Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Sci Rep ; 9(1): 14991, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31628391

RESUMEN

Breast cancer in very young women (≤35 years; BCVY) presents more aggressive and complex biological features than their older counterparts (BCO). Our aim was to evaluate methylation differences between BCVY and BCO and their DNA epigenetic age. EPIC and 450k Illumina methylation arrays were used in 67 breast cancer tumours, including 32 from BCVY, for methylation study and additionally we analysed their epigenetic age. We identified 2 219 CpG sites differently-methylated in BCVY vs. BCO (FDR < 0.05; ß-value difference ± 0.1). The signature showed a general hypomethylation profile with a selective small hypermethylation profile located in open-sea regions in BCVY against BCO and normal tissue. Strikingly, BCVY presented a significant increased epigenetic age-acceleration compared with older women. The affected genes were enriched for pathways in neuronal-system pathways, cell communication, and matrix organisation. Validation in an independent sample highlighted consistent higher expression of HOXD9, and PCDH10 genes in BCVY. Regions implicated in the hypermethylation profile were involved in Notch signalling pathways, the immune system or DNA repair. We further validated HDAC5 expression in BCVY. We have identified a DNA methylation signature that is specific to BCVY and have shown that epigenetic age-acceleration is increased in BCVY.


Asunto(s)
Neoplasias de la Mama/genética , Metilación de ADN/genética , Epigénesis Genética , Adulto , Factores de Edad , Anciano , Cadherinas/genética , Islas de CpG , Epigenómica/métodos , Femenino , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Histona Desacetilasas/genética , Proteínas de Homeodominio/genética , Humanos , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas , Protocadherinas
2.
Mol Nutr Food Res ; 63(19): e1801386, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31327173

RESUMEN

SCOPE: Loci-specific increase in DNA methylation occurs in cancer and may underlie gene silencing. It is investigated whether dietary stilbenoids, resveratrol, and pterostilbene exert time-dependent effects on DNA methylation patterns and specifically methylation-silenced tumor suppressor genes in breast cancer cells. METHODS AND RESULTS: Following genome-wide DNA methylation analysis with Illumina-450K, changes characteristic of early and late response to stilbenoids are identified. Interestingly, often the same genes but at different CpG loci, the same gene families, or the same functional gene categories are affected. CpG loci that lose methylation in exposed cells correspond to genes functionally associated with cancer suppression. There is a group of genes, including SEMA3A, at which the magnitude of hypomethylation in response to stilbenoids rises with increasing invasive potential of cancer cells. Decreased DNA methylation at SEMA3A promoter and concomitant gene upregulation coincide with increased occupancy of active histone marks. Open chromatin upon exposure to stilbenoids may be linked to decreased DNMT3A binding followed by increased NF1C transcription factor occupancy. Sequestration of DNMT3A is possibly a result of stilbenoid-mediated increase in SALL3 expression, which was previously shown to bind and inhibit DNMT3A activity. CONCLUSIONS: The findings define mechanistic players in stilbenoid-mediated epigenetic reactivation of genes suppressing cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Epigénesis Genética/efectos de los fármacos , Factores de Transcripción NFI/metabolismo , Semaforina-3A/genética , Estilbenos/farmacología , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/efectos de los fármacos , ADN Metiltransferasa 3A , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Silenciador del Gen/efectos de los fármacos , Proteínas de Homeodominio/genética , Humanos , Invasividad Neoplásica/prevención & control , Regiones Promotoras Genéticas/genética , Resveratrol/farmacología , Semaforina-3A/fisiología , Factores de Transcripción/genética
3.
Sci Rep ; 8(1): 14373, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30258192

RESUMEN

MiRNAs are part of the epigenetic machinery, and are also epigenetically modified by DNA methylation. MiRNAs regulate expression of different genes, so any alteration in their methylation status may affect their expression. We aimed to identify methylation differences in miRNA encoding genes in breast cancer affecting women under 35 years old (BCVY), in order to identify potential biomarkers in these patients. In Illumina Infinium MethylationEPIC BeadChip samples (metEPICVal), we analysed the methylation of 9,961 CpG site regulators of miRNA-encoding genes present in the array. We identified 193 differentially methylated CpG sites in BCVY (p-value < 0.05 and methylation differences ±0.1) that regulated 83 unique miRNA encoding genes. We validated 10 CpG sites using two independent datasets based on Infinium Human Methylation 450k array. We tested gene expression of miRNAs with differential methylation in BCVY in a meta-analysis using The Cancer Genome Atlas (TCGA), Clariom D and Affymetrix datasets. Five miRNAs (miR-9, miR-124-2, miR-184, miR-551b and miR-196a-1) were differently expressed (FDR p-value < 0.01). Finally, only miR-124-2 shows a significantly different gene expression by quantitative real-time PCR. MiR-124-hypomethylation presents significantly better survival rates for older patients as opposed to the worse prognosis observed in BCVY, identifying it as a potential specific survival biomarker in BCVY.


