#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

H2B Mono-ubiquitylation Facilitates Fork Stalling and Recovery during Replication Stress by Coordinating Rad53 Activation and Chromatin Assembly


Eukaryotic DNA is organized into nucleosomes, which are the fundamental repeating units of chromatin. Coordination of chromatin structure is required for efficient and accurate DNA replication. Aberrant DNA replication results in mutations and chromosome rearrangements that may be associated with human disorders. Therefore, cellular surveillance mechanisms have evolved to counteract potential threats to DNA replication. These mechanisms include checkpoints and specialized enzymatic activities that prevent the replication and segregation of defective DNA molecules. We employed a genome-wide approach to investigate how chromatin structure affects DNA replication under stress. We report that coordination of chromatin assembly and checkpoint activity by a histone modification, H2B ubiquitylation (H2Bub), is critical for the cell response to HU-induced replication stress. In cells with a mutation that abolishes H2Bub, replication progression is enhanced, and the forks are more susceptible to damage by environmental insults. The replication proteins on replicating DNA are akin to a train on the tracks, and movement of this train is carefully controlled. Our data indicate that H2Bub helps organize DNA in the nuclei during DNA replication; this process plays a similar role to the brakes on a train, serving to slow down replication, and maintaining stable progression of replication under environmental stress.


Vyšlo v časopise: H2B Mono-ubiquitylation Facilitates Fork Stalling and Recovery during Replication Stress by Coordinating Rad53 Activation and Chromatin Assembly. PLoS Genet 10(10): e32767. doi:10.1371/journal.pgen.1004667
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004667

Souhrn

Eukaryotic DNA is organized into nucleosomes, which are the fundamental repeating units of chromatin. Coordination of chromatin structure is required for efficient and accurate DNA replication. Aberrant DNA replication results in mutations and chromosome rearrangements that may be associated with human disorders. Therefore, cellular surveillance mechanisms have evolved to counteract potential threats to DNA replication. These mechanisms include checkpoints and specialized enzymatic activities that prevent the replication and segregation of defective DNA molecules. We employed a genome-wide approach to investigate how chromatin structure affects DNA replication under stress. We report that coordination of chromatin assembly and checkpoint activity by a histone modification, H2B ubiquitylation (H2Bub), is critical for the cell response to HU-induced replication stress. In cells with a mutation that abolishes H2Bub, replication progression is enhanced, and the forks are more susceptible to damage by environmental insults. The replication proteins on replicating DNA are akin to a train on the tracks, and movement of this train is carefully controlled. Our data indicate that H2Bub helps organize DNA in the nuclei during DNA replication; this process plays a similar role to the brakes on a train, serving to slow down replication, and maintaining stable progression of replication under environmental stress.


Zdroje

1. RansomM, DenneheyBK, TylerJK (2010) Chaperoning histones during DNA replication and repair. Cell 140: 183–195.

2. SoriaG, PoloSE, AlmouzniG (2012) Prime, repair, restore: the active role of chromatin in the DNA damage response. Mol Cell 46: 722–734.

3. Papamichos-ChronakisM, PetersonCL (2013) Chromatin and the genome integrity network. Nat Rev Genet 14: 62–75.

4. HanJ, ZhouH, HorazdovskyB, ZhangK, XuRM, et al. (2007) Rtt109 acetylates histone H3 lysine 56 and functions in DNA replication. Science 315: 653–655.

5. AdkinsMW, CarsonJJ, EnglishCM, RameyCJ, TylerJK (2007) The histone chaperone anti-silencing function 1 stimulates the acetylation of newly synthesized histone H3 in S-phase. J Biol Chem 282: 1334–1340.

6. BhaskaraS, ChylaBJ, AmannJM, KnutsonSK, CortezD, et al. (2008) Deletion of histone deacetylase 3 reveals critical roles in S phase progression and DNA damage control. Mol Cell 30: 61–72.

