples. Detailed analysis of 41 genes in mice showed progressive drift in DNA methylation from young to old normal bone marrow and spleen; to MDS, where we detected accelerated age-related methylation; and finally to AML, which markedly extends DNA methylation abnormalities. Most of these genes showed similar patterns in human MDS and AML. Repeat element hypomethylation was rare in MDS but marked the transition to AML in some cases. Our data show consistency in patterns of aberrant DNA methylation in human and mouse MDS and suggest that epigenetically, MDS displays an accelerated aging phenotype.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Animales , Células de la Médula Ósea , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Humanos , Leucemia Mieloide Aguda/patología , Ratones , Síndromes Mielodisplásicos/patología , Proteínas de Complejo Poro Nuclear/genética
9.
Gastroenterology ; 146(2): 530-38.e5, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24211491

RESUMEN

BACKGROUND & AIMS: Subgroups of colorectal carcinomas (CRCs) characterized by DNA methylation anomalies are termed CpG island methylator phenotype (CIMP)1, CIMP2, or CIMP-negative. The pathogenesis of CIMP1 colorectal carcinomas, and their effects on patients' prognoses and responses to treatment, differ from those of other CRCs. We sought to identify genetic somatic alterations associated with CIMP1 CRCs. METHODS: We examined genomic DNA samples from 100 primary CRCs, 10 adenomas, and adjacent normal-appearing mucosae from patients undergoing surgery or colonoscopy at 3 tertiary medical centers. We performed exome sequencing of 16 colorectal tumors and their adjacent normal tissues. Extensive comparison with known somatic alterations in CRCs allowed segregation of CIMP1-exclusive alterations. The prevalence of mutations in selected genes was determined from an independent cohort. RESULTS: We found that genes that regulate chromatin were mutated in CIMP1 CRCs; the highest rates of mutation were observed in CHD7 and CHD8, which encode members of the chromodomain helicase/adenosine triphosphate-dependent chromatin remodeling family. Somatic mutations in these 2 genes were detected in 5 of 9 CIMP1 CRCs. A prevalence screen showed that nonsilencing mutations in CHD7 and CHD8 occurred significantly more frequently in CIMP1 tumors (18 of 42 [43%]) than in CIMP2 (3 of 34 [9%]; P < .01) or CIMP-negative tumors (2 of 34 [6%]; P < .001). CIMP1 markers had increased binding by CHD7, compared with all genes. Genes altered in patients with CHARGE syndrome (congenital malformations involving the central nervous system, eye, ear, nose, and mediastinal organs) who had CHD7 mutations were also altered in CRCs with mutations in CHD7. CONCLUSIONS: Aberrations in chromatin remodeling could contribute to the development of CIMP1 CRCs. A better understanding of the biological determinants of CRCs can be achieved when these tumors are categorized according to their epigenetic status.


Asunto(s)
Cromatina , Neoplasias Colorrectales/genética , Islas de CpG , ADN Helicasas/genética , Metilación de ADN , Proteínas de Unión al ADN/genética , Mutación , Factores de Transcripción/genética , Adenoma/genética , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Exoma , Femenino , Silenciador del Gen , Marcadores Genéticos , Humanos , Masculino , Inestabilidad de Microsatélites , Persona de Mediana Edad , Fenotipo , Análisis de Secuencia de ADN
10.
Cancer Prev Res (Phila) ; 6(10): 1093-100, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23943784