Asunto(s)
Neoplasias de la Mama/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Regiones Promotoras Genéticas , Adulto , Factores de Edad , Biomarcadores de Tumor/genética , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/epidemiología , Islas de CpG , Femenino , Humanos , Pronóstico , Análisis de Supervivencia
4.
Epigenetics ; 13(6): 605-626, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29927686

RESUMEN

Late onset of clinical symptoms in hepatocellular carcinoma (HCC) results in late diagnosis and poor disease outcome. Approximately 85% of individuals with HCC have underlying liver cirrhosis. However, not all cirrhotic patients develop cancer. Reliable tools that would distinguish cirrhotic patients who will develop cancer from those who will not are urgently needed. We used the Illumina HumanMethylation450 BeadChip microarray to test whether white blood cell DNA, an easily accessible source of DNA, exhibits site-specific changes in DNA methylation in blood of diagnosed HCC patients (post-diagnostic, 24 cases, 24 controls) and in prospectively collected blood specimens of HCC patients who were cancer-free at blood collection (pre-diagnostic, 21 cases, 21 controls). Out of 22 differentially methylated loci selected for validation by pyrosequencing, 19 loci with neighbouring CpG sites (probes) were confirmed in the pre-diagnostic study group and subjected to verification in a prospective cirrhotic cohort (13 cases, 23 controls). We established for the first time 9 probes that could distinguish HBV-negative cirrhotic patients who subsequently developed HCC from those who stayed cancer-free. These probes were identified within regulatory regions of BARD1, MAGEB3, BRUNOL5, FXYD6, TET1, TSPAN5, DPPA5, KIAA1210, and LSP1. Methylation levels within DPPA5, KIAA1210, and LSP1 were higher in prospective samples from HCC cases vs. cirrhotic controls. The remaining probes were hypomethylated in cases compared with controls. Using blood as a minimally invasive material and pyrosequencing as a straightforward quantitative method, the established probes have potential to be developed into a routine clinical test after validation in larger cohorts.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/genética , Metilación de ADN , Neoplasias Hepáticas/genética , Femenino , Sitios Genéticos , Predisposición Genética a la Enfermedad , Humanos , Masculino
5.
Curr Cancer Drug Targets ; 18(1): 5-15, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28176650

RESUMEN

The efficacy of cancer immunotherapy relies on the ability of the host immune system to recognise the cancer as non-self and eliminate it from the body. Whilst this is an extremely fertile area of medical research, with positive clinical trials showing durable responses, attention must be paid to the subset of patients that do not respond to these treatments. Immune surveillance and immunoediting by the host could itself select for immune-evasive tumour cells during tumour development leading to immunotherapy resistance. One such mechanism of non-efficacy or resistance is the epigenetic silencing of a specific gene required in the immunotherapy response pathway. Epigenetics is the study of the control of expression patterns in a cell via mechanisms not involving a change in DNA sequence. All tumour types show aberrant epigenetic regulation of genes involved in all the hallmarks of cancer, including immunomodulation. Inhibition of key enzymes involved in maintenance of epigenetic states is another important area of research for new treatment strategies for cancer. Could epigenetic therapies be used to successfully enhance the action of immunomodulatory agents in cancer, and are they acting in the way we imagine? An understanding of the effects of epigenetic therapies on immunological pathways in both the tumour and host cells, especially the tumour microenvironment, will be essential to further develop such combination approaches.


