#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Histone Methyltransferase MMSET/NSD2 Alters EZH2 Binding and Reprograms the Myeloma Epigenome through Global and Focal Changes in H3K36 and H3K27 Methylation


Precise spatial and temporal gene expression is required for normal development, and aberrant regulation of gene expression is a common factor in many diseases, including cancer. Histone modifications contribute to the control of gene expression by altering chromatin structure and affecting the recruitment of transcriptional regulators. In this study, we demonstrate interplay between two oncogenic proteins, MMSET and EZH2, known to methylate histone H3 on lysine 36 (H3K36) and lysine 27 (H3K27), respectively. Overexpression of MMSET in myeloma cells increases global levels of H3K36 methylation, alters its normal distribution throughout the genome and decreases global levels of H3K27 methylation. We found that while the majority of the genome loses H3K27 methylation in the presence of MMSET, certain loci have augmented recruitment of EZH2 and enhanced H3K27 methylation, leading to transcriptional repression. Repression of these genes likely plays an important role in the disease because MMSET-overexpressing cells show higher sensitivity to small molecule inhibitors targeting EZH2-mediated methylation. Thus, our study suggests that the specific local changes may outweigh the gross global changes we frequently observe in cancer and implicates EZH2 as a novel therapeutic target in myeloma cells.


Vyšlo v časopise: Histone Methyltransferase MMSET/NSD2 Alters EZH2 Binding and Reprograms the Myeloma Epigenome through Global and Focal Changes in H3K36 and H3K27 Methylation. PLoS Genet 10(9): e32767. doi:10.1371/journal.pgen.1004566
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004566

Souhrn

Precise spatial and temporal gene expression is required for normal development, and aberrant regulation of gene expression is a common factor in many diseases, including cancer. Histone modifications contribute to the control of gene expression by altering chromatin structure and affecting the recruitment of transcriptional regulators. In this study, we demonstrate interplay between two oncogenic proteins, MMSET and EZH2, known to methylate histone H3 on lysine 36 (H3K36) and lysine 27 (H3K27), respectively. Overexpression of MMSET in myeloma cells increases global levels of H3K36 methylation, alters its normal distribution throughout the genome and decreases global levels of H3K27 methylation. We found that while the majority of the genome loses H3K27 methylation in the presence of MMSET, certain loci have augmented recruitment of EZH2 and enhanced H3K27 methylation, leading to transcriptional repression. Repression of these genes likely plays an important role in the disease because MMSET-overexpressing cells show higher sensitivity to small molecule inhibitors targeting EZH2-mediated methylation. Thus, our study suggests that the specific local changes may outweigh the gross global changes we frequently observe in cancer and implicates EZH2 as a novel therapeutic target in myeloma cells.


Zdroje

1. PopovicR, LichtJD (2012) Emerging epigenetic targets and therapies in cancer medicine. Cancer Discov 2: 405–413.

2. MoreyL, HelinK (2010) Polycomb group protein-mediated repression of transcription. Trends Biochem Sci 35: 323–332.

3. VireE, BrennerC, DeplusR, BlanchonL, FragaM, et al. (2006) The Polycomb group protein EZH2 directly controls DNA methylation. Nature 439: 871–874.

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

5. MorinRD, JohnsonNA, SeversonTM, MungallAJ, AnJ, et al. (2010) Somatic mutations altering EZH2 (Tyr641) in follicular and diffuse large B-cell lymphomas of germinal-center origin. Nat Genet 42: 181–185.

6. BeguelinW, PopovicR, TeaterM, JiangY, BuntingKL, et al. (2013) EZH2 is required for germinal center formation and somatic EZH2 mutations promote lymphoid transformation. Cancer Cell 23: 677–692.

7. Abdel-WahabO, AdliM, LaFaveLM, GaoJ, HricikT, et al. (2012) ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell 22: 180–193.

8. ErnstT, ChaseAJ, ScoreJ, Hidalgo-CurtisCE, BryantC, et al. (2010) Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet 42: 722–726.

9. ScoreJ, Hidalgo-CurtisC, JonesAV, WinkelmannN, SkinnerA, et al. (2012) Inactivation of polycomb repressive complex 2 components in myeloproliferative and myelodysplastic/myeloproliferative neoplasms. Blood 119: 1208–1213.

10. LiY, TrojerP, XuCF, CheungP, KuoA, et al. (2009) The target of the NSD family of histone lysine methyltransferases depends on the nature of the substrate. J Biol Chem 284: 34283–34295.

11. KuoAJ, CheungP, ChenK, ZeeBM, KioiM, et al. (2011) NSD2 links dimethylation of histone H3 at lysine 36 to oncogenic programming. Mol Cell 44: 609–620.

