#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

MBD3 Localizes at Promoters, Gene Bodies and Enhancers of Active Genes


The Mi-2/nucleosome remodeling and histone deacetylase (NuRD) complex is a multiprotein machine proposed to regulate chromatin structure by nucleosome remodeling and histone deacetylation activities. Recent reports describing localization of NuRD provide new insights that question previous models on NuRD action, but are not in complete agreement. Here, we provide location analysis of endogenous MBD3, a component of NuRD complex, in two human breast cancer cell lines (MCF-7 and MDA-MB-231) using two independent genomic techniques: DNA adenine methyltransferase identification (DamID) and ChIP-seq. We observed concordance of the resulting genomic localization, suggesting that these studies are converging on a robust map for NuRD in the cancer cell genome. MBD3 preferentially associated with CpG rich promoters marked by H3K4me3 and showed cell-type specific localization across gene bodies, peaking around the transcription start site. A subset of sites bound by MBD3 was enriched in H3K27ac and was in physical proximity to promoters in three-dimensional space, suggesting function as enhancers. MBD3 enrichment was also noted at promoters modified by H3K27me3. Functional analysis of chromatin indicated that MBD3 regulates nucleosome occupancy near promoters and in gene bodies. These data suggest that MBD3, and by extension the NuRD complex, may have multiple roles in fine tuning expression for both active and silent genes, representing an important step in defining regulatory mechanisms by which NuRD complex controls chromatin structure and modification status.


Vyšlo v časopise: MBD3 Localizes at Promoters, Gene Bodies and Enhancers of Active Genes. PLoS Genet 9(12): e32767. doi:10.1371/journal.pgen.1004028
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004028

Souhrn

The Mi-2/nucleosome remodeling and histone deacetylase (NuRD) complex is a multiprotein machine proposed to regulate chromatin structure by nucleosome remodeling and histone deacetylation activities. Recent reports describing localization of NuRD provide new insights that question previous models on NuRD action, but are not in complete agreement. Here, we provide location analysis of endogenous MBD3, a component of NuRD complex, in two human breast cancer cell lines (MCF-7 and MDA-MB-231) using two independent genomic techniques: DNA adenine methyltransferase identification (DamID) and ChIP-seq. We observed concordance of the resulting genomic localization, suggesting that these studies are converging on a robust map for NuRD in the cancer cell genome. MBD3 preferentially associated with CpG rich promoters marked by H3K4me3 and showed cell-type specific localization across gene bodies, peaking around the transcription start site. A subset of sites bound by MBD3 was enriched in H3K27ac and was in physical proximity to promoters in three-dimensional space, suggesting function as enhancers. MBD3 enrichment was also noted at promoters modified by H3K27me3. Functional analysis of chromatin indicated that MBD3 regulates nucleosome occupancy near promoters and in gene bodies. These data suggest that MBD3, and by extension the NuRD complex, may have multiple roles in fine tuning expression for both active and silent genes, representing an important step in defining regulatory mechanisms by which NuRD complex controls chromatin structure and modification status.


Zdroje

1. TylerJK, KadonagaJT (1999) The “dark side” of chromatin remodeling: repressive effects on transcription. Cell 99: 443–446.

2. YoshidaT, HazanI, ZhangJ, NgSY, NaitoT, et al. (2008) The role of the chromatin remodeler Mi-2beta in hematopoietic stem cell self-renewal and multilineage differentiation. Genes Dev 22: 1174–1189.

3. WilliamsCJ, NaitoT, ArcoPG, SeavittJR, CashmanSM, et al. (2004) The chromatin remodeler Mi-2beta is required for CD4 expression and T cell development. Immunity 20: 719–733.

4. ZhangJ, JacksonAF, NaitoT, DoseM, SeavittJ, et al. (2012) Harnessing of the nucleosome-remodeling-deacetylase complex controls lymphocyte development and prevents leukemogenesis. Nat Immunol 13: 86–94.

5. ReynoldsN, LatosP, Hynes-AllenA, LoosR, LeafordD, et al. (2012) NuRD suppresses pluripotency gene expression to promote transcriptional heterogeneity and lineage commitment. Cell Stem Cell 10: 583–594.

6. BowenNJ, FujitaN, KajitaM, WadePA (2004) Mi-2/NuRD: multiple complexes for many purposes. Biochim Biophys Acta 1677: 52–57.