Asunto(s)
Epigénesis Genética , Inmunomodulación/genética , Inmunoterapia , Neoplasias/genética , Neoplasias/terapia , Microambiente Tumoral/genética , Animales , Humanos , Factores Inmunológicos , Neoplasias/inmunología , Microambiente Tumoral/inmunología
6.
Br J Cancer ; 116(10): 1287-1293, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28350786

RESUMEN

BACKGROUND: Survival benefit from surgical debulking of ovarian cancer (OC) is well established, but some women, despite total macroscopic clearance of disease, still have poor prognosis. We aimed to identify biomarkers to predict benefit from conventional surgery. METHODS: Clinical data from women debulked for high-stage OC were analysed (Hammersmith Hospital, London, UK; 2001-2014). Infinium's HumanMethylation27 array interrogated tumour DNA for differentially methylated CpG sites, correlated to survival, in patients with the least residual disease (RD; Hammersmith Array). Validation was performed using bisulphite pyrosequencing (Charité Hospital, Berlin, Germany cohort) and The Cancer Genome Atlas' (TCGA) methylation data set. Kaplan-Meier curves and Cox models tested survival. RESULTS: Altogether 803 women with serous OC were studied. No RD was associated with significantly improved overall survival (OS; hazard ratio (HR) 1.25, 95% CI 1.06-1.47; P=0.0076) and progression-free survival (PFS; HR 1.23, 95% CI 1.05-1.43; P=0.012; Hammersmith database n=430). Differentially methylated loci within FGF4, FGF21, MYLK2, MYLK3, MYL7, and ITGAE associated with survival. Patients with the least RD had significantly better OS with higher methylation of MYLK3 (Hammersmith (HR 0.51, 95% CI 0.31-0.84; P=0.01), Charité (HR 0.46, 95% CI 0.21-1.01; P=0.05), and TCGA (HR 0.64, 95% CI 0.44-0.93; P=0.02)). CONCLUSIONS: MYLK3 methylation is associated with improved OS in patients with the least RD, which could potentially be used to determine response to surgery.


Asunto(s)
Carcinoma/genética , Neoplasias de las Trompas Uterinas/genética , Quinasa de Cadena Ligera de Miosina/genética , Neoplasias Ováricas/genética , Neoplasias Peritoneales/genética , Regiones Promotoras Genéticas , Biomarcadores de Tumor/genética , Carcinoma/cirugía , Islas de CpG , Procedimientos Quirúrgicos de Citorreducción , Metilación de ADN , Neoplasias de las Trompas Uterinas/cirugía , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasia Residual , Neoplasias Ováricas/cirugía , Neoplasias Peritoneales/cirugía , Modelos de Riesgos Proporcionales , Medición de Riesgo/métodos , Tasa de Supervivencia
7.
Clin Cancer Res ; 23(13): 3453-3460, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27986748

RESUMEN

Purpose: We aimed to establish whether programmed cell death-1 (PD-1) and programmed cell death ligand 1 (PD-L1) expression, in ovarian cancer tumor tissue and blood, could be used as biomarkers for discrimination of tumor histology and prognosis of ovarian cancer.Experimental Design: Immune cells were separated from blood, ascites, and tumor tissue obtained from women with suspected ovarian cancer and studied for the differential expression of possible immune biomarkers using flow cytometry. PD-L1 expression on tumor-associated inflammatory cells was assessed by immunohistochemistry and tissue microarray. Plasma soluble PD-L1 was measured using sandwich ELISA. The relationships among immune markers were explored using hierarchical cluster analyses.Results: Biomarkers from the discovery cohort that associated with PD-L1+ cells were found. PD-L1+ CD14+ cells and PD-L1+ CD11c+ cells in the monocyte gate showed a distinct expression pattern when comparing benign tumors and epithelial ovarian cancers (EOCs)-confirmed in the validation cohort. Receiver operating characteristic curves showed PD-L1+ and PD-L1+ CD14+ cells in the monocyte gate performed better than the well-established tumor marker CA-125 alone. Plasma soluble PD-L1 was elevated in patients with EOC compared with healthy women and patients with benign ovarian tumors. Low total PD-1+ expression on lymphocytes was associated with improved survival.Conclusions: Differential expression of immunological markers relating to the PD-1/PD-L1 pathway in blood can be used as potential diagnostic and prognostic markers in EOC. These data have implications for the development and trial of anti-PD-1/PD-L1 therapy in ovarian cancer. Clin Cancer Res; 23(13); 3453-60. ©2016 AACR.