12. Martinez-GarciaE, PopovicR, MinDJ, SweetSM, ThomasPM, et al. (2011) The MMSET histone methyl transferase switches global histone methylation and alters gene expression in t(4;14) multiple myeloma cells. Blood 117: 211–220.

13. WagnerEJ, CarpenterPB (2012) Understanding the language of Lys36 methylation at histone H3. Nat Rev Mol Cell Biol 13: 115–126.

14. BergemannAD, ColeF, HirschhornK (2005) The etiology of Wolf-Hirschhorn syndrome. Trends Genet 21: 188–195.

15. NimuraK, UraK, ShiratoriH, IkawaM, OkabeM, et al. (2009) A histone H3 lysine 36 trimethyltransferase links Nkx2-5 to Wolf-Hirschhorn syndrome. Nature 460: 287–291.

16. KeatsJJ, ReimanT, BelchAR, PilarskiLM (2006) Ten years and counting: so what do we know about t(4;14)(p16;q32) multiple myeloma. Leuk Lymphoma 47: 2289–2300.

17. StecI, WrightTJ, van OmmenGJ, de BoerPA, van HaeringenA, et al. (1998) WHSC1, a 90 kb SET domain-containing gene, expressed in early development and homologous to a Drosophila dysmorphy gene maps in the Wolf-Hirschhorn syndrome critical region and is fused to IgH in t(4;14) multiple myeloma. Hum Mol Genet 7: 1071–1082.

18. KeatsJJ, ReimanT, MaxwellCA, TaylorBJ, LarrattLM, et al. (2003) In multiple myeloma, t(4;14)(p16;q32) is an adverse prognostic factor irrespective of FGFR3 expression. Blood 101: 1520–1529.

19. SantraM, ZhanF, TianE, BarlogieB, ShaughnessyJJr (2003) A subset of multiple myeloma harboring the t(4;14)(p16;q32) translocation lacks FGFR3 expression but maintains an IGH/MMSET fusion transcript. Blood 101: 2374–2376.

20. EzpondaT, PopovicR, ShahMY, Martinez-GarciaE, ZhengY, et al. (2012) The histone methyltransferase MMSET/WHSC1 activates TWIST1 to promote an epithelial-mesenchymal transition and invasive properties of prostate cancer. Oncogene 32 (23) 2882–90..

21. HudlebuschHR, Santoni-RugiuE, SimonR, RalfkiaerE, RossingHH, et al. (2011) The histone methyltransferase and putative oncoprotein MMSET is overexpressed in a large variety of human tumors. Clin Cancer Res 17: 2919–2933.

22. ZhouP, WuLL, WuKM, JiangW, LiJD, et al. (2013) Overexpression of MMSET is correlation with poor prognosis in hepatocellular carcinoma. Pathol Oncol Res 19: 303–309.

23. YangS, ZhangY, MengF, LiuY, XiaB, et al. (2013) Overexpression of multiple myeloma SET domain (MMSET) is associated with advanced tumor aggressiveness and poor prognosis in serous ovarian carcinoma. Biomarkers 18: 257–263.

24. OyerJA, HuangX, ZhengY, ShimJ, EzpondaT, et al. (2013) Point mutation E1099K in MMSET/NSD2 enhances its methyltranferase activity and leads to altered global chromatin methylation in lymphoid malignancies. Leukemia 28 (1) 198–201..

25. JaffeJD, WangY, ChanHM, ZhangJ, HuetherR, et al. (2013) Global chromatin profiling reveals NSD2 mutations in pediatric acute lymphoblastic leukemia. Nat Genet 45: 1386–1391.

26. LauringJ, AbukhdeirAM, KonishiH, GarayJP, GustinJP, et al. (2008) The multiple myeloma associated MMSET gene contributes to cellular adhesion, clonogenic growth, and tumorigenicity. Blood 111: 856–864.

27. ZhengY, SweetSM, PopovicR, Martinez-GarciaE, TiptonJD, et al. (2012) Total kinetic analysis reveals how combinatorial methylation patterns are established on lysines 27 and 36 of histone H3. Proc Natl Acad Sci U S A 109: 13549–13554.

28. BernsteinBE, BirneyE, DunhamI, GreenED, GunterC, et al. (2012) An integrated encyclopedia of DNA elements in the human genome. Nature 489: 57–74.

29. HuangN, KawanoMM, MahmoudMS, MiharaK, TsujimotoT, et al. (1995) Expression of CD21 antigen on myeloma cells and its involvement in their adhesion to bone marrow stromal cells. Blood 85: 3704–3712.