7. CelicI, MasumotoH, GriffithWP, MeluhP, CotterRJ, et al. (2006) The sirtuins hst3 and Hst4p preserve genome integrity by controlling histone h3 lysine 56 deacetylation. Curr Biol 16: 1280–1289.

8. LiQ, ZhouH, WurteleH, DaviesB, HorazdovskyB, et al. (2008) Acetylation of histone H3 lysine 56 regulates replication-coupled nucleosome assembly. Cell 134: 244–255.

9. BurgessRJ, ZhouH, HanJ, ZhangZ (2010) A role for Gcn5 in replication-coupled nucleosome assembly. Mol Cell 37: 469–480.

10. RobzykK, RechtJ, OsleyMA (2000) Rad6-dependent ubiquitination of histone H2B in yeast. Science 287: 501–504.

11. HwangWW, VenkatasubrahmanyamS, IanculescuAG, TongA, BooneC, et al. (2003) A conserved RING finger protein required for histone H2B monoubiquitination and cell size control. Mol Cell 11: 261–266.

12. WoodA, KroganNJ, DoverJ, SchneiderJ, HeidtJ, et al. (2003) Bre1, an E3 ubiquitin ligase required for recruitment and substrate selection of Rad6 at a promoter. Mol Cell 11: 267–274.

13. SongYH, AhnSH (2009) A Bre1-associated protein, large 1 (Lge1), promotes H2B Ubiquitylation during the early stages of transcription elongation. J Biol Chem 285: 2361–7 doi: 10.1074/jbc.M109.039255

14. HenryKW, WyceA, LoWS, DugganLJ, EmreNC, et al. (2003) Transcriptional activation via sequential histone H2B ubiquitylation and deubiquitylation, mediated by SAGA-associated Ubp8. Genes Dev 17: 2648–2663.

15. KaoCF, HillyerC, TsukudaT, HenryK, BergerS, et al. (2004) Rad6 plays a role in transcriptional activation through ubiquitylation of histone H2B. Genes Dev 18: 184–195.

16. BriggsSD, XiaoT, SunZW, CaldwellJA, ShabanowitzJ, et al. (2002) Gene silencing: trans-histone regulatory pathway in chromatin. Nature 418: 498.

17. DoverJ, SchneiderJ, Tawiah-BoatengMA, WoodA, DeanK, et al. (2002) Methylation of histone H3 by COMPASS requires ubiquitination of histone H2B by Rad6. J Biol Chem 277: 28368–28371.

18. NgHH, XuRM, ZhangY, StruhlK (2002) Ubiquitination of histone H2B by Rad6 is required for efficient Dot1-mediated methylation of histone H3 lysine 79. J Biol Chem 277: 34655–34657.

19. SunZW, AllisCD (2002) Ubiquitination of histone H2B regulates H3 methylation and gene silencing in yeast. Nature 418: 104–108.

20. FlemingAB, KaoCF, HillyerC, PikaartM, OsleyMA (2008) H2B ubiquitylation plays a role in nucleosome dynamics during transcription elongation. Mol Cell 31: 57–66.

21. MargaritisT, OrealV, BrabersN, MaestroniL, Vitaliano-PrunierA, et al. (2012) Two distinct repressive mechanisms for histone 3 lysine 4 methylation through promoting 3′-end antisense transcription. PLoS Genet 8: e1002952.

22. PavriR, ZhuB, LiG, TrojerP, MandalS, et al. (2006) Histone H2B monoubiquitination functions cooperatively with FACT to regulate elongation by RNA polymerase II. Cell 125: 703–717.

23. BattaK, ZhangZ, YenK, GoffmanDB, PughBF (2011) Genome-wide function of H2B ubiquitylation in promoter and genic regions. Genes Dev 25: 2254–2265.

24. EmreNC, BergerSL (2004) Histone H2B ubiquitylation and deubiquitylation in genomic regulation. Cold Spring Harb Symp Quant Biol 69: 289–299.

25. FierzB, ChatterjeeC, McGintyRK, Bar-DaganM, RaleighDP, et al. (2011) Histone H2B ubiquitylation disrupts local and higher-order chromatin compaction. Nat Chem Biol 7: 113–119.

