#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Factors That Promote H3 Chromatin Integrity during Transcription Prevent Promiscuous Deposition of CENP-A in Fission Yeast


Specialized chromatin containing CENP-A nucleosomes instead of H3 nucleosomes is found at all centromeres. However, the mechanisms that specify the locations at which CENP-A chromatin is assembled remain elusive in organisms with regional, epigenetically regulated centromeres. It is known that normal centromeric DNA is transcribed in several systems including the fission yeast, Schizosaccharomyces pombe. Here, we show that factors which preserve stable histone H3 chromatin during transcription also play a role in preventing promiscuous CENP-ACnp1 deposition in fission yeast. Mutations in the histone chaperone FACT impair the maintenance of H3 chromatin on transcribed regions and promote widespread CENP-ACnp1 incorporation at non-centromeric sites. FACT has little or no effect on CENP-ACnp1 assembly at endogenous centromeres where CENP-ACnp1 is normally assembled. In contrast, Clr6 complex II (Clr6-CII; equivalent to Rpd3S) histone deacetylase function has a more subtle impact on the stability of transcribed H3 chromatin and acts to prevent the ectopic accumulation of CENP-ACnp1 at specific loci, including subtelomeric regions, where CENP-ACnp1 is preferentially assembled. Moreover, defective Clr6-CII function allows the de novo assembly of CENP-ACnp1 chromatin on centromeric DNA, bypassing the normal requirement for heterochromatin. Thus, our analyses show that alterations in the process of chromatin assembly during transcription can destabilize H3 nucleosomes and thereby allow CENP-ACnp1 to assemble in its place. We propose that normal centromeres provide a specific chromatin context that limits reassembly of H3 chromatin during transcription and thereby promotes the establishment of CENP-ACnp1 chromatin and associated kinetochores. These findings have important implications for genetic and epigenetic processes involved in centromere specification.


Vyšlo v časopise: Factors That Promote H3 Chromatin Integrity during Transcription Prevent Promiscuous Deposition of CENP-A in Fission Yeast. PLoS Genet 8(9): e32767. doi:10.1371/journal.pgen.1002985
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1002985

Souhrn

Specialized chromatin containing CENP-A nucleosomes instead of H3 nucleosomes is found at all centromeres. However, the mechanisms that specify the locations at which CENP-A chromatin is assembled remain elusive in organisms with regional, epigenetically regulated centromeres. It is known that normal centromeric DNA is transcribed in several systems including the fission yeast, Schizosaccharomyces pombe. Here, we show that factors which preserve stable histone H3 chromatin during transcription also play a role in preventing promiscuous CENP-ACnp1 deposition in fission yeast. Mutations in the histone chaperone FACT impair the maintenance of H3 chromatin on transcribed regions and promote widespread CENP-ACnp1 incorporation at non-centromeric sites. FACT has little or no effect on CENP-ACnp1 assembly at endogenous centromeres where CENP-ACnp1 is normally assembled. In contrast, Clr6 complex II (Clr6-CII; equivalent to Rpd3S) histone deacetylase function has a more subtle impact on the stability of transcribed H3 chromatin and acts to prevent the ectopic accumulation of CENP-ACnp1 at specific loci, including subtelomeric regions, where CENP-ACnp1 is preferentially assembled. Moreover, defective Clr6-CII function allows the de novo assembly of CENP-ACnp1 chromatin on centromeric DNA, bypassing the normal requirement for heterochromatin. Thus, our analyses show that alterations in the process of chromatin assembly during transcription can destabilize H3 nucleosomes and thereby allow CENP-ACnp1 to assemble in its place. We propose that normal centromeres provide a specific chromatin context that limits reassembly of H3 chromatin during transcription and thereby promotes the establishment of CENP-ACnp1 chromatin and associated kinetochores. These findings have important implications for genetic and epigenetic processes involved in centromere specification.


Zdroje

1. BlackBE, ClevelandDW (2011) Epigenetic centromere propagation and the nature of CENP-a nucleosomes. Cell 144: 471–479.

2. StimpsonKM, SullivanBA (2010) Epigenomics of centromere assembly and function. Current opinion in cell biology 22: 772–780.

