Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Clin Epigenetics ; 15(1): 102, 2023 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-37309009

RESUMEN

BACKGROUND: Epigenetic alterations are a near-universal feature of human malignancy and have been detected in malignant cells as well as in easily accessible specimens such as blood and urine. These findings offer promising applications in cancer detection, subtyping, and treatment monitoring. However, much of the current evidence is based on findings in retrospective studies and may reflect epigenetic patterns that have already been influenced by the onset of the disease. METHODS: Studying breast cancer, we established genome-scale DNA methylation profiles of prospectively collected buffy coat samples (n = 702) from a case-control study nested within the EPIC-Heidelberg cohort using reduced representation bisulphite sequencing (RRBS). RESULTS: We observed cancer-specific DNA methylation events in buffy coat samples. Increased DNA methylation in genomic regions associated with SURF6 and REXO1/CTB31O20.3 was linked to the length of time to diagnosis in the prospectively collected buffy coat DNA from individuals who subsequently developed breast cancer. Using machine learning methods, we piloted a DNA methylation-based classifier that predicted case-control status in a held-out validation set with 76.5% accuracy, in some cases up to 15 years before clinical diagnosis of the disease. CONCLUSIONS: Taken together, our findings suggest a model of gradual accumulation of cancer-associated DNA methylation patterns in peripheral blood, which may be detected long before clinical manifestation of cancer. Such changes may provide useful markers for risk stratification and, ultimately, personalized cancer prevention.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Estudios de Casos y Controles , Estudios Prospectivos , Estudios Retrospectivos , Metilación de ADN , Proteínas Nucleares
3.
Epigenetics ; 17(13): 2434-2454, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36354000

RESUMEN

Illumina Infinium DNA Methylation (5mC) arrays are a popular technology for low-cost, high-throughput, genome-scale measurement of 5mC distribution, especially in cancer and other complex diseases. After the success of its HumanMethylation450 array (450k), Illumina released the MethylationEPIC array (850k) featuring increased coverage of enhancers. Despite the widespread use of 850k, analysis of the corresponding data remains suboptimal: it still relies mostly on Illumina's default annotation, which underestimates enhancerss and long noncoding RNAs. Results: We have thus developed an approach, based on the ENCODE and LNCipedia databases, which greatly improves upon Illumina's default annotation of enhancers and long noncoding transcripts. We compared the re-annotated 850k with both 450k and reduced-representation bisulphite sequencing (RRBS), another high-throughput 5mC profiling technology. We found 850k to cover at least three times as many enhancers and long noncoding RNAs as either 450k or RRBS. We further investigated the reproducibility of the three technologies, applying various normalization methods to the 850k data. Most of these methods reduced variability to a level below that of RRBS data. We then used 850k with our new annotation and normalization to profile 5mC changes in breast cancer biopsies. 850k highlighted aberrant enhancer methylation as the predominant feature, in agreement with previous reports. Our study provides an updated processing approach for 850k data, based on refined probe annotation and normalization, allowing for improved analysis of methylation at enhancers and long noncoding RNA genes. Our findings will help to further advance understanding of the DNA methylome in health and disease.


Asunto(s)
Metilación de ADN , ARN Largo no Codificante , Humanos , Islas de CpG , ARN Largo no Codificante/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Benchmarking , Reproducibilidad de los Resultados
5.
Nat Cancer ; 2(6): 611-628, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-35121941

RESUMEN

Post-transcriptional modifications of RNA constitute an emerging regulatory layer of gene expression. The demethylase fat mass- and obesity-associated protein (FTO), an eraser of N6-methyladenosine (m6A), has been shown to play a role in cancer, but its contribution to tumor progression and the underlying mechanisms remain unclear. Here, we report widespread FTO downregulation in epithelial cancers associated with increased invasion, metastasis and worse clinical outcome. Both in vitro and in vivo, FTO silencing promotes cancer growth, cell motility and invasion. In human-derived tumor xenografts (PDXs), FTO pharmacological inhibition favors tumorigenesis. Mechanistically, we demonstrate that FTO depletion elicits an epithelial-to-mesenchymal transition (EMT) program through increased m6A and altered 3'-end processing of key mRNAs along the Wnt signaling cascade. Accordingly, FTO knockdown acts via EMT to sensitize mouse xenografts to Wnt inhibition. We thus identify FTO as a key regulator, across epithelial cancers, of Wnt-triggered EMT and tumor progression and reveal a therapeutically exploitable vulnerability of FTO-low tumors.


