#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

DNA–Methylome Analysis of Mouse Intestinal Adenoma Identifies a Tumour-Specific Signature That Is Partly Conserved in Human Colon Cancer


Aberrant CpG methylation is a universal epigenetic trait of cancer cell genomes. However, human cancer samples or cell lines preclude the investigation of epigenetic changes occurring early during tumour development. Here, we have used MeDIP-seq to analyse the DNA methylome of APCMin adenoma as a model for intestinal cancer initiation, and we present a list of more than 13,000 recurring differentially methylated regions (DMRs) characterizing intestinal adenoma of the mouse. We show that Polycomb Repressive Complex (PRC) targets are strongly enriched among hypermethylated DMRs, and several PRC2 components and DNA methyltransferases were up-regulated in adenoma. We further demonstrate by bisulfite pyrosequencing of purified cell populations that the DMR signature arises de novo in adenoma cells rather than by expansion of a pre-existing pattern in intestinal stem cells or undifferentiated crypt cells. We found that epigenetic silencing of tumour suppressors, which occurs frequently in colon cancer, was rare in adenoma. Quite strikingly, we identified a core set of DMRs, which is conserved between mouse adenoma and human colon cancer, thus possibly revealing a global panel of epigenetically modified genes for intestinal tumours. Our data allow a distinction between early conserved epigenetic alterations occurring in intestinal adenoma and late stochastic events promoting colon cancer progression, and may facilitate the selection of more specific clinical epigenetic biomarkers.


Vyšlo v časopise: DNA–Methylome Analysis of Mouse Intestinal Adenoma Identifies a Tumour-Specific Signature That Is Partly Conserved in Human Colon Cancer. PLoS Genet 9(2): e32767. doi:10.1371/journal.pgen.1003250
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003250

Souhrn

Aberrant CpG methylation is a universal epigenetic trait of cancer cell genomes. However, human cancer samples or cell lines preclude the investigation of epigenetic changes occurring early during tumour development. Here, we have used MeDIP-seq to analyse the DNA methylome of APCMin adenoma as a model for intestinal cancer initiation, and we present a list of more than 13,000 recurring differentially methylated regions (DMRs) characterizing intestinal adenoma of the mouse. We show that Polycomb Repressive Complex (PRC) targets are strongly enriched among hypermethylated DMRs, and several PRC2 components and DNA methyltransferases were up-regulated in adenoma. We further demonstrate by bisulfite pyrosequencing of purified cell populations that the DMR signature arises de novo in adenoma cells rather than by expansion of a pre-existing pattern in intestinal stem cells or undifferentiated crypt cells. We found that epigenetic silencing of tumour suppressors, which occurs frequently in colon cancer, was rare in adenoma. Quite strikingly, we identified a core set of DMRs, which is conserved between mouse adenoma and human colon cancer, thus possibly revealing a global panel of epigenetically modified genes for intestinal tumours. Our data allow a distinction between early conserved epigenetic alterations occurring in intestinal adenoma and late stochastic events promoting colon cancer progression, and may facilitate the selection of more specific clinical epigenetic biomarkers.


Zdroje

1. MargueronR, ReinbergD (2010) Chromatin structure and the inheritance of epigenetic information. Nat Rev Genet 11: 285–296 doi:10.1038/nrg2752.

2. CedarH, BergmanY (2009) Linking DNA methylation and histone modification: patterns and paradigms. Nat Rev Genet 10: 295–304 doi:10.1038/nrg2540.

3. LundAH, van LohuizenM (2004) Polycomb complexes and silencing mechanisms. Curr Opin Cell Biol 16: 239–246 doi:10.1016/j.ceb.2004.03.010.

4. ViréE, BrennerC, DeplusR, BlanchonL, FragaM, et al. (2006) The Polycomb group protein EZH2 directly controls DNA methylation. Nature 439: 871–874 doi:10.1038/nature04431.