3. MalikHS, HenikoffS (2009) Major evolutionary transitions in centromere complexity. Cell 138: 1067–1082.

4. MarshallOJ, ChuehAC, WongLH, ChooKH (2008) Neocentromeres: new insights into centromere structure, disease development, and karyotype evolution. American journal of human genetics 82: 261–282.

5. AllshireRC, KarpenGH (2008) Epigenetic regulation of centromeric chromatin: old dogs, new tricks? Nature reviews Genetics 9: 923–937.

6. BuscainoA, AllshireR, PidouxA (2010) Building centromeres: home sweet home or a nomadic existence? Current opinion in genetics & development 20: 118–126.

7. MendiburoMJ, PadekenJ, FulopS, SchepersA, HeunP (2011) Drosophila CENH3 is sufficient for centromere formation. Science 334: 686–690.

8. IshiiK (2009) Conservation and divergence of centromere specification in yeast. Current opinion in microbiology 12: 616–622.

9. KaganskyA, FolcoHD, AlmeidaR, PidouxAL, BoukabaA, et al. (2009) Synthetic heterochromatin bypasses RNAi and centromeric repeats to establish functional centromeres. Science 324: 1716–1719.

10. FolcoHD, PidouxAL, UranoT, AllshireRC (2008) Heterochromatin and RNAi are required to establish CENP-A chromatin at centromeres. Science 319: 94–97.

11. SatoH, MasudaF, TakayamaY, TakahashiK, SaitohS (2012) Epigenetic inactivation and subsequent heterochromatinization of a centromere stabilize dicentric chromosomes. Current biology : CB 22: 658–667.

12. IshiiK, OgiyamaY, ChikashigeY, SoejimaS, MasudaF, et al. (2008) Heterochromatin integrity affects chromosome reorganization after centromere dysfunction. Science 321: 1088–1091.

13. SullivanLL, BoivinCD, MravinacB, SongIY, SullivanBA (2011) Genomic size of CENP-A domain is proportional to total alpha satellite array size at human centromeres and expands in cancer cells. Chromosome research : an international journal on the molecular, supramolecular and evolutionary aspects of chromosome biology 19: 457–470.

14. TomonagaT, MatsushitaK, YamaguchiS, OohashiT, ShimadaH, et al. (2003) Overexpression and mistargeting of centromere protein-A in human primary colorectal cancer. Cancer research 63: 3511–3516.

15. OlszakAM, van EssenD, PereiraAJ, DiehlS, MankeT, et al. (2011) Heterochromatin boundaries are hotspots for de novo kinetochore formation. Nature cell biology 13: 799–808.

16. AmatoA, SchillaciT, LentiniL, Di LeonardoA (2009) CENPA overexpression promotes genome instability in pRb-depleted human cells. Mol Cancer 8: 119.

17. HeunP, ErhardtS, BlowerMD, WeissS, SkoraAD, et al. (2006) Mislocalization of the Drosophila centromere-specific histone CID promotes formation of functional ectopic kinetochores. Developmental cell 10: 303–315.

18. ChanFL, MarshallOJ, SafferyR, KimBW, EarleE, et al. (2012) Active transcription and essential role of RNA polymerase II at the centromere during mitosis. Proceedings of the National Academy of Sciences of the United States of America 109: 1979–1984.

19. BergmannJH, RodriguezMG, MartinsNM, KimuraH, KellyDA, et al. (2011) Epigenetic engineering shows H3K4me2 is required for HJURP targeting and CENP-A assembly on a synthetic human kinetochore. The EMBO journal 30: 328–340.

20. ChuehAC, NorthropEL, Brettingham-MooreKH, ChooKH, WongLH (2009) LINE retrotransposon RNA is an essential structural and functional epigenetic component of a core neocentromeric chromatin. PLoS Genet 5: e1000354 doi:10.1371/journal.pcbi.1000354.

21. CaroneDM, LongoMS, FerreriGC, HallL, HarrisM, et al. (2009) A new class of retroviral and satellite encoded small RNAs emanates from mammalian centromeres. Chromosoma 118: 113–125.

22. WongLH, Brettingham-MooreKH, ChanL, QuachJM, AndersonMA, et al. (2007) Centromere RNA is a key component for the assembly of nucleoproteins at the nucleolus and centromere. Genome research 17: 1146–1160.

23. ToppCN, ZhongCX, DaweRK (2004) Centromere-encoded RNAs are integral components of the maize kinetochore. Proceedings of the National Academy of Sciences of the United States of America 101: 15986–15991.