30. BarskiA, CuddapahS, CuiK, RohTY, SchonesDE, et al. (2007) High-resolution profiling of histone methylations in the human genome. Cell 129: 823–837.

31. KondoY, ShenL, ChengAS, AhmedS, BoumberY, et al. (2008) Gene silencing in cancer by histone H3 lysine 27 trimethylation independent of promoter DNA methylation. Nat Genet 40: 741–750.

32. BrackenAP, DietrichN, PasiniD, HansenKH, HelinK (2006) Genome-wide mapping of Polycomb target genes unravels their roles in cell fate transitions. Genes Dev 20: 1123–1136.

33. 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.

34. KammingaLM, BystrykhLV, de BoerA, HouwerS, DoumaJ, et al. (2006) The Polycomb group gene Ezh2 prevents hematopoietic stem cell exhaustion. Blood 107: 2170–2179.

35. SchmitgesFW, PrustyAB, FatyM, StutzerA, LingarajuGM, et al. (2011) Histone methylation by PRC2 is inhibited by active chromatin marks. Mol Cell 42: 330–341.

36. YuanW, XuM, HuangC, LiuN, ChenS, et al. (2011) H3K36 methylation antagonizes PRC2-mediated H3K27 methylation. J Biol Chem 286: 7983–7989.

37. ShafferAL, WrightG, YangL, PowellJ, NgoV, et al. (2006) A library of gene expression signatures to illuminate normal and pathological lymphoid biology. Immunol Rev 210: 67–85.

38. GranitRZ, GabaiY, HadarT, KaramanshaY, LibermanL, et al. (2012) EZH2 promotes a bi-lineage identity in basal-like breast cancer cells. Oncogene 32 (33) 3886–95..

39. MendenhallEM, KocheRP, TruongT, ZhouVW, IssacB, et al. (2010) GC-rich sequence elements recruit PRC2 in mammalian ES cells. PLoS Genet 6: e1001244.

40. VermaSK, XT, LaFranceLV, CD, SuarezDP, et al. (2012) Identification of Potent, Selective, Cell-Active Inhibitors of the Histone Lysine Methyltransferase EZH2. ACS Medical Chemistry Letters 3: 1091–1096.

41. ShouY, MartelliML, GabreaA, QiY, BrentsLA, et al. (2000) Diverse karyotypic abnormalities of the c-myc locus associated with c-myc dysregulation and tumor progression in multiple myeloma. Proc Natl Acad Sci U S A 97: 228–233.

42. ChngWJ, HuangGF, ChungTH, NgSB, Gonzalez-PazN, et al. (2011) Clinical and biological implications of MYC activation: a common difference between MGUS and newly diagnosed multiple myeloma. Leukemia 25: 1026–1035.

43. MinDJ, EzpondaT, KimMK, WillCM, Martinez-GarciaE, et al. (2013) MMSET stimulates myeloma cell growth through microRNA-mediated modulation of c-MYC. Leukemia 27: 686–694.

44. BakerLA, AllisCD, WangGG (2008) PHD fingers in human diseases: disorders arising from misinterpreting epigenetic marks. Mutat Res 647: 3–12.

45. WuH, ZengH, LamR, TempelW, AmayaMF, et al. (2011) Structural and histone binding ability characterizations of human PWWP domains. PLoS One 6: e18919.

46. KurotakiN, ImaizumiK, HaradaN, MasunoM, KondohT, et al. (2002) Haploinsufficiency of NSD1 causes Sotos syndrome. Nat Genet 30: 365–366.

47. MardisER, DingL, DoolingDJ, LarsonDE, McLellanMD, et al. (2009) Recurring mutations found by sequencing an acute myeloid leukemia genome. N Engl J Med 361: 1058–1066.

48. DelhommeauF, DupontS, Della ValleV, JamesC, TrannoyS, et al. (2009) Mutation in TET2 in myeloid cancers. N Engl J Med 360: 2289–2301.

49. DalglieshGL, FurgeK, GreenmanC, ChenL, BignellG, et al. (2010) Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463: 360–363.

50. KalushkovaA, FryknasM, LemaireM, FristedtC, AgarwalP, et al. (2010) Polycomb target genes are silenced in multiple myeloma. PLoS One 5: e11483.

51. VillaR, PasiniD, GutierrezA, MoreyL, OcchionorelliM, et al. (2007) Role of the polycomb repressive complex 2 in acute promyelocytic leukemia. Cancer Cell 11: 513–525.

52. PengJC, ValouevA, SwigutT, ZhangJ, ZhaoY, et al. (2009) Jarid2/Jumonji coordinates control of PRC2 enzymatic activity and target gene occupancy in pluripotent cells. Cell 139: 1290–1302.