26. MoyalL, LerenthalY, Gana-WeiszM, MassG, SoS, et al. (2011) Requirement of ATM-dependent monoubiquitylation of histone H2B for timely repair of DNA double-strand breaks. Mol Cell 41: 529–542.

27. NakamuraK, KatoA, KobayashiJ, YanagiharaH, SakamotoS, et al. (2011) Regulation of homologous recombination by RNF20-dependent H2B ubiquitination. Mol Cell 41: 515–528.

28. TrujilloKM, OsleyMA (2012) A Role for H2B Ubiquitylation in DNA Replication. Mol Cell 48: 734–46 doi: 10.1016/j.molcel.2012.09.019

29. FriedelAM, PikeBL, GasserSM (2009) ATR/Mec1: coordinating fork stability and repair. Curr Opin Cell Biol 21: 237–244.

30. BranzeiD, FoianiM (2009) The checkpoint response to replication stress. DNA Repair (Amst) 8: 1038–1046.

31. ZegermanP, DiffleyJF (2009) DNA replication as a target of the DNA damage checkpoint. DNA Repair (Amst) 8: 1077–1088.

32. AshtonTM, HicksonID (2010) Yeast as a model system to study RecQ helicase function. DNA Repair (Amst) 9: 303–314.

33. BjergbaekL, CobbJA, Tsai-PflugfelderM, GasserSM (2005) Mechanistically distinct roles for Sgs1p in checkpoint activation and replication fork maintenance. EMBO J 24: 405–417.

34. BranzeiD, FoianiM (2010) Maintaining genome stability at the replication fork. Nat Rev Mol Cell Biol 11: 208–219.

35. RodriguezJ, TsukiyamaT (2013) ATR-like kinase Mec1 facilitates both chromatin accessibility at DNA replication forks and replication fork progression during replication stress. Genes Dev 27: 74–86.

36. AlvinoGM, CollingwoodD, MurphyJM, DelrowJ, BrewerBJ, et al. (2007) Replication in hydroxyurea: it's a matter of time. Mol Cell Biol 27: 6396–6406.

37. PoliJ, TsaponinaO, CrabbeL, KeszthelyiA, PantescoV, et al. (2012) dNTP pools determine fork progression and origin usage under replication stress. EMBO J 31: 883–894.

38. XuW, AparicioJG, AparicioOM, TavareS (2006) Genome-wide mapping of ORC and Mcm2p binding sites on tiling arrays and identification of essential ARS consensus sequences in S. cerevisiae. BMC Genomics 7: 276.

39. ChabesA, StillmanB (2007) Constitutively high dNTP concentration inhibits cell cycle progression and the DNA damage checkpoint in yeast Saccharomyces cerevisiae. Proc Natl Acad Sci U S A 104: 1183–1188.

40. DavidsonMB, KatouY, KeszthelyiA, SingTL, XiaT, et al. (2012) Endogenous DNA replication stress results in expansion of dNTP pools and a mutator phenotype. EMBO J 31: 895–907.

41. GeorgievaB, ZhaoX, RothsteinR (2000) Damage response and dNTP regulation: the interaction between ribonucleotide reductase and its inhibitor, Sml1. Cold Spring Harb Symp Quant Biol 65: 343–346.

42. ZhaoX, ChabesA, DomkinV, ThelanderL, RothsteinR (2001) The ribonucleotide reductase inhibitor Sml1 is a new target of the Mec1/Rad53 kinase cascade during growth and in response to DNA damage. EMBO J 20: 3544–3553.

43. ZhaoX, RothsteinR (2002) The Dun1 checkpoint kinase phosphorylates and regulates the ribonucleotide reductase inhibitor Sml1. Proc Natl Acad Sci U S A 99: 3746–3751.

44. NavasTA, ZhouZ, ElledgeSJ (1995) DNA polymerase epsilon links the DNA replication machinery to the S phase checkpoint. Cell 80: 29–39.