Asunto(s)
Neoplasias Glandulares y Epiteliales , ARN , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Animales , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/genética , Humanos , Ratones
7.
Nucleic Acids Res ; 47(W1): W561-W565, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31114869

RESUMEN

The recent growth in the number of publicly available cancer omics databases has been accompanied by the development of various tools that allow researchers to visually explore these data. In 2015, we built MEXPRESS, an online tool for the integration and visualization of gene expression, DNA methylation and clinical data from The Cancer Genome Atlas (TCGA), a large collection of publicly available multi-omics cancer data. MEXPRESS addresses the need for an easy-to-use, interactive application that allows researchers to identify dysregulated genes and their clinical relevance in cancer. Furthermore, while other tools typically do not support integrated visualization of expression and DNA methylation data in combination with the precise genomic location of the methylation, MEXPRESS is unique in how it depicts these diverse data types together. Motivated by the large number of users MEXPRESS has managed to attract over the past 3 years and the recent migration of all TCGA data to a new data portal, we developed a new version of MEXPRESS (https://mexpress.be). It contains the latest TCGA data, additional types of omics and clinical data and extra functionality, allowing users to explore mechanisms of gene dysregulation beyond expression and DNA methylation.


Asunto(s)
Metilación de ADN , Expresión Génica , Neoplasias/genética , Programas Informáticos , Genómica , Humanos , RNA-Seq
8.
J Clin Invest ; 127(8): 3090-3102, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28714863

RESUMEN

BACKGROUND: The tumor immune response is increasingly associated with better clinical outcomes in breast and other cancers. However, the evaluation of tumor-infiltrating lymphocytes (TILs) relies on histopathological measurements with limited accuracy and reproducibility. Here, we profiled DNA methylation markers to identify a methylation of TIL (MeTIL) signature that recapitulates TIL evaluations and their prognostic value for long-term outcomes in breast cancer (BC). METHODS: MeTIL signature scores were correlated with clinical endpoints reflecting overall or disease-free survival and a pathologic complete response to preoperative anthracycline therapy in 3 BC cohorts from the Jules Bordet Institute in Brussels and in other cancer types from The Cancer Genome Atlas. RESULTS: The MeTIL signature measured TIL distributions in a sensitive manner and predicted survival and response to chemotherapy in BC better than did histopathological assessment of TILs or gene expression-based immune markers, respectively. The MeTIL signature also improved the prediction of survival in other malignancies, including melanoma and lung cancer. Furthermore, the MeTIL signature predicted differences in survival for malignancies in which TILs were not known to have a prognostic value. Finally, we showed that MeTIL markers can be determined by bisulfite pyrosequencing of small amounts of DNA from formalin-fixed, paraffin-embedded tumor tissue, supporting clinical applications for this methodology. CONCLUSIONS: This study highlights the power of DNA methylation to evaluate tumor immune responses and the potential of this approach to improve the diagnosis and treatment of breast and other cancers. FUNDING: This work was funded by the Fonds National de la Recherche Scientifique (FNRS) and Télévie, the INNOVIRIS Brussels Region BRUBREAST Project, the IUAP P7/03 program, the Belgian "Foundation against Cancer," the Breast Cancer Research Foundation (BCRF), and the Fonds Gaston Ithier.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Metilación de ADN , Anciano , Antraciclinas/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Separación Celular , Estudios de Cohortes , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Humanos , Sistema Inmunológico , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Linfocitos Infiltrantes de Tumor/citología , Masculino , Melanoma/diagnóstico , Melanoma/genética , Melanoma/terapia , Persona de Mediana Edad , Periodo Preoperatorio , Pronóstico , Modelos de Riesgos Proporcionales , Análisis de Secuencia de ADN , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/terapia , Resultado del Tratamiento
9.
PLoS One ; 12(6): e0179501, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28622390