53. MargueronR, JustinN, OhnoK, SharpeML, SonJ, et al. (2009) Role of the polycomb protein EED in the propagation of repressive histone marks. Nature 461: 762–767.

54. Van BortleK, RamosE, TakenakaN, YangJ, WahiJE, et al. (2012) Drosophila CTCF tandemly aligns with other insulator proteins at the borders of H3K27me3 domains. Genome Res 22: 2176–2187.

55. LhoumaudP, HennionM, GamotA, CuddapahS, QueilleS, et al. (2014) Insulators recruit histone methyltransferase dMes4 to regulate chromatin of flanking genes. EMBO J 33 (14) 1599–613..

56. GaydosLJ, RechtsteinerA, EgelhoferTA, CarrollCR, StromeS (2012) Antagonism between MES-4 and Polycomb Repressive Complex 2 Promotes Appropriate Gene Expression in C. elegans Germ Cells. Cell Rep 2 (5) 1169–77..

57. BlackledgeNP, ZhouJC, TolstorukovMY, FarcasAM, ParkPJ, et al. (2010) CpG islands recruit a histone H3 lysine 36 demethylase. Mol Cell 38: 179–190.

58. SchwartzentruberJ, KorshunovA, LiuXY, JonesDT, PfaffE, et al. (2012) Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482: 226–231.

59. AsanganiIA, AteeqB, CaoQ, DodsonL, PandhiM, et al. (2013) Characterization of the EZH2-MMSET histone methyltransferase regulatory axis in cancer. Mol Cell 49: 80–93.

60. HeC, LiF, ZhangJ, WuJ, ShiY (2013) The methyltransferase NSD3 has chromatin-binding motifs, PHD5-C5HCH, that are distinct from other NSD (nuclear receptor SET domain) family members in their histone H3 recognition. J Biol Chem 288: 4692–4703.

61. HollinkIH, van den Heuvel-EibrinkMM, Arentsen-PetersST, PratcoronaM, AbbasS, et al. (2011) NUP98/NSD1 characterizes a novel poor prognostic group in acute myeloid leukemia with a distinct HOX gene expression pattern. Blood 118: 3645–3656.

62. WangGG, CaiL, PasillasMP, KampsMP (2007) NUP98-NSD1 links H3K36 methylation to Hox-A gene activation and leukaemogenesis. Nat Cell Biol 9: 804–812.

63. FaravelliF (2005) NSD1 mutations in Sotos syndrome. Am J Med Genet C Semin Med Genet 137C: 24–31.

64. ZhangJ, DingL, HolmfeldtL, WuG, HeatleySL, et al. (2012) The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature 481: 157–163.

65. van HaaftenG, DalglieshGL, DaviesH, ChenL, BignellG, et al. (2009) Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet 41: 521–523.

66. CroonquistPA, Van NessB (2005) The polycomb group protein enhancer of zeste homolog 2 (EZH 2) is an oncogene that influences myeloma cell growth and the mutant ras phenotype. Oncogene 24: 6269–6280.

67. DelmoreJE, IssaGC, LemieuxME, RahlPB, ShiJ, et al. (2011) BET bromodomain inhibition as a therapeutic strategy to target c-Myc. Cell 146: 904–917.

68. Creasy CL, McCabe MT, Korenchuk S, Diaz E, Ott H, et al.. (2012) A novel selective EZH2 inhibitor exhibits anti-tumor activity in lymphoma with EZH2 activating mutations. Chicago: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research. pp. 492.

69. McCabeMT, OttHM, GanjiG, KorenchukS, ThompsonC, et al. (2012) EZH2 inhibition as a therapeutic strategy for lymphoma with EZH2-activating mutations. Nature 492 (7427) 108–12..

70. ZhangY, LiuT, MeyerCA, EeckhouteJ, JohnsonDS, et al. (2008) Model-based analysis of ChIP-Seq (MACS). Genome Biol 9: R137.

71. GiannopoulouEG, ElementoO (2011) An integrated ChIP-seq analysis platform with customizable workflows. BMC Bioinformatics 12: 277.

72. ElementoO, SlonimN, TavazoieS (2007) A universal framework for regulatory element discovery across all genomes and data types. Mol Cell 28: 337–350.

73. HeinzS, BennerC, SpannN, BertolinoE, LinYC, et al. (2010) Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell 38: 576–589.

74. GoodarziH, ElementoO, TavazoieS (2009) Revealing global regulatory perturbations across human cancers. Mol Cell 36: 900–911.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 9
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#