#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Role of DNA Methylation and Epigenetic Silencing of in Endometrial Cancer Development


Background:
Endometrial cancer incidence is continuing to rise in the wake of the current ageing and obesity epidemics. Much of the risk for endometrial cancer development is influenced by the environment and lifestyle. Accumulating evidence suggests that the epigenome serves as the interface between the genome and the environment and that hypermethylation of stem cell polycomb group target genes is an epigenetic hallmark of cancer. The objective of this study was to determine the functional role of epigenetic factors in endometrial cancer development.

Methods and Findings:
Epigenome-wide methylation analysis of >27,000 CpG sites in endometrial cancer tissue samples (n = 64) and control samples (n = 23) revealed that HAND2 (a gene encoding a transcription factor expressed in the endometrial stroma) is one of the most commonly hypermethylated and silenced genes in endometrial cancer. A novel integrative epigenome-transcriptome-interactome analysis further revealed that HAND2 is the hub of the most highly ranked differential methylation hotspot in endometrial cancer. These findings were validated using candidate gene methylation analysis in multiple clinical sample sets of tissue samples from a total of 272 additional women. Increased HAND2 methylation was a feature of premalignant endometrial lesions and was seen to parallel a decrease in RNA and protein levels. Furthermore, women with high endometrial HAND2 methylation in their premalignant lesions were less likely to respond to progesterone treatment. HAND2 methylation analysis of endometrial secretions collected using high vaginal swabs taken from women with postmenopausal bleeding specifically identified those patients with early stage endometrial cancer with both high sensitivity and high specificity (receiver operating characteristics area under the curve = 0.91 for stage 1A and 0.97 for higher than stage 1A). Finally, mice harbouring a Hand2 knock-out specifically in their endometrium were shown to develop precancerous endometrial lesions with increasing age, and these lesions also demonstrated a lack of PTEN expression.

Conclusions:
HAND2 methylation is a common and crucial molecular alteration in endometrial cancer that could potentially be employed as a biomarker for early detection of endometrial cancer and as a predictor of treatment response. The true clinical utility of HAND2 DNA methylation, however, requires further validation in prospective studies.

Please see later in the article for the Editors' Summary


Vyšlo v časopise: Role of DNA Methylation and Epigenetic Silencing of in Endometrial Cancer Development. PLoS Med 10(11): e32767. doi:10.1371/journal.pmed.1001551
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pmed.1001551

Souhrn

Background:
Endometrial cancer incidence is continuing to rise in the wake of the current ageing and obesity epidemics. Much of the risk for endometrial cancer development is influenced by the environment and lifestyle. Accumulating evidence suggests that the epigenome serves as the interface between the genome and the environment and that hypermethylation of stem cell polycomb group target genes is an epigenetic hallmark of cancer. The objective of this study was to determine the functional role of epigenetic factors in endometrial cancer development.

Methods and Findings:
Epigenome-wide methylation analysis of >27,000 CpG sites in endometrial cancer tissue samples (n = 64) and control samples (n = 23) revealed that HAND2 (a gene encoding a transcription factor expressed in the endometrial stroma) is one of the most commonly hypermethylated and silenced genes in endometrial cancer. A novel integrative epigenome-transcriptome-interactome analysis further revealed that HAND2 is the hub of the most highly ranked differential methylation hotspot in endometrial cancer. These findings were validated using candidate gene methylation analysis in multiple clinical sample sets of tissue samples from a total of 272 additional women. Increased HAND2 methylation was a feature of premalignant endometrial lesions and was seen to parallel a decrease in RNA and protein levels. Furthermore, women with high endometrial HAND2 methylation in their premalignant lesions were less likely to respond to progesterone treatment. HAND2 methylation analysis of endometrial secretions collected using high vaginal swabs taken from women with postmenopausal bleeding specifically identified those patients with early stage endometrial cancer with both high sensitivity and high specificity (receiver operating characteristics area under the curve = 0.91 for stage 1A and 0.97 for higher than stage 1A). Finally, mice harbouring a Hand2 knock-out specifically in their endometrium were shown to develop precancerous endometrial lesions with increasing age, and these lesions also demonstrated a lack of PTEN expression.

Conclusions:
HAND2 methylation is a common and crucial molecular alteration in endometrial cancer that could potentially be employed as a biomarker for early detection of endometrial cancer and as a predictor of treatment response. The true clinical utility of HAND2 DNA methylation, however, requires further validation in prospective studies.

