A METILAÇÃO DO DNA COMO BIOMARCADOR NO PROGNÓSTICO E DIAGNÓSTICO DO CÂNCER OVARIANO: REVISÃO DE LITERATURA

2021 ◽  
Author(s):  
Manuella Amlid Pimenta de Castro Cavalcanti Silva ◽  
Caio Victor Barros Gonçalves da Silva ◽  
Dryelli Frances Santana da Silva ◽  
Laura Lucena Serafim ◽  
Ryan Cristian da Silva ◽  
...  

Introdução: O câncer ovariano é caracterizado pelo crescimento anormal de células provenientes dos ovários. Mutações no gene BRCA podem aumentar, entre 20 a 50%, o risco do paciente desenvolver câncer ovariano (1). Assim, o gene BRCA1 é expresso como autossômico dominante, com penetrância incompleta e atua como supressor de tumor, sendo importante para a reparação do DNA, pois atua na manutenção da estabilidade do genoma e no controle do ciclo celular no checkpoint (3). Desse modo, a metilação do DNA ocorre na região promotora do gene, local dos sítios CpG – sendo denominados assim pois um grupo metil é adicionado a uma citosina seguida de uma guanina – e grupos metil são adicionados ao nucleotídeo citosina (1). Portanto, a metilação do DNA é essencial na regulação da expressão gênica (3). No câncer ovariano, anomalias nesta metilação são comuns, sendo potenciais biomarcadores na detecção desta doença. Objetivos: Realizar uma revisão narrativa sobre a metilação do DNA, que ocorre nos sítios CpG do gene BRCA1, ser um importante biomarcador no prognóstico e diagnóstico do câncer ovariano. Métodos: Para esta revisão narrativa, foi realizado buscas através do Pubmed e Google Scholar,através dos descritores: “ovarian cancer”, “BRCA1”, “DNA methylation” e “biomarker”. Como critérios para a pesquisa, foram selecionados artigos de língua inglesa e publicados entre os anos de 2016 e 2021. Resultados: A análise da metilação do DNA permite a identificação de alterações na região promotora do gene, nos sítios CpG que podem estar hipermetilados – quando há o silenciamento do gene BRCA1 – ou hipometilados – quando não há o silenciamento do gene BRCA1 (2). Devido ao envolvimento da metilação do DNA com o câncer ovariano, a metilação nos sítios CpG no gene BRCA1 apresenta um alto potencial como biomarcador. Portanto, analisar anomalias na metilação do DNA, nos sítios CpG, significa que não seria necessário codificar todo o gene para identificar se há uma mutação no gene BRCA1. Um dos métodos mais conhecidos e mais utilizados é o método que envolve o bissulfito na conversão de citosina em uracila. Citosinas metiladas não serão convertidas e inúmeros métodos podem ser utilizados na identificação de CpGs que não foram convertidas (1). Conclusão: Alterações na metilação do DNA, nos sítios CpG do gene BRCA1, apresentam um alto potencial como biomarcadores, mesmo sendo algo novo na área. Portanto, mais estudos são necessários para aperfeiçoar o entendimento deste biomarcador como prognóstico e diagnóstico do câncer ovariano.

2019 ◽  
Vol 11 (1) ◽  
Author(s):  
Hua Tian ◽  
Li Yan ◽  
Li Xiao-fei ◽  
Sun Hai-yan ◽  
Chen Juan ◽  
...  

Abstract Purpose One major reason of the high mortality of epithelial ovarian cancer (EOC) is due to platinum-based chemotherapy resistance. Aberrant DNA methylation may be a potential mechanism underlying the development of platinum resistance in EOC. The purpose of this study is to discover potential aberrant DNA methylation that contributes to drug resistance. Methods By initially screening of 16 platinum-sensitive/resistant samples from EOC patients with reduced representation bisulfite sequencing (RRBS), the upstream region of the hMSH2 gene was discovered hypermethylated in the platinum-resistant group. The effect of hMSH2 methylation on the cellular response to cisplatin was explored by demethylation and knockdown assays in ovarian cancer cell line A2780. Matrix-assisted laser desorption ionization time-of-flight (MALDI-TOF) mass spectrometry was employed to examine the methylation levels of hMSH2 upstream region in additional 40 EOC patient samples. RT-qPCR and IHC assay was used to detect the hMSH2 mRNA and protein expression in extended 150 patients. Results RRBS assay discovered an upstream region from − 1193 to − 1125 of hMSH2 was significant hypermethylated in resistant EOC patients (P = 1.06 × 10−14). In vitro analysis demonstrated that global demethylation increased cisplatin sensitivity along with a higher expression of the hMSH2 mRNA and protein. Knockdown hMSH2 reduced the cell sensitivity to cisplatin. MALDI-TOF mass spectrometry assay validated the strong association of hypermethylation of hMSH2 upstream region with platinum resistance. Spearman’s correlation analysis revealed a significantly negative connection between methylation level of hMSH2 upstream region and its expression. The Kaplan-Meier analyses showed the high methylation of hMSH2 promoter region, and its low expressions are associated with worse survival. In multivariable models, hMSH2 low expression was an independent factor predicting poor outcome (P = 0.03, HR = 1.91, 95%CI = 1.85–2.31). Conclusion The hypermethylation of hMSH2 upstream region is associated with platinum resistant in EOC, and low expression of hMSH2 may be an index for the poor prognosis.