24. ChoiES, StralforsA, CastilloAG, Durand-DubiefM, EkwallK, et al. (2011) Identification of noncoding transcripts from within CENP-A chromatin at fission yeast centromeres. The Journal of biological chemistry 286: 23600–23607.

25. HenikoffS (2008) Nucleosome destabilization in the epigenetic regulation of gene expression. Nature reviews Genetics 9: 15–26.

26. LiB, CareyM, WorkmanJL (2007) The role of chromatin during transcription. Cell 128: 707–719.

27. SaundersA, WernerJ, AndrulisED, NakayamaT, HiroseS, et al. (2003) Tracking FACT and the RNA polymerase II elongation complex through chromatin in vivo. Science 301: 1094–1096.

28. KaplanCD, LapradeL, WinstonF (2003) Transcription elongation factors repress transcription initiation from cryptic sites. Science 301: 1096–1099.

29. BelotserkovskayaR, OhS, BondarenkoVA, OrphanidesG, StuditskyVM, et al. (2003) FACT facilitates transcription-dependent nucleosome alteration. Science 301: 1090–1093.

30. KeoghMC, KurdistaniSK, MorrisSA, AhnSH, PodolnyV, et al. (2005) Cotranscriptional set2 methylation of histone H3 lysine 36 recruits a repressive Rpd3 complex. Cell 123: 593–605.

31. CarrozzaMJ, LiB, FlorensL, SuganumaT, SwansonSK, et al. (2005) Histone H3 methylation by Set2 directs deacetylation of coding regions by Rpd3S to suppress spurious intragenic transcription. Cell 123: 581–592.

32. MasonPB, StruhlK (2003) The FACT complex travels with elongating RNA polymerase II and is important for the fidelity of transcriptional initiation in vivo. Molecular and cellular biology 23: 8323–8333.

33. NicolasE, YamadaT, CamHP, FitzgeraldPC, KobayashiR, et al. (2007) Distinct roles of HDAC complexes in promoter silencing, antisense suppression and DNA damage protection. Nature structural & molecular biology 14: 372–380.

34. WittmeyerJ, JossL, FormosaT (1999) Spt16 and Pob3 of Saccharomyces cerevisiae form an essential, abundant heterodimer that is nuclear, chromatin-associated, and copurifies with DNA polymerase alpha. Biochemistry 38: 8961–8971.

35. OrphanidesG, WuWH, LaneWS, HampseyM, ReinbergD (1999) The chromatin-specific transcription elongation factor FACT comprises human SPT16 and SSRP1 proteins. Nature 400: 284–288.

36. LejeuneE, BortfeldM, WhiteSA, PidouxAL, EkwallK, et al. (2007) The chromatin-remodeling factor FACT contributes to centromeric heterochromatin independently of RNAi. Current biology : CB 17: 1219–1224.

37. ForsburgSL (1993) Comparison of Schizosaccharomyces pombe expression systems. Nucleic acids research 21: 2955–2956.

38. CastilloAG, MelloneBG, PartridgeJF, RichardsonW, HamiltonGL, et al. (2007) Plasticity of fission yeast CENP-A chromatin driven by relative levels of histone H3 and H4. PLoS Genet 3: e121 doi:10.1371/journal.pgen.0030121.

39. PidouxAL, RichardsonW, AllshireRC (2003) Sim4: a novel fission yeast kinetochore protein required for centromeric silencing and chromosome segregation. The Journal of cell biology 161: 295–307.

40. JamaiA, PuglisiA, StrubinM (2009) Histone chaperone spt16 promotes redeposition of the original h3-h4 histones evicted by elongating RNA polymerase. Molecular cell 35: 377–383.

41. SchwabishMA, StruhlK (2004) Evidence for eviction and rapid deposition of histones upon transcriptional elongation by RNA polymerase II. Molecular and cellular biology 24: 10111–10117.

42. FoltzDR, JansenLE, BlackBE, BaileyAO, YatesJR3rd, et al. (2006) The human CENP-A centromeric nucleosome-associated complex. Nature cell biology 8: 458–469.