RESUMEN

Whole blood DNA methylation analysis has been proposed to be a risk marker for cancer that can be used to target patients for preventive interventions. To test this, we examined whole blood DNA methylation of 16 CpG island promoters and LINE1 repetitive element in patients with gastric cancer and control subjects. Bisulfite pyrosequencing was used to quantify the methylation of 14 CpG island promoters (MINT25, RORA, GDNF, CDH1, RARAB2, ER, CDH13, MYOD1, SFRP1, P2RX7, SLC16A12, IGF2, DPYS, and N33) and LINE1 from 72 patients with gastric cancer, 67 control, and 52 healthy young individuals. Quantitative methylation-specific real-time PCR was also conducted for 3 CpG island promoters (MINT25, MYO3A, and SOX11). Among all sites tested, only a marginal increase in the methylation of the SFRP1 promoter was observed in the blood of patients with gastric cancer when compared with the control group (11.3 % vs 10.5%; age-adjusted P value: P = 0.009), and this association was also seen in a validation set of 91 patients with gastric cancer (11.5% vs 10.5%; age-adjusted P value: P = 0.001). The methylation of 9 sites (GDNF, CDH1, RARAB2, CDH13, MYOD1, SFRP1, SLC16A12, DPYS, N33, and LINE1) and their mean Z score was correlated with higher age (R = 0.41, P < 0.0001) and marginally with telomere shortening (R = -0.18, P = 0.01) but not with gastric cancer risk (other than SFRP1 methylation). Variability in whole blood DNA methylation of cancer markers is primarily associated with aging, reflecting turnover of white blood cells, and has no direct link to gastric cancer predisposition. SFRP1 methylation in whole blood may be associated with gastric cancer risk.


Asunto(s)
Biomarcadores de Tumor/sangre , Metilación de ADN , ADN/sangre , Neoplasias Gástricas/genética , Adulto , Anciano , Estudios de Casos y Controles , Islas de CpG , Femenino , Humanos , Masculino , Persona de Mediana Edad , Riesgo , Análisis de Secuencia de ADN , Neoplasias Gástricas/metabolismo , Telómero/ultraestructura , Adulto Joven
11.
Epigenetics ; 7(12): 1368-78, 2012 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23075513

RESUMEN

Genome wide analysis of DNA methylation provides important information in a variety of diseases, including cancer. Here, we describe a simple method, Digital Restriction Enzyme Analysis of Methylation (DREAM), based on next generation sequencing analysis of methylation-specific signatures created by sequential digestion of genomic DNA with SmaI and XmaI enzymes. DREAM provides information on 150,000 unique CpG sites, of which 39,000 are in CpG islands and 30,000 are at transcription start sites of 13,000 RefSeq genes. We analyzed DNA methylation in healthy white blood cells and found methylation patterns to be remarkably uniform. Inter individual differences > 30% were observed only at 227 of 28,331 (0.8%) of autosomal CpG sites. Similarly, > 30% differences were observed at only 59 sites when we comparing the cord and adult blood. These conserved methylation patterns contrasted with extensive changes affecting 18-40% of CpG sites in a patient with acute myeloid leukemia and in two leukemia cell lines. The method is cost effective, quantitative (r ( 2) = 0.93 when compared with bisulfite pyrosequencing) and reproducible (r ( 2) = 0.997). Using 100-fold coverage, DREAM can detect differences in methylation greater than 10% or 30% with a false positive rate below 0.05 or 0.001, respectively. DREAM can be useful in quantifying epigenetic effects of environment and nutrition, correlating developmental epigenetic variation with phenotypes, understanding epigenetics of cancer and chronic diseases, measuring the effects of drugs on DNA methylation or deriving new biological insights into mammalian genomes.


Asunto(s)
Metilación de ADN , Leucemia/genética , Leucocitos/fisiología , Análisis de Secuencia de ADN/métodos , Secuencia de Bases , Línea Celular Tumoral , Cromosomas Humanos X , Secuencia Conservada , Islas de CpG , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Femenino , Impresión Genómica , Humanos , Leucemia/patología , Leucemia Mieloide Aguda/genética , Masculino , Valores de Referencia , Secuencias Repetitivas de Ácidos Nucleicos , Análisis de Secuencia de ADN/economía
12.
Mol Cancer Res ; 10(10): 1332-42, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22952045