RESUMEN

Innovative therapies for solid tumors are urgently needed. Recently, therapies that harness the host immune system to fight cancer cells have successfully treated a subset of patients with solid tumors. These responses have been strong and durable but observed in subsets of patients. Work from our group and others has shown that epigenetic therapy, specifically inhibiting the silencing DNA methylation mark, activates immune signaling in tumor cells and can sensitize to immune therapy in murine models. Here we show that colon and ovarian cancer cell lines exhibit lower expression of transcripts involved in antigen processing and presentation to immune cells compared to normal tissues. In addition, treatment with clinically relevant low doses of DNMT inhibitors (that remove DNA methylation) increases expression of both antigen processing and presentation and Cancer Testis Antigens in these cell lines. We confirm that treatment with DNMT inhibitors upregulates expression of the antigen processing and presentation molecules B2M, CALR, CD58, PSMB8, PSMB9 at the RNA and protein level in a wider range of colon and ovarian cancer cell lines and treatment time points than had been described previously. In addition, we show that DNMTi treatment upregulates many Cancer Testis Antigens common to both colon and ovarian cancer. This increase of both antigens and antigen presentation by epigenetic therapy may be one mechanism to sensitize patients to immune therapies.


Asunto(s)
Presentación de Antígeno/genética , Antígenos de Neoplasias , Metilación de ADN , ADN de Neoplasias , Neoplasias Ováricas , Neoplasias Testiculares , Animales , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Células CACO-2 , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Metilación de ADN/genética , Metilación de ADN/inmunología , ADN de Neoplasias/genética , ADN de Neoplasias/inmunología , ADN de Neoplasias/metabolismo , Femenino , Humanos , Masculino , Ratones , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Testiculares/genética , Neoplasias Testiculares/inmunología , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patología
10.
Clin Cancer Res ; 23(8): 2006-2018, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-27756787

RESUMEN

Purpose: The currently used prognostic models for patients with nonmetastatic clear cell renal cell carcinoma (ccRCC) are based on clinicopathologic features and might be improved by adding molecular markers. Epigenetic alterations occur frequently in ccRCC and are promising biomarkers. The aim of this study is to identify prognostic promoter methylation markers for ccRCC.Experimental Design: We integrated data generated by massive parallel sequencing of methyl-binding domain enriched DNA and microarray-based RNA expression profiling of 5-aza-2'-deoxycytidine-treated ccRCC cell lines to comprehensively characterize the ccRCC methylome. A selection of the identified methylation markers was evaluated in two independent series of primary ccRCC (n = 150 and n = 185) by methylation-specific PCR. Kaplan-Meier curves and log-rank tests were used to estimate cause-specific survival. HRs and corresponding 95% confidence intervals (CI) were assessed using Cox proportional hazard models. To assess the predictive capacity and fit of models combining several methylation markers, HarrellC statistic and the Akaike Information Criterion were used.Results: We identified four methylation markers, that is, GREM1, NEURL, LAD1, and NEFH, that individually predicted prognosis of patients with ccRCC. The four markers combined were associated with poorer survival in two independent patient series (HR, 3.64; 95% CI, 1.02-13.00 and HR, 7.54; 95% CI, 2.68-21.19). These findings were confirmed in a third series of ccRCC cases from The Cancer Genome Atlas (HR, 3.60; 95% CI, 2.02-6.40).Conclusions: A four-gene promoter methylation marker panel consisting of GREM1, NEURL, LAD1, and NEFH predicts outcome of patients with ccRCC and might be used to improve current prognostic models. Clin Cancer Res; 23(8); 2006-18. ©2016 AACR.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Adulto , Anciano , Autoantígenos/genética , Carcinoma de Células Renales/mortalidad , Metilación de ADN/genética , Supervivencia sin Enfermedad , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Estimación de Kaplan-Meier , Neoplasias Renales/mortalidad , Masculino , Persona de Mediana Edad , Proteínas de Neurofilamentos/genética , Colágenos no Fibrilares/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Regiones Promotoras Genéticas/genética , Modelos de Riesgos Proporcionales , Ubiquitina-Proteína Ligasas/genética , Colágeno Tipo XVII
11.
Curr Opin Genet Dev ; 36: 16-26, 2016 02.
Artículo en Inglés | MEDLINE | ID: mdl-26875115