2008 ◽  
Vol 123 (3) ◽  
pp. 725-729 ◽  
Author(s):  
Heidi Fiegl ◽  
Gudrun Windbichler ◽  
Elisabeth Mueller-Holzner ◽  
Georg Goebel ◽  
Matthias Lechner ◽  
...  

2021 ◽  
Vol 12 ◽  
Author(s):  
Min Zhou ◽  
Shasha Hong ◽  
Bingshu Li ◽  
Cheng Liu ◽  
Ming Hu ◽  
...  

Background: DNA methylation affects the development, progression, and prognosis of various cancers. This study aimed to identify DNA methylated-differentially expressed genes (DEGs) and develop a methylation-driven gene model to evaluate the prognosis of ovarian cancer (OC).Methods: DNA methylation and mRNA expression profiles of OC patients were downloaded from The Cancer Genome Atlas, Genotype-Tissue Expression, and Gene Expression Omnibus databases. We used the R package MethylMix to identify DNA methylation-regulated DEGs and built a prognostic signature using LASSO Cox regression. A quantitative nomogram was then drawn based on the risk score and clinicopathological features.Results: We identified 56 methylation-related DEGs and constructed a prognostic risk signature with four genes according to the LASSO Cox regression algorithm. A higher risk score not only predicted poor prognosis, but also was an independent poor prognostic indicator, which was validated by receiver operating characteristic (ROC) curves and the validation cohort. A nomogram consisting of the risk score, age, FIGO stage, and tumor status was generated to predict 3- and 5-year overall survival (OS) in the training cohort. The joint survival analysis of DNA methylation and mRNA expression demonstrated that the two genes may serve as independent prognostic biomarkers for OS in OC.Conclusion: The established qualitative risk score model was found to be robust for evaluating individualized prognosis of OC and in guiding therapy.


2021 ◽  
Vol 13 (1) ◽  
Author(s):  
David W. Chan ◽  
Wai-Yip Lam ◽  
Fushun Chen ◽  
Mingo M. H. Yung ◽  
Yau-Sang Chan ◽  
...  

Abstract Background In contrast to stable genetic events, epigenetic changes are highly plastic and play crucial roles in tumor evolution and development. Epithelial ovarian cancer (EOC) is a highly heterogeneous disease that is generally associated with poor prognosis and treatment failure. Profiling epigenome-wide DNA methylation status is therefore essential to better characterize the impact of epigenetic alterations on the heterogeneity of EOC. Methods An epigenome-wide association study was conducted to evaluate global DNA methylation in a retrospective cohort of 80 mixed subtypes of primary ovarian cancers and 30 patients with high-grade serous ovarian carcinoma (HGSOC). Three demethylating agents, azacytidine, decitabine, and thioguanine, were tested their anti-cancer and anti-chemoresistant effects on HGSOC cells. Results Global DNA hypermethylation was significantly associated with high-grade tumors, platinum resistance, and poor prognosis. We determined that 9313 differentially methylated probes (DMPs) were enriched in their relative gene regions of 4938 genes involved in small GTPases and were significantly correlated with the PI3K-AKT, MAPK, RAS, and WNT oncogenic pathways. On the other hand, global DNA hypermethylation was preferentially associated with recurrent HGSOC. A total of 2969 DMPs corresponding to 1471 genes were involved in olfactory transduction, and calcium and cAMP signaling. Co-treatment with demethylating agents showed significant growth retardation in ovarian cancer cells through differential inductions, such as cell apoptosis by azacytidine or G2/M cell cycle arrest by decitabine and thioguanine. Notably, azacytidine and decitabine, though not thioguanine, synergistically enhanced cisplatin-mediated cytotoxicity in HGSOC cells. Conclusions This study demonstrates the significant association of global hypermethylation with poor prognosis and drug resistance in high-grade EOC and highlights the potential of demethylating agents in cancer treatment. Graphic abstract


2016 ◽  
Vol 2 (5) ◽  
pp. 261 ◽  
Author(s):  
Yogita Lugani ◽  
Smita Asthana ◽  
Satyanarayana Labani