Asunto(s)
Antígeno B7-H1/sangre , Biomarcadores de Tumor/sangre , Neoplasias Ováricas/sangre , Receptor de Muerte Celular Programada 1/sangre , Adulto , Anciano , Antígeno Ca-125/sangre , Antígeno Ca-125/inmunología , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Pronóstico
8.
Clin Cancer Res ; 23(9): 2213-2222, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-27663594

RESUMEN

Purpose: DNA damage repair can lead to epigenetic changes. DNA mismatch repair proteins bind to platinum DNA adducts and at sites of DNA damage can recruit the DNA methylating enzyme DNMT1, resulting in aberrant methylation. We hypothesised that DNA damage repair during platinum-based chemotherapy may cause aberrant DNA methylation in normal tissues of patients such as blood.Experimental Design: We used Illumina 450k methylation arrays and bisulphite pyrosequencing to investigate methylation at presentation and relapse in blood DNA from patients with ovarian cancer enrolled in the SCOTROC1 trial (n = 247) and in a cohort of ovarian tumor DNA samples collected at first relapse (n = 46). We used an ovarian cancer cell line model to investigate the role of the DNA mismatch repair gene MLH1 in platinum-induced methylation changes.Results: Specific CpG methylation changes in blood at relapse are observed following platinum-based chemotherapy and are associated with patient survival, independent of other clinical factors [hazard ratio, 3.7; 95% confidence interval, 1.8-7.6, P = 2.8 × 10-4]. Similar changes occur in ovarian tumors at relapse, also associated with patient survival (hazard ratio, 2.6; 95% confidence interval, 1.0-6.8, P = 0.048). Using an ovarian cancer cell line model, we demonstrate that functional mismatch repair increases the frequency of platinum-induced methylation.Conclusions: DNA methylation in blood at relapse following chemotherapy, and not at presentation, is informative regarding survival of patients with ovarian cancer. Functional DNA mismatch repair increases the frequency of DNA methylation changes induced by platinum. DNA methylation in blood following chemotherapy could provide a noninvasive means of monitoring patients' epigenetic responses to treatment without requiring a tumor biopsy. Clin Cancer Res; 23(9); 2213-22. ©2016 AACR.


Asunto(s)
Metilación de ADN/genética , ADN de Neoplasias/sangre , Neoplasias Ováricas/tratamiento farmacológico , Platino (Metal)/administración & dosificación , Anciano , Línea Celular Tumoral , Aductos de ADN/genética , Daño del ADN/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , ADN de Neoplasias/genética , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Platino (Metal)/efectos adversos , Regiones Promotoras Genéticas
9.
Carcinogenesis ; 37(7): 656-68, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27207652

RESUMEN

DNA hypomethylation was previously implicated in cancer progression and metastasis. The purpose of this study was to examine whether stilbenoids, resveratrol and pterostilbene thought to exert anticancer effects, target genes with oncogenic function for de novo methylation and silencing, leading to inactivation of related signaling pathways. Following Illumina 450K, genome-wide DNA methylation analysis reveals that stilbenoids alter DNA methylation patterns in breast cancer cells. On average, 75% of differentially methylated genes have increased methylation, and these genes are enriched for oncogenic functions, including NOTCH signaling pathway. MAML2, a coactivator of NOTCH targets, is methylated at the enhancer region and transcriptionally silenced in response to stilbenoids, possibly explaining the downregulation of NOTCH target genes. The increased DNA methylation at MAML2 enhancer coincides with increased occupancy of repressive histone marks and decrease in activating marks. This condensed chromatin structure is associated with binding of DNMT3B and decreased occupancy of OCT1 transcription factor at MAML2 enhancer, suggesting a role of DNMT3B in increasing methylation of MAML2 after stilbenoid treatment. Our results deliver a novel insight into epigenetic regulation of oncogenic signals in cancer and provide support for epigenetic-targeting strategies as an effective anticancer approach.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Proteínas Nucleares/genética , Transportador 1 de Catión Orgánico/genética , Factores de Transcripción/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cromatina/efectos de los fármacos , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genoma Humano , Humanos , Transportador 1 de Catión Orgánico/biosíntesis , Regiones Promotoras Genéticas , Receptores Notch/genética , Resveratrol , Transducción de Señal/efectos de los fármacos , Estilbenos/administración & dosificación , Transactivadores , Activación Transcripcional/genética , ADN Metiltransferasa 3B
10.
Clin Epigenetics ; 7: 67, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26244061