RESUMEN

The discovery of TET-mediated DNA hydroxymethylation as a mechanism of DNA demethylation, along with the observation of disrupted hydroxymethylation patterns in cancer, sparked high hopes of better understanding malignant processes. In this review, we discuss a plethora of recent studies that have shed light on the mechanisms and biological consequences of DNA hydroxymethylation pattern changes in various cancers. A picture is taking shape, in which TET proteins appear as both promoters and suppressors of cancer. Their impairment at multiple levels creates abnormal 5hmC landscapes that affect, often in concert with key cancer pathways, a wider range of biological processes than initially proposed. As the picture gains in scope and precision, the prospect of 5hmC-pattern-targeting cancer therapies shimmers in the distance.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética , Neoplasias/genética , 5-Metilcitosina/metabolismo , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Humanos , Oxigenasas de Función Mixta/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética
12.
Sci Rep ; 5: 12714, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26239807

RESUMEN

The TET enzymes convert methylcytosine to the newly discovered base hydroxymethylcytosine. While recent reports suggest that TETs may play a role in response to oxidative stress, this role remains uncertain, and results lack in vivo models. Here we show a global decrease of hydroxymethylcytosine in cells treated with buthionine sulfoximine, and in mice depleted for the major antioxidant enzymes GPx1 and 2. Furthermore, genome-wide profiling revealed differentially hydroxymethylated regions in coding genes, and intriguingly in microRNA genes, both involved in response to oxidative stress. These results thus suggest a profound effect of in vivo oxidative stress on the global hydroxymethylome.


Asunto(s)
5-Metilcitosina/metabolismo , Proteínas de Unión al ADN/genética , Genoma , MicroARNs/genética , Neuronas/metabolismo , Proteínas Proto-Oncogénicas/genética , 5-Metilcitosina/análogos & derivados , Animales , Antimetabolitos/farmacología , Butionina Sulfoximina/farmacología , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Glutatión/antagonistas & inhibidores , Glutatión/biosíntesis , Glutatión Peroxidasa/deficiencia , Glutatión Peroxidasa/genética , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Estrés Oxidativo , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal , Glutatión Peroxidasa GPX1
13.
BMC Genomics ; 16: 636, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26306699

RESUMEN

BACKGROUND: In recent years, increasing amounts of genomic and clinical cancer data have become publically available through large-scale collaborative projects such as The Cancer Genome Atlas (TCGA). However, as long as these datasets are difficult to access and interpret, they are essentially useless for a major part of the research community and their scientific potential will not be fully realized. To address these issues we developed MEXPRESS, a straightforward and easy-to-use web tool for the integration and visualization of the expression, DNA methylation and clinical TCGA data on a single-gene level ( http://mexpress.be ). RESULTS: In comparison to existing tools, MEXPRESS allows researchers to quickly visualize and interpret the different TCGA datasets and their relationships for a single gene, as demonstrated for GSTP1 in prostate adenocarcinoma. We also used MEXPRESS to reveal the differences in the DNA methylation status of the PAM50 marker gene MLPH between the breast cancer subtypes and how these differences were linked to the expression of MPLH. CONCLUSIONS: We have created a user-friendly tool for the visualization and interpretation of TCGA data, offering clinical researchers a simple way to evaluate the TCGA data for their genes or candidate biomarkers of interest.