Please see later in the article for the Editors' Summary


Zdroje

1. FeilR, FragaMF (2011) Epigenetics and the environment: emerging patterns and implications. Nat Rev Genet 13: 97–109 doi:10.1038/nrg3142

2. JirtleRL, SkinnerMK (2007) Environmental epigenomics and disease susceptibility. Nat Rev Genet 8: 253–262.

3. SuvaML, RiggiN, BernsteinBE (2013) Epigenetic reprogramming in cancer. Science 339: 1567–1570 doi:10.1126/science.1230184

4. ShenH, LairdPW (2013) Interplay between the cancer genome and epigenome. Cell 153: 38–55.

5. TeschendorffAE, MenonU, Gentry-MaharajA, RamusSJ, WeisenbergerDJ, et al. (2010) Age-dependent DNA methylation of genes that are suppressed in stem cells is a hallmark of cancer. Genome Res 20: 440–446.

6. TeschendorffAE, JonesA, FieglH, SargentA, ZhuangJJ, et al. (2012) Epigenetic variability in cells of normal cytology is associated with the risk of future morphological transformation. Genome Med 4: 24 doi:10.1186/gm323

7. ZhuangJ, JonesA, LeeSH, NgE, FieglH, et al. (2012) The dynamics and prognostic potential of DNA methylation changes at stem cell gene loci in women's cancer. PLoS Genet 8: e1002517 doi:10.1371/journal.pgen.1002517

8. BeermanI, BockC, GarrisonBS, SmithZD, GuH, et al. (2013) Proliferation-dependent alterations of the DNA methylation landscape underlie hematopoietic stem cell aging. Cell Stem Cell 12: 413–425 doi:10.1016/j.stem.2013.01.017

9. BrackenAP, HelinK (2009) Polycomb group proteins: navigators of lineage pathways led astray in cancer. Nat Rev Cancer 9: 773–784 doi:10.1038/nrc2736

10. EaswaranH, JohnstoneSE, VanNL, OhmJ, MosbrugerT, et al. (2012) A DNA hypermethylation module for the stem/progenitor cell signature of cancer. Genome Res 22: 837–849 doi:10.1101/gr.131169.111

11. LichtensteinP, HolmNV, VerkasaloPK, IliadouA, KaprioJ, et al. (2000) Environmental and heritable factors in the causation of cancer—analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl J Med 343: 78–85.

12. SchoutenLJ, GoldbohmRA, van den BrandtPA (2004) Anthropometry, physical activity, and endometrial cancer risk: results from the Netherlands Cohort Study. J Natl Cancer Inst 96: 1635–1638 doi:10.1093/jnci/djh291

13. AmantF, MoermanP, NevenP, TimmermanD, VanLE, et al. (2005) Endometrial cancer. Lancet 366: 491–505 doi:10.1016/S0140-6736(05)67063-8

14. YangS, ThielKW, LeslieKK (2011) Progesterone: the ultimate endometrial tumor suppressor. Trends Endocrinol Metab 22: 145–152 doi:10.1016/j.tem.2011.01.005

15. KuritaT, YoungP, BrodyJR, LydonJP, O'MalleyBW, et al. (1998) Stromal progesterone receptors mediate the inhibitory effects of progesterone on estrogen-induced uterine epithelial cell deoxyribonucleic acid synthesis. Endocrinology 139: 4708–4713.

16. SalvesenHB, CarterSL, MannelqvistM, DuttA, GetzG, et al. (2009) Integrated genomic profiling of endometrial carcinoma associates aggressive tumors with indicators of PI3 kinase activation. Proc Natl Acad Sci U S A 106: 4834–4839.

17. WiegandKC, ShahSP, Al-AghaOM, ZhaoY, TseK, et al. (2010) ARID1A mutations in endometriosis-associated ovarian carcinomas. N Engl J Med 363: 1532–1543 doi:10.1056/NEJMoa1008433

18. WikE, TrovikJ, IversenOE, EngelsenIB, StefanssonIM, et al. (2009) Deoxyribonucleic acid ploidy in endometrial carcinoma: a reproducible and valid prognostic marker in a routine diagnostic setting. Am J Obstet Gynecol 201: 603–607 doi:10.1016/j.ajog.2009.07.029

19. DayRS, McDadeKK, ChandranUR, LisovichA, ConradsTP, et al. (2011) Identifier mapping performance for integrating transcriptomics and proteomics experimental results. BMC Bioinformatics 12: 213 doi:10.1186/1471-2105-12-213

20. TaoMH, FreudenheimJL (2010) DNA methylation in endometrial cancer. Epigenetics 5: 491–498 pii:12431.

21. WidschwendterM, FieglH, EgleD, Mueller-HolznerE, SpizzoG, et al. (2007) Epigenetic stem cell signature in cancer. Nat Genet 39: 157–158.