RESUMEN

BACKGROUND: Interest in the potential of DNA methylation in peripheral blood as a biomarker of cancer risk is increasing. We aimed to assess whether epigenome-wide DNA methylation measured in peripheral blood samples obtained before onset of the disease is associated with increased risk of breast cancer. We report on three independent prospective nested case-control studies from the European Prospective Investigation into Cancer and Nutrition (EPIC-Italy; n = 162 matched case-control pairs), the Norwegian Women and Cancer study (NOWAC; n = 168 matched pairs), and the Breakthrough Generations Study (BGS; n = 548 matched pairs). We used the Illumina 450k array to measure methylation in the EPIC and NOWAC cohorts. Whole-genome bisulphite sequencing (WGBS) was performed on the BGS cohort using pooled DNA samples, combined to reach 50× coverage across ~16 million CpG sites in the genome including 450k array CpG sites. Mean ß values over all probes were calculated as a measurement for epigenome-wide methylation. RESULTS: In EPIC, we found that high epigenome-wide methylation was associated with lower risk of breast cancer (odds ratio (OR) per 1 SD = 0.61, 95 % confidence interval (CI) 0.47-0.80; -0.2 % average difference in epigenome-wide methylation for cases and controls). Specifically, this was observed in gene bodies (OR = 0.51, 95 % CI 0.38-0.69) but not in gene promoters (OR = 0.92, 95 % CI 0.64-1.32). The association was not replicated in NOWAC (OR = 1.03 95 % CI 0.81-1.30). The reasons for heterogeneity across studies are unclear. However, data from the BGS cohort was consistent with epigenome-wide hypomethylation in breast cancer cases across the overlapping 450k probe sites (difference in average epigenome-wide methylation in case and control DNA pools = -0.2 %). CONCLUSIONS: We conclude that epigenome-wide hypomethylation of DNA from pre-diagnostic blood samples may be predictive of breast cancer risk and may thus be useful as a clinical biomarker.

11.
BMC Cancer ; 15: 337, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25927974

RESUMEN

BACKGROUND: DNA methylation variability regions (MVRs) across the oestrogen receptor alpha (ESR1) gene have been identified in peripheral blood cells from breast cancer patients and healthy individuals. In contrast to promoter methylation, gene body methylation may be important in maintaining active transcription. This study aimed to assess MVRs in ESR1 in breast cancer cell lines, tumour biopsies and exfoliated epithelial cells from expressed breast milk (EBM), to determine their significance for ESR1 transcription. METHODS: DNA methylation levels in eight MVRs across ESR1 were assessed by pyrosequencing bisulphite-converted DNA from three oestrogen receptor (ER)-positive and three ER-negative breast cancer cell lines. DNA methylation and expression were assessed following treatment with DAC (1 µM), or DMSO (controls). ESR1 methylation levels were also assayed in DNA from 155 invasive ductal carcinoma biopsies provided by the Breast Cancer Campaign Tissue Bank, and validated with DNA methylation profiles from the TCGA breast tumours (n = 356 ER-pos, n = 109 ER-neg). DNA methylation was profiled in exfoliated breast epithelial cells from EBM using the Illumina 450 K (n = 36) and pyrosequencing in a further 53 donor samples. ESR1 mRNA levels were measured by qRT-PCR. RESULTS: We show that ER-positive cell lines had unmethylated ESR1 promoter regions and highly methylated intragenic regions (median, 80.45%) while ER-negative cells had methylated promoters and lower intragenic methylation levels (median, 38.62%). DAC treatment increased ESR1 expression in ER-negative cells, but significantly reduced methylation and expression of ESR1 in ER-positive cells. The ESR1 promoter was unmethylated in breast tumour biopsies with high levels of intragenic methylation, independent of ER status. However, ESR1 methylation in the strongly ER-positive EBM DNA samples were very similar to ER-positive tumour cell lines. CONCLUSION: DAC treatment inhibited ESR1 transcription in cells with an unmethylated ESR1 promoter and reduced intragenic DNA methylation. Intragenic methylation levels correlated with ESR1 expression in homogenous cell populations (cell lines and exfoliated primary breast epithelial cells), but not in heterogeneous tumour biopsies, highlighting the significant differences between the in vivo tumour microenvironment and individual homogenous cell types. These findings emphasise the need for care when choosing material for epigenetic research and highlights the presence of aberrant intragenic methylation levels in tumour tissue.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma Ductal de Mama/genética , Receptor alfa de Estrógeno/genética , Regiones Promotoras Genéticas , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Línea Celular Tumoral , Metilación de ADN , Epigénesis Genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Glándulas Mamarias Humanas/metabolismo , Leche Humana/citología , Análisis de Secuencia de ADN , Transcripción Genética
12.
Epigenetics ; 10(12): 1121-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26727311