45. HolmesAM, HaberJE (1999) Double-strand break repair in yeast requires both leading and lagging strand DNA polymerases. Cell 96: 415–424.

46. PaciottiV, ClericiM, ScottiM, LucchiniG, LongheseMP (2001) Characterization of mec1 kinase-deficient mutants and of new hypomorphic mec1 alleles impairing subsets of the DNA damage response pathway. Mol Cell Biol 21: 3913–3925.

47. WeinertTA, KiserGL, HartwellLH (1994) Mitotic checkpoint genes in budding yeast and the dependence of mitosis on DNA replication and repair. Genes Dev 8: 652–665.

48. CobbJA, BjergbaekL, ShimadaK, FreiC, GasserSM (2003) DNA polymerase stabilization at stalled replication forks requires Mec1 and the RecQ helicase Sgs1. EMBO J 22: 4325–4336.

49. CobbJA, SchlekerT, RojasV, BjergbaekL, TerceroJA, et al. (2005) Replisome instability, fork collapse, and gross chromosomal rearrangements arise synergistically from Mec1 kinase and RecQ helicase mutations. Genes Dev 19: 3055–3069.

50. HegnauerAM, HustedtN, ShimadaK, PikeBL, VogelM, et al. (2012) An N-terminal acidic region of Sgs1 interacts with Rpa70 and recruits Rad53 kinase to stalled forks. EMBO J 31: 3768–3783.

51. GrothA, CorpetA, CookAJ, RocheD, BartekJ, et al. (2007) Regulation of replication fork progression through histone supply and demand. Science 318: 1928–1931.

52. FrancoAA, LamWM, BurgersPM, KaufmanPD (2005) Histone deposition protein Asf1 maintains DNA replisome integrity and interacts with replication factor C. Genes Dev 19: 1365–1375.

53. DriscollR, HudsonA, JacksonSP (2007) Yeast Rtt109 promotes genome stability by acetylating histone H3 on lysine 56. Science 315: 649–652.

54. GreenEM, AntczakAJ, BaileyAO, FrancoAA, WuKJ, et al. (2005) Replication-independent histone deposition by the HIR complex and Asf1. Curr Biol 15: 2044–2049.

55. EmiliA, SchieltzDM, YatesJR3rd, HartwellLH (2001) Dynamic interaction of DNA damage checkpoint protein Rad53 with chromatin assembly factor Asf1. Mol Cell 7: 13–20.

56. HuF, AlcasabasAA, ElledgeSJ (2001) Asf1 links Rad53 to control of chromatin assembly. Genes Dev 15: 1061–1066.

57. TerceroJA, DiffleyJF (2001) Regulation of DNA replication fork progression through damaged DNA by the Mec1/Rad53 checkpoint. Nature 412: 553–557.

58. SeguradoM, DiffleyJF (2008) Separate roles for the DNA damage checkpoint protein kinases in stabilizing DNA replication forks. Genes Dev 22: 1816–1827.

59. Clemente-RuizM, PradoF (2009) Chromatin assembly controls replication fork stability. EMBO Rep 10: 790–796.

60. Clemente-RuizM, Gonzalez-PrietoR, PradoF (2011) Histone H3K56 acetylation, CAF1, and Rtt106 coordinate nucleosome assembly and stability of advancing replication forks. PLoS Genet 7: e1002376.

61. MejlvangJ, FengY, AlabertC, NeelsenKJ, JasencakovaZ, et al. (2014) New histone supply regulates replication fork speed and PCNA unloading. J Cell Biol 204: 29–43.

62. GrothA, Ray-GalletD, QuivyJP, LukasJ, BartekJ, et al. (2005) Human Asf1 regulates the flow of S phase histones during replicational stress. Mol Cell 17: 301–311.

63. HoekM, StillmanB (2003) Chromatin assembly factor 1 is essential and couples chromatin assembly to DNA replication in vivo. Proc Natl Acad Sci U S A 100: 12183–12188.