Asunto(s)
Biología Computacional/métodos , Metilación de ADN , Genómica/métodos , Programas Informáticos , Humanos , Navegador Web
14.
Epigenetics ; 10(7): 622-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25985363

RESUMEN

Neurofilament heavy polypeptide (NEFH) has recently been identified as a candidate DNA hypermethylated gene within the functional breast cancer hypermethylome. NEFH exists in a complex with neurofilament medium polypeptide (NEFM) and neurofilament light polypeptide (NEFL) to form neurofilaments, which are structural components of the cytoskeleton in mature neurons. Recent studies reported the deregulation of these proteins in several malignancies, suggesting that neurofilaments may have a role in other cell types as well. Using a comprehensive approach, we studied the epigenetic inactivation of neurofilament genes in breast cancer and the functional significance of this event. We report that DNA methylation-associated silencing of NEFH, NEFL, and NEFM in breast cancer is frequent, cancer-specific, and correlates with clinical features of disease progression. DNA methylation-mediated inactivation of these genes occurs also in multiple other cancer histologies including pancreas, gastric, and colon. Restoration of NEFH function, the major subunit of the neurofilament complex, reduces proliferation and growth of breast cancer cells and arrests them in Go/G1 phase of the cell cycle along with a reduction in migration and invasion. These findings suggest that DNA methylation-mediated silencing of the neurofilament genes NEFH, NEFM, and NEFL are frequent events that may contribute to the progression of breast cancer and possibly other malignancies.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Epigénesis Genética , Silenciador del Gen , Proteínas de Neurofilamentos/genética , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Metilación de ADN , Femenino , Humanos , Filamentos Intermedios/patología , Regiones Promotoras Genéticas
15.
Clin Cancer Res ; 21(15): 3492-500, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25904753

RESUMEN

PURPOSE: In this era of molecular diagnostics, prediction of clear-cell renal cell cancer (ccRCC) survival requires optimization, as current prognostic markers fail to determine individual patient outcome. Epigenetic events are promising molecular markers. Promoter CpG island methylation of cysteine dioxygenase type 1 (CDO1), which was identified as prognostic marker for breast cancer, is studied as a potential marker for ccRCC survival. EXPERIMENTAL DESIGN: We collected primary tissues of 365 ccRCC cases identified within the prospective Netherlands Cohort Study (NLCS). In this population-based series, CDO1 promoter methylation was observed in 124 of 324 (38.3%) patients with successful methylation-specific PCR analysis. Kaplan-Meier curves and Wilcoxon tests were used to evaluate 10-year ccRCC-specific survival. Cox regression analysis was used to obtain crude and multivariate HRs and 95% confidence intervals (CI). The relative prognostic value of multivariate models with and without CDO1 promoter methylation was compared using likelihood-ratio tests. RESULTS: Patients with CDO1 promoter methylation have a significantly poorer survival than those without (Wilcoxon P = 0.006). Differences in survival were independent of other prognostic factors, including age and sex (HR, 1.66; 95% CI, 1.12-2.45) and TNM stage, tumor size, and Fuhrman grade (HR, 1.89; 95% CI, 1.25-2.85). Multivariate models performed better with than without CDO1 promoter methylation status (likelihood-ratio P = 0.003). Survival curves were validated in an independent series of 280 ccRCC cases from The Cancer Genome Atlas (TCGA; Wilcoxon P < 0.001). CONCLUSIONS: CDO1 promoter methylation may not substitute common prognostic makers to predict ccRCC survival, but offers additional, relevant prognostic information, indicating that it might be a novel molecular marker to determine ccRCC prognosis.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Renales/genética , Cisteína-Dioxigenasa/genética , Metilación de ADN/genética , Anciano , Carcinoma de Células Renales/patología , Islas de CpG/genética , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Países Bajos , Pronóstico , Regiones Promotoras Genéticas , Factores de Riesgo
16.
FEBS J ; 282(9): 1801-14, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25331982

RESUMEN

Breast cancer, one of the most common and deadliest malignancies in developed countries, is a remarkably heterogeneous disease, which is clinically reflected by patients who display similar pathological features but respond differently to treatments. In the search for mediators of responsiveness, the tumor microenvironment (TME), in particular tumor-associated immune cells, has been pushed into the spotlight as it has become clear that the TME is an active component of breast cancer disease that affects clinical outcomes. Thus, the characterization of the TME in terms of cell identities and their frequencies has generated a great deal of interest. The common methods currently used for this purpose are either limited in accuracy or application, and DNA methylation has recently been proposed as an alternative approach. The aim of this review is to discuss DNA methylation profiling beyond promoters as a potential clinical tool for TME characterization and cell typing within tumors. With respect to this, we review the role of DNA methylation in breast cancer and cell-lineage specification, as well as inform about the composition and clinical relevance of the TME.