7. Le GuezennecX, VermeulenM, BrinkmanAB, HoeijmakersWA, CohenA, et al. (2006) MBD2/NuRD and MBD3/NuRD, two distinct complexes with different biochemical and functional properties. Mol Cell Biol 26: 843–851.

8. HendrichB, BirdA (1998) Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol Cell Biol 18: 6538–6547.

9. YildirimO, LiR, HungJH, ChenPB, DongX, et al. (2011) Mbd3/NURD complex regulates expression of 5-hydroxymethylcytosine marked genes in embryonic stem cells. Cell 147: 1498–1510.

10. WhyteWA, BilodeauS, OrlandoDA, HokeHA, FramptonGM, et al. (2012) Enhancer decommissioning by LSD1 during embryonic stem cell differentiation. Nature 482: 221–225.

11. ReynoldsN, Salmon-DivonM, DvingeH, Hynes-AllenA, BalasooriyaG, et al. (2011) NuRD-mediated deacetylation of H3K27 facilitates recruitment of Polycomb Repressive Complex 2 to direct gene repression. Embo J 31: 593–605.

12. HuG, WadePA (2012) NuRD and pluripotency: a complex balancing act. Cell Stem Cell 10: 497–503.

13. RamO, GorenA, AmitI, ShoreshN, YosefN, et al. (2011) Combinatorial patterning of chromatin regulators uncovered by genome-wide location analysis in human cells. Cell 147: 1628–1639.

14. van SteenselB, HenikoffS (2000) Identification of in vivo DNA targets of chromatin proteins using tethered dam methyltransferase. Nat Biotechnol 18: 424–428.

15. NeveRM, ChinK, FridlyandJ, YehJ, BaehnerFL, et al. (2006) A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell 10: 515–527.

16. 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.

17. FujitaN, JayeDL, KajitaM, GeigermanC, MorenoCS, et al. (2003) MTA3, a Mi-2/NuRD complex subunit, regulates an invasive growth pathway in breast cancer. Cell 113: 207–219.

18. FujitaN, WadePA (2004) Use of bifunctional cross-linking reagents in mapping genomic distribution of chromatin remodeling complexes. Methods 33: 81–85.

19. ZangC, SchonesDE, ZengC, CuiK, ZhaoK, et al. (2009) A clustering approach for identification of enriched domains from histone modification ChIP-Seq data. Bioinformatics 25: 1952–1958.

20. ENCODE_Project_Consortium (2011) A user's guide to the encyclopedia of DNA elements (ENCODE). PLoS Biol 9: e1001046.

21. HashimotoH, LiuY, UpadhyayAK, ChangY, HowertonSB, et al. (2012) Recognition and potential mechanisms for replication and erasure of cytosine hydroxymethylation. Nucleic Acids Res 40: 4841–4849.

22. SpruijtCG, GnerlichF, SmitsAH, PfaffenederT, JansenPW, et al. (2013) Dynamic Readers for 5-(Hydroxy)Methylcytosine and Its Oxidized Derivatives. Cell 152: 1146–1159.

23. CarterD, ChakalovaL, OsborneCS, DaiYF, FraserP (2002) Long-range chromatin regulatory interactions in vivo. Nat Genet 32: 623–626.

24. ZhangJ, PohHM, PehSQ, SiaYY, LiG, et al. (2012) ChIA-PET analysis of transcriptional chromatin interactions. Methods 58: 289–299.

25. DekkerJ, RippeK, DekkerM, KlecknerN (2002) Capturing chromosome conformation. Science 295: 1306–1311.

26. ButlerJS, KoutelouE, SchiblerAC, DentSY (2012) Histone-modifying enzymes: regulators of developmental decisions and drivers of human disease. Epigenomics 4: 163–177.

27. ShuXS, LiL, TaoQ (2012) Chromatin regulators with tumor suppressor properties and their alterations in human cancers. Epigenomics 4: 537–549.

28. NishiokaK, ChuikovS, SarmaK, Erdjument-BromageH, AllisCD, et al. (2002) Set9, a novel histone H3 methyltransferase that facilitates transcription by precluding histone tail modifications required for heterochromatin formation. Genes Dev 16: 479–489.

29. ZegermanP, CanasB, PappinD, KouzaridesT (2002) Histone H3 lysine 4 methylation disrupts binding of nucleosome remodeling and deacetylase (NuRD) repressor complex. J Biol Chem 277: 11621–11624.

30. GuntherK, RustM, LeersJ, BoettgerT, ScharfeM, et al. (2013) Differential roles for MBD2 and MBD3 at methylated CpG islands, active promoters and binding to exon sequences. Nucleic Acids Res 41: 3010–3021.