RESUMEN

Germline pathogenic mutations in BRCA1 increase risk of developing breast cancer. Screening for mutations in BRCA1 frequently identifies sequence variants of unknown pathogenicity and recent work has aimed to develop methods for determining pathogenicity. We previously observed that tumor DNA methylation can differentiate BRCA1-mutated from BRCA1-wild type tumors. We hypothesized that we could predict pathogenicity of variants based on DNA methylation profiles of tumors that had arisen in carriers of unclassified variants. We selected 150 FFPE breast tumor DNA samples [47 BRCA1 pathogenic mutation carriers, 65 BRCAx (BRCA1-wild type), 38 BRCA1 test variants] and analyzed a subset (n=54) using the Illumina 450K methylation platform, using the remaining samples for bisulphite pyrosequencing validation. Three validated markers (BACH2, C8orf31, and LOC654342) were combined with sequence bioinformatics in a model to predict pathogenicity of 27 variants (independent test set). Predictions were compared with standard multifactorial likelihood analysis. Prediction was consistent for c.5194-12G>A (IVS 19-12 G>A) (P>0.99); 13 variants were considered not pathogenic or likely not pathogenic using both approaches. We conclude that tumor DNA methylation data alone has potential to be used in prediction of BRCA1 variant pathogenicity but is not independent of estrogen receptor status and grade, which are used in current multifactorial models to predict pathogenicity.


Asunto(s)
Neoplasias de la Mama/genética , Metilación de ADN , Genes BRCA1 , Predisposición Genética a la Enfermedad , Neoplasias de la Mama/patología , Biología Computacional , Análisis Mutacional de ADN , Femenino , Marcadores Genéticos , Humanos
13.
J Virol ; 86(23): 12494-502, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23015699

RESUMEN

The Epstein-Barr virus (EBV) genome sustains substantial epigenetic modification involving chromatin remodelling and DNA methylation during lytic replication. Zta (ZEBRA, BZLF1), a key regulator of the EBV lytic cycle, is a transcription and replication factor, binding to Zta response elements (ZREs) in target promoters and EBV lytic origins of replication. In vitro, Zta binding is modulated by DNA methylation; a subset of CpG-containing Zta binding sites (CpG ZREs) is bound only in a DNA methylation-dependent manner. The question of how the dynamic epigenetic environment impacts Zta interaction during the EBV lytic cycle is unknown. To address this, we used chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-Seq) to identify Zta binding sites across the EBV genome before and after viral DNA replication. Replication did not alter the association of Zta across many regions of the EBV genome, but a striking reduction in Zta binding occurred at some loci that contain CpG ZREs. Separating Zta-bound DNA into methylated and nonmethylated fractions, we found that promoters that contain CpG ZREs were enriched in the methylated fraction but that Zta binding to promoters lacking CpG ZREs was not reduced. We hypothesize that the loss of DNA methylation on the EBV genome during the lytic cycle causes the reduced binding to CpG ZREs; this may act as a lytic cycle epigenetic switch. However, the epigenetic changes associated with the replicated EBV genome do not affect the interaction of Zta with many loci that are rich in non-CpG ZREs; this leads to sustained binding at these regions.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética/genética , Genoma Viral/genética , Herpesvirus Humano 4/genética , Transactivadores/metabolismo , Replicación Viral/fisiología , Sitios de Unión/genética , Western Blotting , Inmunoprecipitación de Cromatina/métodos , Cartilla de ADN/genética , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Unión Proteica/genética , Transactivadores/genética , Replicación Viral/genética
14.
J Virol ; 86(3): 1809-19, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22090141