RESUMEN

Almost half of the human genome and as much as 40% of the mouse genome is composed of repetitive DNA sequences. The majority of these repeats are retrotransposons of the SINE and LINE families, and such repeats are generally repressed by epigenetic mechanisms. It has been proposed that these elements can act as methylation centers from which DNA methylation spreads into gene promoters in cancer. Contradictory to a methylation center function, we have found that retrotransposons are enriched near promoter CpG islands that stay methylation-free in cancer. Clearly, it is important to determine which influence, if any, these repetitive elements have on nearby gene promoters. Using an in vitro system, we confirm here that SINE B1 elements can influence the activity of downstream gene promoters, with acquisition of DNA methylation and loss of activating histone marks, thus resulting in a repressed state. SINE sequences themselves did not immediately acquire DNA methylation but were marked by H3K9me2 and H3K27me3. Moreover, our bisulfite sequencing data did not support that gain of DNA methylation in gene promoters occurred by methylation spreading from SINE B1 repeats. Genome-wide analysis of SINE repeats distribution showed that their enrichment is directly correlated with the presence of USF1, USF2, and CTCF binding, proteins with insulator function. In summary, our work supports the concept that SINE repeats interfere negatively with gene expression and that their presence near gene promoters is counter-selected, except when the promoter is protected by an insulator element.


Asunto(s)
Reprogramación Celular/genética , Epigénesis Genética , Genes/genética , Regiones Promotoras Genéticas , Elementos de Nucleótido Esparcido Corto/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Elementos Alu/genética , Animales , Antígenos CD , Cadherinas/genética , Cromatina/metabolismo , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/genética , Metilación de ADN/genética , Silenciador del Gen , Humanos , Elementos Aisladores/genética , Elementos de Nucleótido Esparcido Largo/genética , Ratones , Homólogo 1 de la Proteína MutL , Células 3T3 NIH , Proteínas Nucleares/genética , Unión Proteica/genética , Sitio de Iniciación de la Transcripción , Transcripción Genética , Proteína p14ARF Supresora de Tumor/genética
13.
Nucleic Acids Res ; 40(15): 7257-68, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22600741

RESUMEN

Repression of many tumor suppressor genes in cancer is concurrent with aberrantly increased DNA methylation levels at promoter CpG islands (CGIs). About one-fourth of empirically defined human promoters are surrounded by or contain clustered repetitive elements. It was previously observed that a sharp transition of methylation exists between highly methylated repetitive elements and unmethylated promoter-CGIs in normal tissues. The factors that lead to aberrant CGI hypermethylation in cancer remain poorly understood. Here, we established a site-specific integration system with enforced local transcriptional repression in colorectal cancer cells and monitored the occurrence of initial de novo methylation at specific CG sites adjacent to the CGI of the INSL6 promoter, which could be accelerated by binding a KRAB-containing transcriptional factor. Additional repetitive elements from P16 and RIL (PDLIM4), if situated adjacent to the promoter of INSL6, could confer DNA methylation spreading into the CGI particularly in the setting of KRAB-factor binding. However, a repressive chromatin alone was not sufficient to initiate DNA methylation, which required specific DNA sequences and was integration-site (and/or cell-line) specific. Overall, these results demonstrate a requirement for specific DNA sequences to trigger de novo DNA methylation, and repetitive elements as cis-regulatory factors to cooperate with advanced transcriptional repression in promoting methylation spreading.