Asunto(s)
Neoplasias de la Mama/genética , Metilación de ADN , Epigénesis Genética , Microambiente Tumoral , Neoplasias de la Mama/patología , Linaje de la Célula , Femenino , Humanos
17.
Epigenetics ; 9(5): 738-46, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24518818

RESUMEN

CpG island methylator phenotype (CIMP) has been found in multiple precancerous and cancerous lesions, including colorectal adenomas, colorectal cancers, and duodenal adenocarcinomas. There are no reports in the literature of a relationship between CIMP status and clinicopathologic features of sporadic duodenal adenomas. This study sought to elucidate the role of methylation in duodenal adenomas and correlate it with KRAS and BRAF mutations. CIMP+ (with more than 2 markers methylated) was seen in 33.3% of duodenal adenomas; 61% of these CIMP+ adenomas were CIMP-high (with more than 3 markers methylated). Furthermore, CIMP+ status significantly correlated with older age of patients, larger size and villous type of tumor, coexistent dysplasia and periampullary location. MLH1 methylation was seen in 11.1% of duodenal adenomas and was significantly associated with CIMP+ tumors, while p16 methylation was an infrequent event. KRAS mutations were frequent and seen in 26.3% of adenomas; however, no BRAF mutations were detected. Furthermore, CIMP-high status was associated with larger size and villous type of tumor and race (non-white). These results suggest that CIMP+ duodenal adenomas may have a higher risk for developing malignancy and may require more aggressive management and surveillance.


Asunto(s)
Adenoma/genética , Neoplasias Colorrectales/genética , Islas de CpG , Metilación de ADN , Adenoma/patología , Neoplasias Colorrectales/patología , Epigénesis Genética , Femenino , Genes p16 , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras) , Riesgo , Proteínas ras/genética
18.
PLoS One ; 8(7): e68630, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23844228

RESUMEN

Downregulation of the tight junction protein claudin 1 is a frequent event in breast cancer and is associated with recurrence, metastasis, and reduced survival, suggesting a tumor suppressor role for this protein. Tumor suppressor genes are often epigenetically silenced in cancer. Downregulation of claudin 1 via DNA promoter methylation may thus be an important determinant in breast cancer development and progression. To investigate if silencing of claudin 1 has an epigenetic etiology in breast cancer we compared gene expression and methylation data from 217 breast cancer samples and 40 matched normal samples available through the Cancer Genome Atlas (TCGA). Moreover, we analyzed claudin 1 expression and methylation in 26 breast cancer cell lines. We found that methylation of the claudin 1 promoter CpG island is relatively frequent in estrogen receptor positive (ER+) breast cancer and is associated with low claudin 1 expression. In contrast, the claudin 1 promoter was not methylated in most of the ER-breast cancers samples and some of these tumors overexpress claudin 1. In addition, we observed that the demethylating agents, azacitidine and decitabine can upregulate claudin 1 expression in breast cancer cell lines that have a methylated claudin 1 promoter. Taken together, our results indicate that DNA promoter methylation is causally associated with downregulation of claudin 1 in a subgroup of breast cancer that includes mostly ER+ tumors, and suggest that epigenetic therapy to restore claudin 1 expression might represent a viable therapeutic strategy in this subtype of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Claudina-1/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Regiones Promotoras Genéticas , Receptores de Estrógenos/metabolismo , Línea Celular , Claudina-1/metabolismo , Análisis por Conglomerados , Islas de CpG , Femenino , Orden Génico , Silenciador del Gen , Humanos , Transcriptoma
19.
Clin Cancer Res ; 19(12): 3201-11, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23630167