RESUMEN

The ability of Epstein-Barr virus (EBV) to establish latency allows it to evade the immune system and to persist for the lifetime of its host; one distinguishing characteristic is the lack of transcription of the majority of viral genes. Entry into the lytic cycle is coordinated by the viral transcription factor, Zta (BZLF1, ZEBRA, and EB1), and downstream effectors, while viral genome replication requires the concerted action of Zta and six other viral proteins at the origins of lytic replication. We explored the chromatin context at key EBV lytic cycle promoters (BZLF1, BRLF1, BMRF1, and BALF5) and the origins of lytic replication during latency and lytic replication. We show that a repressive heterochromatin-like environment (trimethylation of histone H3 at lysine 9 [H3K9me3] and lysine 27 [H3K27me3]), which blocks the interaction of some transcription factors with DNA, encompasses the key early lytic regulatory regions. Epigenetic silencing of the EBV genome is also imposed by DNA methylation during latency. The chromatin environment changes during the lytic cycle with activation of histones H3, H4, and H2AX occurring at both the origins of replication and at the key lytic regulatory elements. We propose that Zta is able to reverse the effects of latency-associated repressive chromatin at EBV early lytic promoters by interacting with Zta response elements within the H3K9me3-associated chromatin and demonstrate that these interactions occur in vivo. Since the interaction of Zta with DNA is not inhibited by DNA methylation, it is clear that Zta uses two routes to overcome epigenetic silencing of its genome.


Asunto(s)
Cromatina/metabolismo , Genoma Viral , Herpesvirus Humano 4/genética , Secuencia de Bases , Inmunoprecipitación de Cromatina , Metilación de ADN , Cartilla de ADN , Silenciador del Gen , Herpesvirus Humano 4/fisiología , Metilación , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Transcripción/fisiología , Replicación Viral
15.
PLoS One ; 6(10): e25922, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022468

RESUMEN

Epstein-Barr virus (EBV) encoded transcription factor Zta (BZLF1, ZEBRA, EB1) is the prototype of a class of transcription factor (including C/EBPalpha) that interact with CpG-containing DNA response elements in a methylation-dependent manner. The EBV genome undergoes a biphasic methylation cycle; it is extensively methylated during viral latency but is reset to an unmethylated state following viral lytic replication. Zta is expressed transiently following infection and again during the switch between latency and lytic replication. The requirement for CpG-methylation at critical Zta response elements (ZREs) has been proposed to regulate EBV replication, specifically it could aid the activation of viral lytic gene expression from silenced promoters on the methylated genome during latency in addition to preventing full lytic reactivation from the non-methylated EBV genome immediately following infection. We developed a computational approach to predict the location of ZREs which we experimentally assessed using in vitro and in vivo DNA association assays. A remarkably different binding motif is apparent for the CpG and non-CpG ZREs. Computational prediction of the location of these binding motifs in EBV revealed that the majority of lytic cycle genes have at least one and many have multiple copies of methylation-dependent CpG ZREs within their promoters. This suggests that the abundance of Zta protein coupled with the methylation status of the EBV genome act together to co-ordinate the expression of lytic cycle genes at the majority of EBV promoters.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética , Herpesvirus Humano 4/crecimiento & desarrollo , Herpesvirus Humano 4/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Algoritmos , Secuencia de Bases , Sitios de Unión , Inmunoprecipitación de Cromatina , Islas de CpG/genética , Genoma Viral/genética , Modelos Genéticos , Datos de Secuencia Molecular , Posición Específica de Matrices de Puntuación , Unión Proteica , Elementos de Respuesta/genética
16.
PLoS One ; 5(2): e9443, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20195470

RESUMEN

BACKGROUND: Epstein Barr virus (EBV) infects the majority of the human population, causing fatal diseases in a small proportion in conjunction with environmental factors. Following primary infection, EBV remains latent in the memory B cell population for life. Recurrent reactivation of the virus occurs, probably due to activation of the memory B-lymphocytes, resulting in viral replication and re-infection of B-lymphocytes. Methylation of the viral DNA at CpG motifs leads to silencing of viral gene expression during latency. Zta, the key viral protein that mediates the latency/reactivation balance, interacts with methylated DNA. Zta is a transcription factor for both viral and host genes. A sub-set of its DNA binding sites (ZREs) contains a CpG motif, which is recognised in its methylated form. Detailed analysis of the promoter of the viral gene BRLF1 revealed that interaction with a methylated CpG ZRE (RpZRE3) is key to overturning the epigenetic silencing of the gene. METHODOLOGY AND PRINCIPAL FINDINGS: Here we question whether we can use this information to identify which host genes contain promoters with similar response elements. A computational search of human gene promoters identified 274 targets containing the 7-nucleotide RpZRE3 core element. DNA binding analysis of Zta with 17 of these targets revealed that the flanking context of the core element does not have a profound effect on the ability of Zta to interact with the methylated sites. A second juxtaposed ZRE was observed for one promoter. Zta was able to interact with this site, although co-occupancy with the RpZRE3 core element was not observed. CONCLUSIONS/SIGNIFICANCE: This research demonstrates 274 human promoters have the potential to be regulated by Zta to overturn epigenetic silencing of gene expression during viral reactivation from latency.