64. NabatiyanA, KrudeT (2004) Silencing of chromatin assembly factor 1 in human cells leads to cell death and loss of chromatin assembly during DNA synthesis. Mol Cell Biol 24: 2853–2862.

65. YeX, FrancoAA, SantosH, NelsonDM, KaufmanPD, et al. (2003) Defective S phase chromatin assembly causes DNA damage, activation of the S phase checkpoint, and S phase arrest. Mol Cell 11: 341–351.

66. KatsES, AlbuquerqueCP, ZhouH, KolodnerRD (2006) Checkpoint functions are required for normal S-phase progression in Saccharomyces cerevisiae RCAF- and CAF-I-defective mutants. Proc Natl Acad Sci U S A 103: 3710–3715.

67. RykowskiMC, WallisJW, ChoeJ, GrunsteinM (1981) Histone H2B subtypes are dispensable during the yeast cell cycle. Cell 25: 477–487.

68. NorrisD, OsleyMA (1987) The two gene pairs encoding H2A and H2B play different roles in the Saccharomyces cerevisiae life cycle. Mol Cell Biol 7: 3473–3481.

69. GunjanA, VerreaultA (2003) A Rad53 kinase-dependent surveillance mechanism that regulates histone protein levels in S. cerevisiae. Cell 115: 537–549.

70. HwangWW, MadhaniHD (2009) Nonredundant requirement for multiple histone modifications for the early anaphase release of the mitotic exit regulator Cdc14 from nucleolar chromatin. PLoS Genet 5: e1000588.

71. BuddME, CampbellJL (1993) DNA polymerases delta and epsilon are required for chromosomal replication in Saccharomyces cerevisiae. Mol Cell Biol 13: 496–505.

72. SchererS, DavisRW (1979) Replacement of chromosome segments with altered DNA sequences constructed in vitro. Proc Natl Acad Sci U S A 76: 4951–4955.

73. FachinettiD, BermejoR, CocitoA, MinardiS, KatouY, et al. (2010) Replication termination at eukaryotic chromosomes is mediated by Top2 and occurs at genomic loci containing pausing elements. Mol Cell 39: 595–605.

74. KatouY, KanohY, BandoM, NoguchiH, TanakaH, et al. (2003) S-phase checkpoint proteins Tof1 and Mrc1 form a stable replication-pausing complex. Nature 424: 1078–1083.

75. ScaffidiP, MisteliT (2008) Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing. Nat Cell Biol 10: 452–459.

76. DahlKN, ScaffidiP, IslamMF, YodhAG, WilsonKL, et al. (2006) Distinct structural and mechanical properties of the nuclear lamina in Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A 103: 10271–10276.

77. SchubelerD, ScalzoD, KooperbergC, van SteenselB, DelrowJ, et al. (2002) Genome-wide DNA replication profile for Drosophila melanogaster: a link between transcription and replication timing. Nat Genet 32: 438–442.

78. WuhrM, ChenY, DumontS, GroenAC, NeedlemanDJ, et al. (2008) Evidence for an upper limit to mitotic spindle length. Curr Biol 18: 1256–1261.

79. PellicioliA, LuccaC, LiberiG, MariniF, LopesM, et al. (1999) Activation of Rad53 kinase in response to DNA damage and its effect in modulating phosphorylation of the lagging strand DNA polymerase. EMBO J 18: 6561–6572.

80. BermejoR, DoksaniY, CapraT, KatouYM, TanakaH, et al. (2007) Top1- and Top2-mediated topological transitions at replication forks ensure fork progression and stability and prevent DNA damage checkpoint activation. Genes Dev 21: 1921–1936.

81. KumarD, VibergJ, NilssonAK, ChabesA (2010) Highly mutagenic and severely imbalanced dNTP pools can escape detection by the S-phase checkpoint. Nucleic Acids Res 38: 3975–3983.

82. JiangC, PughBF (2009) A compiled and systematic reference map of nucleosome positions across the Saccharomyces cerevisiae genome. Genome Biol 10: R109.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 10
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#