Asunto(s)
Islas de CpG , Metilación de ADN , Silenciador del Gen , Regiones Promotoras Genéticas , Secuencias Repetitivas de Ácidos Nucleicos , Transcripción Genética , Línea Celular Tumoral , Cromatina/metabolismo , ADN/química , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Transgenes
14.
Cancer Res ; 72(5): 1170-81, 2012 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22219169

RESUMEN

DNA methylation is commonly thought of as a "molecular lock" that leads to permanent gene silencing. To investigate this notion, we tested 24 different histone deacetylase inhibitors (HDACi) on colon cancer cells that harbor a GFP locus stably integrated and silenced by a hypermethylated cytomegalovirus (CMV) promoter. We found that HDACi efficiently reactivated expression of GFP and many other endogenous genes silenced by DNA hypermethylation. After treatment, all promoters were marked with active chromatin, yet DNA hypermethylation did not change. Thus, DNA methylation could not prevent gene reactivation by drug-induced resetting of the chromatin state. In evaluating the relative contribution of DNA methylation and histone modifications to stable gene silencing, we followed expression levels of GFP and other genes silenced by DNA hypermethylation over time after treatment with HDACi or DNA-demethylating drugs. Reactivation of methylated loci by HDACi was detectable for only 2 weeks, whereas DNA-demethylating drugs induced permanent epigenetic reprogramming. Therefore, DNA methylation cannot be considered as a lock for gene expression but rather as a memory signal for long-term maintenance of gene silencing. These findings define chromatin as an important druggable target for cancer epigenetic therapy and suggest that removal of DNA methylation signals is required to achieve long-term gene reactivation.


Asunto(s)
Neoplasias del Colon/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Metilación de ADN/efectos de los fármacos , Depsipéptidos/farmacología , Proteínas Fluorescentes Verdes/genética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Regiones Promotoras Genéticas , Activación Transcripcional/efectos de los fármacos
15.
PLoS One ; 6(8): e23320, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21853109

RESUMEN

BACKGROUND: MLL3 is a histone 3-lysine 4 methyltransferase with tumor-suppressor properties that belongs to a family of chromatin regulator genes potentially altered in neoplasia. Mutations in MLL3 were found in a whole genome analysis of colorectal cancer but have not been confirmed by a separate study. METHODS AND RESULTS: We analyzed mutations of coding region and promoter methylation in MLL3 using 126 cases of colorectal cancer. We found two isoforms of MLL3 and DNA sequencing revealed frameshift and other mutations affecting both isoforms of MLL3 in colorectal cancer cells and 19 of 134 (14%) primary colorectal samples analyzed. Moreover, frameshift mutations were more common in cases with microsatellite instability (31%) both in CRC cell lines and primary tumors. The largest isoform of MLL3 is transcribed from a CpG island-associated promoter that has highly homology with a pseudo-gene on chromosome 22 (psiTPTE22). Using an assay which measured both loci simultaneously we found prominent age related methylation in normal colon (from 21% in individuals less than 25 years old to 56% in individuals older than 70, R = 0.88, p<0.001) and frequent hypermethylation (83%) in both CRC cell lines and primary tumors. We next studied the two loci separately and found that age and cancer related methylation was solely a property of the pseudogene CpG island and that the MLL3 loci was unmethylated. CONCLUSIONS: We found that frameshift mutations of MLL3 in both CRC cells and primary tumor that were more common in cases with microsatellite instability. Moreover, we have shown CpG island-associated promoter of MLL3 gene has no DNA methylation in CRC cells but also primary tumor and normal colon, and this region has a highly homologous of pseudo gene (psiTPTE22) that was age relate DNA methylation.


Asunto(s)
Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/genética , Mutación del Sistema de Lectura/genética , Repeticiones de Microsatélite/genética , Secuencia de Bases , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Metilación de ADN/genética , Análisis Mutacional de ADN , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genoma Humano/genética , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Reproducibilidad de los Resultados
16.
PLoS One ; 6(7): e22110, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21760961