31. BaubecT, IvanekR, LienertF, SchubelerD (2013) Methylation-Dependent and -Independent Genomic Targeting Principles of the MBD Protein Family. Cell 153: 480–492.

32. JacksonV, ShiresA, ChalkleyR, GrannerDK (1975) Studies on highly metabolically active acetylation and phosphorylation of histones. J Biol Chem 250: 4856–4863.

33. WangZ, ZangC, CuiK, SchonesDE, BarskiA, et al. (2009) Genome-wide mapping of HATs and HDACs reveals distinct functions in active and inactive genes. Cell 138: 1019–1031.

34. MusselmanCA, RamirezJ, SimsJK, MansfieldRE, OliverSS, et al. (2012) Bivalent recognition of nucleosomes by the tandem PHD fingers of the CHD4 ATPase is required for CHD4-mediated repression. Proc Natl Acad Sci U S A 109: 787–792.

35. MusselmanCA, MansfieldRE, GarskeAL, DavrazouF, KwanAH, et al. (2009) Binding of the CHD4 PHD2 finger to histone H3 is modulated by covalent modifications. Biochem J 423: 179–187.

36. HendrichB, BirdA (1998) Identification and characterization of a family of mammalian methyl-CpG binding proteins. Molecular and cellular biology 18: 6538–6547.

37. WadePA (2001) Methyl CpG binding proteins: coupling chromatin architecture to gene regulation. Oncogene 20: 3166–3173.

38. ZhaoS, ChoiM, OvertonJD, BelloneS, RoqueDM, et al. (2013) Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proceedings of the National Academy of Sciences 110: 2916–2921.

39. Le GalloM, O'HaraAJ, RuddML, UrickME, HansenNF, et al. (2012) Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes. Nature genetics 44: 1310–1315.

40. LaiAY, FatemiM, DhasarathyA, MaloneC, SobolSE, et al. (2010) DNA methylation prevents CTCF-mediated silencing of the oncogene BCL6 in B cell lymphomas. J Exp Med 207: 1939–1950.

41. OnderTT, KaraN, CherryA, SinhaAU, ZhuN, et al. (2012) Chromatin-modifying enzymes as modulators of reprogramming. Nature 483: 598–602.

42. GreilF, MoormanC, van SteenselB (2006) DamID: mapping of in vivo protein-genome interactions using tethered DNA adenine methyltransferase. Methods Enzymol 410: 342–359.

43. WandM (1997) Data-based choice of histogram bin width. The American Statistician 51: 59–64.

44. BiedaM, XuX, SingerMA, GreenR, FarnhamPJ (2006) Unbiased location analysis of E2F1-binding sites suggests a widespread role for E2F1 in the human genome. Genome research 16: 595–605.

45. Hastie T, Tibshirani R (1990) Generalized additive models. New York: Chapman & Hall/CRC.

46. ScacheriPC, CrawfordGE, DavisS (2006) Statistics for ChIP-chip and DNase hypersensitivity experiments on NimbleGen arrays. Methods in enzymology 411: 270.

47. Charafe-JauffretE, GinestierC, MonvilleF, FinettiP, AdelaideJ, et al. (2006) Gene expression profiling of breast cell lines identifies potential new basal markers. Oncogene 25: 2273–2284.

48. WeberM, HellmannI, StadlerMB, RamosL, PääboS, et al. (2007) Distribution, silencing potential and evolutionary impact of promoter DNA methylation in the human genome. Nature genetics 39: 457–466.

49. WhyteWA, OrlandoDA, HniszD, AbrahamBJ, LinCY, et al. (2013) Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153: 307–319.

50. LiZ, GadueP, ChenK, JiaoY, TutejaG, et al. (2012) Foxa2 and H2A. Z mediate nucleosome depletion during embryonic stem cell differentiation. Cell 151: 1608–1616.

51. LangmeadB, TrapnellC, PopM, SalzbergSL (2009) Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol 10: R25.

52. QuinlanAR, HallIM (2010) BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26: 841–842.

53. KentWJ, SugnetCW, FureyTS, RoskinKM, PringleTH, et al. (2002) The human genome browser at UCSC. Genome Res 12: 996–1006.

54. KhaitanD, SankpalUT, WekslerB, MeisterEA, RomeroIA, et al. (2009) Role of KCNMA1 gene in breast cancer invasion and metastasis to brain. BMC Cancer 9: 258.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 12
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#