RESUMEN

PURPOSE: Genome-wide DNA methylation analyses have identified hundreds of candidate DNA-hypermethylated genes in cancer. Comprehensive functional analyses provide an understanding of the biologic significance of this vast amount of DNA methylation data that may allow the determination of key epigenetic events associated with tumorigenesis. EXPERIMENTAL DESIGN: To study mechanisms of cysteine dioxygenase type 1 (CDO1) inactivation and its functional significance in breast cancer in a comprehensive manner, we screened for DNA methylation and gene mutations in primary breast cancers and analyzed growth, survival, and reactive oxygen species (ROS) production in breast cancer cells with restored CDO1 function in the context of anthracycline treatment. RESULTS: DNA methylation-associated silencing of CDO1 in breast cancer is frequent (60%), cancer specific, and correlates with disease progression and outcome. CDO1 function can alternatively be silenced by repressive chromatin, and we describe protein-damaging missense mutations in 7% of tumors without DNA methylation. Restoration of CDO1 function in breast cancer cells increases levels of ROS and leads to reduced viability and growth, as well as sensitization to anthracycline treatment. Priming with 5-azacytidine of breast cancer cells with epigenetically silenced CDO1 resulted in restored expression and increased sensitivity to anthracyclines. CONCLUSION: We report that silencing of CDO1 is a critical epigenetic event that contributes to the survival of oxidative-stressed breast cancer cells through increased detoxification of ROS and thus leads to the resistance to ROS-generating chemotherapeutics including anthracyclines. Our study shows the importance of CDO1 inactivation in breast cancer and its clinical potential as a biomarker and therapeutic target to overcome resistance to anthracyclines.


Asunto(s)
Antraciclinas/administración & dosificación , Neoplasias de la Mama/genética , Cisteína-Dioxigenasa/genética , Resistencia a Antineoplásicos/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisteína-Dioxigenasa/antagonistas & inhibidores , Metilación de ADN/genética , Resistencia a Antineoplásicos/genética , Femenino , Silenciador del Gen , Humanos , Especies Reactivas de Oxígeno/metabolismo
20.
Int J Cancer ; 132(11): 2502-9, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23065691

RESUMEN

KRAS mutations have been found in duodenal adenocarcinomas and may have prognostic significance. The purpose of this study was to classify clinicopathological characteristics, microsatellite instability and KRAS mutations and identify possible prognostic role of KRAS mutations in duodenal adenocarcinomas. Demographics, tumor characteristics and survival were recorded for 78 patients with duodenal adenocarcinomas (Stages I-III). KRAS mutations were detected in 27 (34.6%) cases, of which the majority (74.1%) were G>A transitions. Multivariate logistic regression analysis showed that KRAS G>A mutation was significantly associated with late stage (p = 0.025) and poor tumor differentiation (p = 0.035), when compared with wild-type and other than G>A mutations. KRAS G>A mutation carriers were at increased risk for distant relapse (p = 0.022) and had significantly shorter overall survival (OS; log-rank p = 0.045) and a trend toward shorter relapse-free survival (RFS; log-rank p = 0.062) when compared with those who did not carry the KRAS G>A mutation. In multivariate analyses, there was a significant correlation between ≥ 3 positive lymph nodes and poor OS (p < 0.001) and RFS (p = 0.001) and KRAS G>A mutation carriers demonstrated no effect on clinical outcome. In conclusion, KRAS G>A mutation correlates significantly with late stage and poor tumor differentiation in duodenal adenocarcinoma. Among patients who undergo a curative resection of duodenal adenocarcinoma, KRAS G>A mutation carriers will more likely experience distant relapse but may not exhibit a poor prognosis. The number of positive lymph nodes should be incorporated in future staging systems.


Asunto(s)
Adenocarcinoma/genética , Biomarcadores de Tumor/genética , Diferenciación Celular , Neoplasias Duodenales/genética , Mutación/genética , Recurrencia Local de Neoplasia/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Adenocarcinoma/cirugía , ADN de Neoplasias , Neoplasias Duodenales/mortalidad , Neoplasias Duodenales/patología , Neoplasias Duodenales/cirugía , Femenino , Humanos , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/cirugía , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa , Pronóstico , Proteínas Proto-Oncogénicas p21(ras) , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...