RESUMEN

The epigenetic impact of DNA methylation in chronic myelogenous leukemia (CML) is not completely understood. To elucidate its role we analyzed 120 patients with CML for methylation of promoter-associated CpG islands of 10 genes. Five genes were identified by DNA methylation screening in the K562 cell line and 3 genes in patients with myeloproliferative neoplasms. The CDKN2B gene was selected for its frequent methylation in myeloid malignancies and ABL1 as the target of BCR-ABL translocation. Thirty patients were imatinib-naïve (mostly treated by interferon-alpha before the imatinib era), 30 were imatinib-responsive, 50 were imatinib-resistant, and 10 were imatinib-intolerant. We quantified DNA methylation by bisulfite pyrosequencing. The average number of methylated genes was 4.5 per patient in the chronic phase, increasing significantly to 6.2 in the accelerated and 6.4 in the blastic phase. Higher numbers of methylated genes were also observed in patients resistant or intolerant to imatinib. These patients also showed almost exclusive methylation of a putative transporter OSCP1. Abnormal methylation of a Src suppressor gene PDLIM4 was associated with shortened survival independently of CML stage and imatinib responsiveness. We conclude that aberrant DNA methylation is associated with CML progression and that DNA methylation could be a marker associated with imatinib resistance. Finally, DNA methylation of PDLIM4 may help identify a subset of CML patients that would benefit from treatment with Src/Abl inhibitors.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Piperazinas/farmacología , Pirimidinas/farmacología , Adolescente , Adulto , Anciano , Benzamidas , Línea Celular Tumoral , Femenino , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Genes Relacionados con las Neoplasias/genética , Genoma Humano/genética , Humanos , Mesilato de Imatinib , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Análisis Multivariante , Estadificación de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Piperazinas/efectos adversos , Piperazinas/uso terapéutico , Pirimidinas/efectos adversos , Pirimidinas/uso terapéutico , Análisis de Secuencia de ADN , Análisis de Supervivencia , Resultado del Tratamiento , Adulto Joven
17.
FEBS Lett ; 585(13): 2078-86, 2011 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-21146531

RESUMEN

There is compelling evidence to support the importance of DNA methylation alterations in cancer development. Both losses and gains of DNA methylation are observed, thought to contribute pathophysiologically by inactivating tumor suppressor genes, inducing chromosomal instability and ectopically activating gene expression. Lesser known are the causes of aberrant DNA methylation. Recent studies have pointed out that intrinsic gene susceptibility to DNA methylation, environmental factors and gene function all have an intertwined participation in this process. Overall, these data support a deterministic rather than a stochastic mechanism for de novo DNA methylation in cancer. In this review article, we discuss the technologies available to study DNA methylation and the endogenous and exogenous factors that influence the onset of de novo methylation in cancer.


Asunto(s)
Metilación de ADN/genética , Neoplasias/genética , Animales , Humanos
18.
Genome Res ; 20(10): 1369-82, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20716667

RESUMEN

Epigenetic silencing plays an important role in cancer development. An attractive hypothesis is that local DNA features may participate in differential predisposition to gene hypermethylation. We found that, compared with methylation-resistant genes, methylation-prone genes have a lower frequency of SINE and LINE retrotransposons near their transcription start site. In several large testing sets, this distribution was highly predictive of promoter methylation. Genome-wide analysis showed that 22% of human genes were predicted to be methylation-prone in cancer; these tended to be genes that are down-regulated in cancer and that function in developmental processes. Moreover, retrotransposon distribution marks a larger fraction of methylation-prone genes compared to Polycomb group protein (PcG) marking in embryonic stem cells; indeed, PcG marking and our predictive model based on retrotransposon frequency appear to be correlated but also complementary. In summary, our data indicate that retrotransposon elements, which are widespread in our genome, are strongly associated with gene promoter DNA methylation in cancer and may in fact play a role in influencing epigenetic regulation in normal and abnormal physiological states.