<p>Ovarian carcinoma is one of the leading causes of morbidity and mortality associated with carcinomas affecting women. It comprises a heterogeneous group of neoplasms that represents the seventh most lethal malignancy in women worldwide, and is a major cause of death from gynecological carcinoma. Specific to different geographical locations all over the globe, there are variations in the magnitude and trends of ovarian carcinoma, and the scenario of the disease keeps changing. As such, it is necessary to update and review the existing study on ovarian carcinoma. Reviews on ovarian cancer from 2000 to 2015 were extracted from PubMed and Google Scholar, and a few selected landmark studies that incorporated old data were also included. The focus of the present study is to consolidate an updated global view on epithelial ovarian carcinoma, the most prevalent type of ovarian carcinoma. This article covers the epidemiology, types, diagnosis, prognosis, and treatment of epithelial ovarian carcinoma.</p>


2020 ◽  
Vol 21 (5) ◽  
pp. 1848 ◽  
Author(s):  
Sadia Mehdi ◽  
Magdalena Bachvarova ◽  
Marie-Pier Scott-Boyer ◽  
Arnaud Droit ◽  
Dimcho Bachvarov

Growing evidence demonstrates that epithelial–mesenchymal transition (EMT) plays an important role in epithelial ovarian cancer (EOC) progression and spreading; however, its molecular mechanisms remain poorly defined. We have previously shown that the antigen receptor LY75 can modulate EOC cell phenotype and metastatic potential, as LY75 depletion directed mesenchymal–epithelial transition (MET) in EOC cell lines with mesenchymal phenotype. We used the LY75-mediated modulation of EMT as a model to investigate for DNA methylation changes during EMT in EOC cells, by applying the reduced representation bisulfite sequencing (RRBS) methodology. Numerous genes have displayed EMT-related DNA methylation patterns alterations in their promoter/exon regions. Ten selected genes, whose DNA methylation alterations were further confirmed by alternative methods, were further identified, some of which could represent new EOC biomarkers/therapeutic targets. Moreover, our methylation data were strongly indicative for the predominant implication of the Wnt/β-catenin pathway in the EMT-induced DNA methylation variations in EOC cells. Consecutive experiments, including alterations in the Wnt/β-catenin pathway activity in EOC cells with a specific inhibitor and the identification of LY75-interacting partners by a proteomic approach, were strongly indicative for the direct implication of the LY75 receptor in modulating the Wnt/β-catenin signaling in EOC cells.


2020 ◽  
Vol 12 (1) ◽  
Author(s):  
Pietro Lo Riso ◽  
Carlo Emanuele Villa ◽  
Gilles Gasparoni ◽  
Andrea Vingiani ◽  
Raffaele Luongo ◽  
...  

Abstract Background High-grade serous ovarian cancer (HGSOC) is a major unmet need in oncology. The remaining uncertainty on its originating tissue has hampered the discovery of molecular oncogenic pathways and the development of effective therapies. Methods We used an approach based on the retention in tumors of a DNA methylation trace (OriPrint) that distinguishes the two putative tissues of origin of HGSOC, the fimbrial (FI) and ovarian surface epithelia (OSE), to stratify HGSOC by several clustering methods, both linear and non-linear. The identified tumor subtypes (FI-like and OSE-like HGSOC) were investigated at the RNAseq level to stratify an in-house cohort of macrodissected HGSOC FFPE samples to derive overall and disease-free survival and identify specific transcriptional alterations of the two tumor subtypes, both by classical differential expression and weighted correlation network analysis. We translated our strategy to published datasets and verified the co-occurrence of previously described molecular classification of HGSOC. We performed cytokine analysis coupled to immune phenotyping to verify alterations in the immune compartment associated with HGSOC. We identified genes that are both differentially expressed and methylated in the two tumor subtypes, concentrating on PAX8 as a bona fide marker of FI-like HGSOC. Results We show that: - OriPrint is a robust DNA methylation tracer that exposes the tissue of origin of HGSOC. - The tissue of origin of HGSOC is the main determinant of DNA methylation variance in HGSOC. - The tissue of origin is a prognostic factor for HGSOC patients. - FI-like and OSE-like HGSOC are endowed with specific transcriptional alterations that impact patients’ prognosis. - OSE-like tumors present a more invasive and immunomodulatory phenotype, compatible with its worse prognostic impact. - Among genes that are differentially expressed and regulated in FI-like and OSE-like HGSOC, PAX8 is a bona fide marker of FI-like tumors. Conclusions Through an integrated approach, our work demonstrates that both FI and OSE are possible origins for human HGSOC, whose derived subtypes are both molecularly and clinically distinct. These results will help define a new roadmap towards rational, subtype-specific therapeutic inroads and improved patients’ care.


2011 ◽  
Vol 412 (15-16) ◽  
pp. 1472-1475 ◽  
Author(s):  
Rémy Bosviel ◽  
Emilie Michard ◽  
Guillaume Lavediaux ◽  
Fabrice Kwiatkowski ◽  
Yves-Jean Bignon ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document