5. RazinA, ShemerR (1995) DNA methylation in early development. Hum Mol Genet 4 Spec No: 1751–1755.

6. WeberM, DaviesJJ, WittigD, OakeleyEJ, HaaseM, et al. (2005) Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells. Nat Genet 37: 853–862 doi:10.1038/ng1598.

7. KeshetI, SchlesingerY, FarkashS, RandE, HechtM, et al. (2006) Evidence for an instructive mechanism of de novo methylation in cancer cells. Nat Genet 38: 149–153 doi:10.1038/ng1719.

8. JiaD, JurkowskaRZ, ZhangX, JeltschA, ChengX (2007) Structure of Dnmt3a bound to Dnmt3L suggests a model for de novo DNA methylation. Nature 449: 248–251 doi:10.1038/nature06146.

9. OoiSKT, QiuC, BernsteinE, LiK, JiaD, et al. (2007) DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448: 714–717 doi:10.1038/nature05987.

10. JonesPA, BaylinSB (2007) The epigenomics of cancer. Cell 128: 683–692 doi:10.1016/j.cell.2007.01.029.

11. EstellerM (2008) Epigenetics in cancer. N Engl J Med 358: 1148–1159 doi:10.1056/NEJMra072067.

12. BaylinSB (2009) Stem cells, cancer, and epigenetics. StemBook 1–14 doi:10.3824/stembook.1.50.1.

13. JonesPA, BaylinSB (2002) The fundamental role of epigenetic events in cancer. Nat Rev Genet 3: 415–428 doi:10.1038/nrg816.

14. WidschwendterM, FieglH, EgleD, Mueller-HolznerE, SpizzoG, et al. (2007) Epigenetic stem cell signature in cancer. Nat Genet 39: 157–158 doi:10.1038/ng1941.

15. 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 doi:10.1038/ng1950.

16. 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 doi:10.1038/ng1972.

17. ZhuangJ, JonesA, LeeS-H, 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.

18. EaswaranH, JohnstoneSE, Van NesteL, 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.

19. SparmannA, van LohuizenM (2006) Polycomb silencers control cell fate, development and cancer. Nat Rev Cancer 6: 846–856 doi:10.1038/nrc1991.

20. MillsAA (2010) Throwing the cancer switch: reciprocal roles of polycomb and trithorax proteins. Nat Rev Cancer 10: 669–682 doi:10.1038/nrc2931.

21. SchuettengruberB, MartinezA-M, IovinoN, CavalliG (2011) Trithorax group proteins: switching genes on and keeping them active. Nat Rev Mol Cell Biol 12: 799–814 doi:10.1038/nrm3230.

22. FearonER, VogelsteinB (1990) A genetic model for colorectal tumorigenesis. Cell 61: 759–767.

23. SuLK, KinzlerKW, VogelsteinB, PreisingerAC, MoserAR, et al. (1992) Multiple intestinal neoplasia caused by a mutation in the murine homolog of the APC gene. Science 256: 668–670.

24. EstellerM (2007) Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet 8: 286–298 doi:10.1038/nrg2005.

25. LaoVV, GradyWM (2011) Epigenetics and colorectal cancer. Nat Rev Gastroenterol Hepatol doi:10.1038/nrgastro.2011.173.

26. DownTA, RakyanVK, TurnerDJ, FlicekP, LiH, et al. (2008) A Bayesian deconvolution strategy for immunoprecipitation-based DNA methylome analysis. Nat Biotechnol 26: 779–785 doi:10.1038/nbt1414.

27. ChavezL, JozefczukJ, GrimmC, DietrichJ, TimmermannB, et al. (2010) Computational analysis of genome-wide DNA methylation during the differentiation of human embryonic stem cells along the endodermal lineage. Genome Res 20: 1441–1450 doi:10.1101/gr.110114.110.

28. EstécioMRH, GharibyanV, ShenL, IbrahimAEK, DoshiK, et al. (2007) LINE-1 hypomethylation in cancer is highly variable and inversely correlated with microsatellite instability. PLoS ONE 2: e399 doi:10.1371/journal.pone.0000399.