Asunto(s)
Metilación de ADN , Neoplasias/genética , Retroelementos/genética , Línea Celular Tumoral , Epigenómica , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genoma Humano , Humanos , Leucemia Mieloide Aguda , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria
19.
Melanoma Res ; 19(3): 146-55, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19441164

RESUMEN

A better understanding of key molecular changes during the pathogenesis of melanoma could impact strategies to reduce mortality from this cancer. Two epigenetic events involved in the pathogenesis of cancer are hypermethylation of tumor-suppressor gene promoters associated with transcriptional repression and hypomethylation associated with gene reexpression and genomic instability. We analyzed 16 melanoma cell lines for aberrant hypermethylation of 15 cancer-linked genes (ER alpha, MGMT, RAR beta 2, RIL, RASSF1A, PAX7, PGR beta, PAX2, NKX2-3, OLIG2, HAND1, ECAD, CDH13, MLH1, and p16) and hypomethylation of two genes (MAGEA1, maspin) and two repetitive sequences (LINE-1 and Alu) using pyrosequencing. We observed hypermethylation of ER alpha in 50% of the cell lines, MGMT (50%), RAR beta 2 (44%), RIL (88%), RASSF1A (69%), PAX7 (31%), PGR beta (56%), PAX2 (38%), NKX2-3 (63%), OLIG2 (63%), HAND1 (63%), ECAD (88%), CDH13 (44%), MLH1 (0%), and p16 (6%). In human melanoma cell lines, hypomethylation of MAGEA1 (44%), maspin (25%), LINE-1 (75%), and Alu (13%) is frequently observed. We analyzed a panel of cell lines for BRAF V600E and NRAS codon 61 mutations. In melanoma cell lines, the BRAF and NRAS mutations had no association with aberrant methylation. We found that the cumulative aberrant hypermethylation of the gene promoters was correlated with the level of global DNA methylation. We conclude that aberrant hypermethylation, is frequent in melanoma cell lines, directly correlated with global DNA methylation, and independent of BRAF and NRAS mutations.


Asunto(s)
Islas de CpG/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Neoplasias Cutáneas/genética , Línea Celular Tumoral , Epigénesis Genética , Perfilación de la Expresión Génica , Frecuencia de los Genes , Humanos , Mutación/genética , Regiones Promotoras Genéticas
20.
Gastroenterology ; 136(7): 2149-58, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19375421

RESUMEN

BACKGROUND & AIMS: Aberrant DNA methylation is an early and frequent process in gastric carcinogenesis and could be useful for detection of gastric neoplasia. We hypothesized that methylation analysis of DNA recovered from gastric washes could be used to detect gastric cancer. METHODS: We studied 51 candidate genes in 7 gastric cancer cell lines and 24 samples (training set) and identified 6 for further studies. We examined the methylation status of these genes in a test set consisting of 131 gastric neoplasias at various stages. Finally, we validated the 6 candidate genes in a different population of 40 primary gastric cancer samples and 113 nonneoplastic gastric mucosa samples. RESULTS: Six genes (MINT25, RORA, GDNF, ADAM23, PRDM5, MLF1) showed frequent differential methylation between gastric cancer and normal mucosa in the training, test, and validation sets. GDNF and MINT25 were most sensitive molecular markers of early stage gastric cancer, whereas PRDM5 and MLF1 were markers of a field defect. There was a close correlation (r = 0.5-0.9, P = .03-.001) between methylation levels in tumor biopsy and gastric washes. MINT25 methylation had the best sensitivity (90%), specificity (96%), and area under the receiver operating characteristic curve (0.961) in terms of tumor detection in gastric washes. CONCLUSIONS: These findings suggest MINT25 is a sensitive and specific marker for screening in gastric cancer. Additionally, we have developed a new method for gastric cancer detection by DNA methylation in gastric washes.


Asunto(s)
Metilación de ADN , Predisposición Genética a la Enfermedad , Lesiones Precancerosas/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteínas Supresoras de Tumor/genética , Anciano , Análisis de Varianza , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Línea Celular Tumoral , ADN de Neoplasias/análisis , Detección Precoz del Cáncer , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Lesiones Precancerosas/patología , Probabilidad , Curva ROC , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...