Asunto(s)
Epigénesis Genética , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/genética , Neoplasias/virología , Regiones Promotoras Genéticas/genética , Elementos de Respuesta/genética , Secuencia de Bases , Sitios de Unión/genética , Biología Computacional/métodos , Islas de CpG/genética , Metilación de ADN , Infecciones por Virus de Epstein-Barr/genética , Regulación Viral de la Expresión Génica , Genoma Humano/genética , Herpesvirus Humano 4/metabolismo , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Humanos , Neoplasias/genética , Unión Proteica , Homología de Secuencia de Ácido Nucleico , Transactivadores/genética , Transactivadores/metabolismo
17.
J Gen Virol ; 90(Pt 6): 1450-1454, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19264650

RESUMEN

Activation of the host gene egr1 is essential for the lytic replication of Epstein-Barr virus (EBV). egr1 is activated by Zta (BZLF1, ZEBRA). Zta interacts directly with DNA through a series of closely related Zta-response elements (ZREs). Here we dissect the mechanism used by Zta to interact with the egr1 promoter and identify a weak interaction with egr1ZRE that is dependent on the distal part of egr1ZRE. Furthermore, we demonstrate that the ability of Zta to interact with egr1ZRE is enhanced at least tenfold by methylation. The ability of Zta to transactivate a reporter construct driven by the egr1 promoter can be enhanced by methylation. As the ability of Zta to interact with a methylated ZRE in the EBV genome correlates with its ability to activate the expression of the endogenous viral gene BRLF1, this suggests that Zta may also have the capability to overturn epigenetic control of egr1.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/biosíntesis , Herpesvirus Humano 4/fisiología , Transactivadores/metabolismo , Replicación Viral , Sitios de Unión , Línea Celular , ADN/metabolismo , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Genes Reporteros , Humanos , Metilación , Regiones Promotoras Genéticas , Unión Proteica
18.
J Virol ; 81(13): 7149-55, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17459922

RESUMEN

The Epstein-Barr virus transcription factor Zta (encoded by BZLF1) is a bZIP protein containing an alpha-helical coiled-coil homodimerization motif (zipper). The Zta zipper forms less-stable dimers than other bZIP proteins, and an adjacent region (CT) interacts with the zipper to form a novel structure that is proposed to strengthen the dimer. Here we question the role of the CT region for Zta function. Cross-linking experiments demonstrate that the entire CT region lies adjacent to the zipper. Detailed analyses of Zta truncation mutations identify an involvement of the proximal CT region (221 to 230) in dimer formation with a further contribution from the distal region (236 to 243). Biophysical analyses reveal that residues 221 to 230 enhance the stability of the coiled coil. The ability of the Zta truncation mutants to interact with three Zta-binding sites also requires the proximal CT region. Fine mapping of DNA-binding requirements highlighted the contribution of these amino acids for Zta function. Thus, the proximal part of the CT region is required to aid the dimerization of Zta and thereby its DNA-binding ability. In contrast, although the distal part of the CT region aids dimerization, it promotes only a modest increase in DNA binding. To probe this further, we defined the contribution from the CT region for Zta to transactivate a promoter embedded within the viral genome. From this we conclude that the proximal part of the CT region is absolutely required, whereas the distal part is dispensable.


Asunto(s)
ADN Viral/metabolismo , Proteínas de Unión al ADN/metabolismo , Herpesvirus Humano 4/fisiología , Regiones Promotoras Genéticas/fisiología , Transactivadores/metabolismo , Transcripción Genética/fisiología , Activación Transcripcional/fisiología , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos/genética , Animales , Línea Celular , ADN Viral/química , ADN Viral/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Dimerización , Genoma Viral/fisiología , Herpesvirus Humano 4/química , Humanos , Eliminación de Secuencia , Transactivadores/química , Transactivadores/genética , Proteínas Virales/química , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...