29. SunamiE, de MaatM, VuA, TurnerRR, HoonDSB (2011) LINE-1 hypomethylation during primary colon cancer progression. PLoS ONE 6: e18884 doi:10.1371/journal.pone.0018884.

30. El-MaarriO (2004) SIRPH analysis: SNuPE with IP-RP-HPLC for quantitative measurements of DNA methylation at specific CpG sites. Methods Mol Biol 287: 195–205 doi:10.1385/1-59259-828-5:195.

31. BoyerLA, PlathK, ZeitlingerJ, BrambrinkT, MedeirosLA, et al. (2006) Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature 441: 349–353 doi:10.1038/nature04733.

32. Ben-PorathI, ThomsonMW, CareyVJ, GeR, BellGW, et al. (2008) An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet 40: 499–507 doi:10.1038/ng.127.

33. GuentherMG, LawtonLN, RozovskaiaT, FramptonGM, LevineSS, et al. (2008) Aberrant chromatin at genes encoding stem cell regulators in human mixed-lineage leukemia. Genes Dev 22: 3403–3408 doi:10.1101/gad.1741408.

34. WilliamsK, ChristensenJ, PedersenMT, JohansenJV, CloosPAC, et al. (2011) TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature 473: 343–348 doi:10.1038/nature10066.

35. 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 USA 102: 15545–15550 doi:10.1073/pnas.0506580102.

36. BarkerN, RidgwayRA, van EsJH, van de WeteringM, BegthelH, et al. (2009) Crypt stem cells as the cells-of-origin of intestinal cancer. Nature 457: 608–611 doi:10.1038/nature07602.

37. BarkerN, van EsJH, KuipersJ, KujalaP, van den BornM, et al. (2007) Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449: 1003–1007 doi:10.1038/nature06196.

38. DietrichWF, LanderES, SmithJS, MoserAR, GouldKA, et al. (1993) Genetic identification of Mom-1, a major modifier locus affecting Min-induced intestinal neoplasia in the mouse. Cell 75: 631–639.

39. HlubekF, BrabletzT, BudcziesJ, PfeifferS, JungA, et al. (2007) Heterogeneous expression of Wnt/beta-catenin target genes within colorectal cancer. Int J Cancer 121: 1941–1948 doi:10.1002/ijc.22916.

40. SatoT, van EsJH, SnippertHJ, StangeDE, VriesRG, et al. (2010) Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature 469: 415 doi:10.1038/nature09637.

41. Merlos-SuárezA, BarrigaFM, JungP, IglesiasM, CéspedesMV, et al. (2011) The intestinal stem cell signature identifies colorectal cancer stem cells and predicts disease relapse. Cell Stem Cell 8: 511–524 doi:10.1016/j.stem.2011.02.020.

42. FarrallAL, RiemerP, LeushackeM, SreekumarA, GrimmC, et al. (2012) Wnt and BMP signals control intestinal adenoma cell fates. Int J Cancer doi:10.1002/ijc.27500.

43. LiQL, ItoK, SakakuraC, FukamachiH, InoueKI, et al. (2002) Causal relationship between the loss of RUNX3 expression and gastric cancer. Cell 109: 113–124.

44. NishioM, SakakuraC, NagataT, KomiyamaS, MiyashitaA, et al. (2010) RUNX3 promoter methylation in colorectal cancer: its relationship with microsatellite instability and its suitability as a novel serum tumor marker. Anticancer Res 30: 2673–2682.

45. TanakaK, ImotoI, InoueJ, KozakiK, TsudaH, et al. (2007) Frequent methylation-associated silencing of a candidate tumor-suppressor, CRABP1, in esophageal squamous-cell carcinoma. Oncogene 26: 6456–6468 doi:10.1038/sj.onc.1210459.

46. GasparC, CardosoJ, FrankenP, MolenaarL, MorreauH, et al. (2008) Cross-species comparison of human and mouse intestinal polyps reveals conserved mechanisms in adenomatous polyposis coli (APC)-driven tumorigenesis. Am J Pathol 172: 1363–1380 doi:10.2353/ajpath.2008.070851.