22. WidschwendterM, SiegmundKD, MullerHM, FieglH, MarthC, et al. (2004) Association of breast cancer DNA methylation profiles with hormone receptor status and response to tamoxifen. Cancer Res 64: 3807–3813 doi:10.1158/0008-5472.CAN-03-3852

23. LiQ, KannanA, DeMayoFJ, LydonJP, CookePS, et al. (2011) The antiproliferative action of progesterone in uterine epithelium is mediated by Hand2. Science 331: 912–916 doi:10.1126/science.1197454

24. StoreyJD, TibshiraniR (2003) Statistical significance for genomewide studies. Proc Natl Acad Sci U S A 100: 9440–9445.

25. SubramanianA, TamayoP, MoothaVK, MukherjeeS, EbertBL, et al. (2005) Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 102: 15545–15550.

26. LeeTI, JennerRG, BoyerLA, GuentherMG, LevineSS, et al. (2006) Control of developmental regulators by Polycomb in human embryonic stem cells. Cell 125: 301–313.

27. HochbergY, BenjaminiY (1990) More powerful procedures for multiple significance testing. Stat Med 9: 811–818.

28. WestJ, BeckS, WangX, TeschendorffAE (2013) An integrative network algorithm identifies age-associated differential methylation interactome hotspots targeting stem-cell differentiation pathways. Sci Rep 3: 1630 doi:10.1038/srep01630

29. KatoHD, KondohH, InoueT, AsanomaK, MatsudaT, et al. (2004) Expression of DCC and netrin-1 in normal human endometrium and its implication in endometrial carcinogenesis. Gynecol Oncol 95: 281–289 doi:10.1016/j.ygyno.2004.07.050

30. SrivastavaD, ThomasT, LinQ, KirbyML, BrownD, et al. (1997) Regulation of cardiac mesodermal and neural crest development by the bHLH transcription factor, dHAND. Nat Genet 16: 154–160 doi:10.1038/ng0697-154

31. BagchiIC, LiQ, CheonYP, MantenaSR, KannanA, et al. (2005) Use of the progesterone receptor antagonist RU 486 to identify novel progesterone receptor-regulated pathways in implantation. Semin Reprod Med 23: 38–45 doi:10.1055/s-2005-864032

32. DassenH, PunyadeeraC, KampsR, KlompJ, DunselmanG, et al. (2007) Progesterone regulation of implantation-related genes: new insights into the role of oestrogen. Cell Mol Life Sci 64: 1009–1032 doi:10.1007/s00018-007-6553-9

33. LeitaoMMJr, HanG, LeeLX, Abu-RustumNR, BrownCL, et al. (2010) Complex atypical hyperplasia of the uterus: characteristics and prediction of underlying carcinoma risk. Am J Obstet Gynecol 203: 349–346 doi:10.1016/j.ajog.2010.05.004

34. TurnerN, GroseR (2010) Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer 10: 116–129 doi:10.1038/nrc2780

35. HollanderMC, BlumenthalGM, DennisPA (2011) PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nat Rev Cancer 11: 289–301 doi:10.1038/nrc3037

36. ButtittaF, MarchettiA, AthanassiadouS, GadducciA, BellinaS, et al. (1997) p53 nuclear accumulation in preneoplastic lesions and stage I uterine endometrioid adenocarcinoma. Oncol Rep 4: 315–318.

37. WescheJ, HaglundK, HaugstenEM (2011) Fibroblast growth factors and their receptors in cancer. Biochem J 437: 199–213 doi:10.1042/BJ20101603

38. HayesMP, WangH, Espinal-WitterR, DouglasW, SolomonGJ, et al. (2006) PIK3CA and PTEN mutations in uterine endometrioid carcinoma and complex atypical hyperplasia. Clin Cancer Res 12: 5932–5935 doi:10.1158/1078-0432.CCR-06-1375

39. YeramianA, Moreno-BuenoG, DolcetX, CatasusL, AbalM, et al. (2012) Endometrial carcinoma: molecular alterations involved in tumor development and progression. Oncogene 32: 403–413 doi:10.1038/onc.2012.76

40. OhmJE, McGarveyKM, YuX, ChengL, SchuebelKE, et al. (2007) A stem cell-like chromatin pattern may predispose tumor suppressor genes to DNA hypermethylation and heritable silencing. Nat Genet 39: 237–242.

41. OhmJE, BaylinSB (2007) Stem cell chromatin patterns: an instructive mechanism for DNA hypermethylation? Cell Cycle 6: 1040–1043 pii:4210.

42. SchlesingerY, StraussmanR, KeshetI, FarkashS, HechtM, et al. (2007) Polycomb-mediated methylation on Lys27 of histone H3 pre-marks genes for de novo methylation in cancer. Nat Genet 39: 232–236.

43. WidschwendterM, JonesA, TeschendorffAE (2013) Epigenetics makes its mark on women-specific cancers-an opportunity to redefine oncological approaches? Gynecol Oncol 128: 134–143 doi:10.1016/j.ygyno.2012.09.027

Štítky
Interné lekárstvo

Článok vyšiel v časopise

PLOS Medicine


2013 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#