43. KielyCM, MargueratS, GarciaJF, MadhaniHD, BahlerJ, et al. (2011) Spt6 is required for heterochromatic silencing in the fission yeast Schizosaccharomyces pombe. Molecular and cellular biology

44. HayashiT, FujitaY, IwasakiO, AdachiY, TakahashiK, et al. (2004) Mis16 and Mis18 are required for CENP-A loading and histone deacetylation at centromeres. Cell 118: 715–729.

45. TakahashiK, ChenES, YanagidaM (2000) Requirement of Mis6 centromere connector for localizing a CENP-A-like protein in fission yeast. Science 288: 2215–2219.

46. WilliamsJS, HayashiT, YanagidaM, RussellP (2009) Fission yeast Scm3 mediates stable assembly of Cnp1/CENP-A into centromeric chromatin. Molecular cell 33: 287–298.

47. PidouxAL, ChoiES, AbbottJK, LiuX, KaganskyA, et al. (2009) Fission yeast Scm3: A CENP-A receptor required for integrity of subkinetochore chromatin. Molecular cell 33: 299–311.

48. DunleavyEM, PidouxAL, MonetM, BonillaC, RichardsonW, et al. (2007) A NASP (N1/N2)-related protein, Sim3, binds CENP-A and is required for its deposition at fission yeast centromeres. Molecular cell 28: 1029–1044.

49. GovindCK, QiuH, GinsburgDS, RuanC, HofmeyerK, et al. (2010) Phosphorylated Pol II CTD recruits multiple HDACs, including Rpd3C(S), for methylation-dependent deacetylation of ORF nucleosomes. Molecular cell 39: 234–246.

50. OkadaT, OhzekiJ, NakanoM, YodaK, BrinkleyWR, et al. (2007) CENP-B controls centromere formation depending on the chromatin context. Cell 131: 1287–1300.

51. WinklerDD, LugerK (2011) The Histone Chaperone FACT: Structural Insights and Mechanisms for Nucleosome Reorganization. The Journal of biological chemistry 286: 18369–18374.

52. JansenLE, BlackBE, FoltzDR, ClevelandDW (2007) Propagation of centromeric chromatin requires exit from mitosis. The Journal of cell biology 176: 795–805.

53. OkadaM, OkawaK, IsobeT, FukagawaT (2009) CENP-H-containing complex facilitates centromere deposition of CENP-A in cooperation with FACT and CHD1. Molecular biology of the cell 20: 3986–3995.

54. StuweT, HothornM, LejeuneE, RybinV, BortfeldM, et al. (2008) The FACT Spt16 “peptidase” domain is a histone H3-H4 binding module. Proceedings of the National Academy of Sciences of the United States of America 105: 8884–8889.

55. GassmannR, RechtsteinerA, YuenKW, MuroyamaA, EgelhoferT, et al. (2012) An inverse relationship to germline transcription defines centromeric chromatin in C. elegans. Nature 484: 534–537.

56. ChanFL, MarshallOJ, SafferyR, KimBW, EarleE, et al. (2012) Active transcription and essential role of RNA polymerase II at the centromere during mitosis. Proceedings of the National Academy of Sciences of the United States of America 109: 1979–1984.

57. KetelC, WangHS, McClellanM, BouchonvilleK, SelmeckiA, et al. (2009) Neocentromeres form efficiently at multiple possible loci in Candida albicans. PLoS Genet 5: e1000400 doi:10.1371/journal.pgen.1000400.

58. MorenoS, KlarA, NurseP (1991) Molecular genetic analysis of fission yeast Schizosaccharomyces pombe. Methods in enzymology 194: 795–823.

59. NelsonJD, DenisenkoO, BomsztykK (2006) Protocol for the fast chromatin immunoprecipitation (ChIP) method. Nature protocols 1: 179–185.

60. Durand-DubiefM, EkwallK (2009) Chromatin immunoprecipitation using microarrays. Methods in molecular biology 529: 279–295.

61. SadeghiL, BonillaC, StralforsA, EkwallK, SvenssonJP (2011) Podbat: a novel genomic tool reveals Swr1-independent H2A.Z incorporation at gene coding sequences through epigenetic meta-analysis. PLoS Comput Biol 7: e1002163 doi:10.1371/journal.pcbi.1002163.

62. Durand-DubiefM, PerssonJ, NormanU, HartsuikerE, EkwallK (2010) Topoisomerase I regulates open chromatin and controls gene expression in vivo. The EMBO journal 29: 2126–2134.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 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#