47. JinB, YaoB, LiJ-L, FieldsCR, DelmasAL, et al. (2009) DNMT1 and DNMT3B modulate distinct polycomb-mediated histone modifications in colon cancer. Cancer Res 69: 7412–7421 doi:10.1158/0008-5472.CAN-09-0116.

48. FacklerMJ, UmbrichtCB, WilliamsD, ArganiP, CruzL-A, et al. (2011) Genome-wide methylation analysis identifies genes specific to breast cancer hormone receptor status and risk of recurrence. Cancer Res 71: 6195–6207 doi:10.1158/0008-5472.CAN-11-1630.

49. PeterzielH, MüllerJ, DannerA, BarbusS, LiuH-K, et al. (2012) Expression of podoplanin in human astrocytic brain tumors is controlled by the PI3K-AKT-AP-1 signaling pathway and promoter methylation. Neuro-oncology 14: 426–439 doi:10.1093/neuonc/nos055.

50. IzumiH, InoueJ, YokoiS, HosodaH, ShibataT, et al. (2005) Frequent silencing of DBC1 is by genetic or epigenetic mechanisms in non-small cell lung cancers. Hum Mol Genet 14: 997–1007 doi:10.1093/hmg/ddi092.

51. LuxenS, BelinskySA, KnausUG (2008) Silencing of DUOX NADPH oxidases by promoter hypermethylation in lung cancer. Cancer Res 68: 1037–1045 doi:10.1158/0008-5472.CAN-07-5782.

52. MiottoE, SabbioniS, VeroneseA, CalinGA, GulliniS, et al. (2004) Frequent aberrant methylation of the CDH4 gene promoter in human colorectal and gastric cancer. Cancer Res 64: 8156–8159 doi:10.1158/0008-5472.CAN-04-3000.

53. HarukiS, ImotoI, KozakiK-I, MatsuiT, KawachiH, et al. (2010) Frequent silencing of protocadherin 17, a candidate tumour suppressor for esophageal squamous cell carcinoma. Carcinogenesis 31: 1027–1036 doi:10.1093/carcin/bgq053.

54. LindGE, KleiviK, MelingGI, TeixeiraMR, Thiis-EvensenE, et al. (2006) ADAMTS1, CRABP1, and NR3C1 identified as epigenetically deregulated genes in colorectal tumorigenesis. Cell Oncol 28: 259–272.

55. MeissnerA, MikkelsenTS, GuH, WernigM, HannaJ, et al. (2008) Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature 454: 766–770 doi:10.1038/nature07107.

56. HalbergRB, WaggonerJ, RasmussenK, WhiteA, ClipsonL, et al. (2009) Long-lived Min mice develop advanced intestinal cancers through a genetically conservative pathway. Cancer Res 69: 5768–5775 doi:10.1158/0008-5472.CAN-09-0446.

57. SatoT, StangeDE, FerranteM, VriesRGJ, van EsJH, et al. (2011) Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett's epithelium. Gastroenterology 141: 1762–1772 doi:10.1053/j.gastro.2011.07.050.

58. GrimmC, AdjayeJ (2012) Analysis of the methylome of human embryonic stem cells employing methylated DNA immunoprecipitation coupled to next-generation sequencing. Methods Mol Biol 873: 281–295 doi:10.1007/978-1-61779-794-1_19.

59. ParkhomchukD, BorodinaT, AmstislavskiyV, BanaruM, HallenL, et al. (2009) Transcriptome analysis by strand-specific sequencing of complementary DNA. Nucleic Acids Res 37: e123 doi:10.1093/nar/gkp596.

60. FujitaPA, RheadB, ZweigAS, HinrichsAS, KarolchikD, et al. (2011) The UCSC Genome Browser database: update 2011. Nucleic Acids Res 39: D876–D882 doi:10.1093/nar/gkq963.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 2
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#