#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Nucleosome Acidic Patch Promotes RNF168- and RING1B/BMI1-Dependent H2AX and H2A Ubiquitination and DNA Damage Signaling


Post-translational modifications of histones play important roles in regulating both the structure and function of chromatin. As all DNA based processes, including transcription, DNA replication and DNA repair, occur within the context of chromatin, the actual in vivo substrate of these reactions is chromatin. Thus, understanding these processes within the context of chromatin is vital for providing mechanistic insights into chromatin-based processes, including DNA damage signaling and genome maintenance. Here we identify a structure within H2A and H2AX termed the acidic patch that promotes the activity of two independent ubiquitin E3 ligase complexes, RNF168 and RING1B/BMI1, and is required for DNA damage ubiquitin signaling. We show directly in vitro and in vivo that this nucleosome structure is critical for histone H2A and H2AX ubiquitinations and the DNA damage response in cells. In addition, we engineered a novel biological tool that blocked the nucleosome acidic patch of all histone H2A species leading to the repression of the DNA damage response in cells. Collectively, DNA damage factors elicit their response not only through histone modifications such as ubiquitin but also through interactions within nucleosome surface structures to activate DNA damage signaling.


Vyšlo v časopise: Nucleosome Acidic Patch Promotes RNF168- and RING1B/BMI1-Dependent H2AX and H2A Ubiquitination and DNA Damage Signaling. PLoS Genet 10(3): e32767. doi:10.1371/journal.pgen.1004178
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004178

Souhrn

Post-translational modifications of histones play important roles in regulating both the structure and function of chromatin. As all DNA based processes, including transcription, DNA replication and DNA repair, occur within the context of chromatin, the actual in vivo substrate of these reactions is chromatin. Thus, understanding these processes within the context of chromatin is vital for providing mechanistic insights into chromatin-based processes, including DNA damage signaling and genome maintenance. Here we identify a structure within H2A and H2AX termed the acidic patch that promotes the activity of two independent ubiquitin E3 ligase complexes, RNF168 and RING1B/BMI1, and is required for DNA damage ubiquitin signaling. We show directly in vitro and in vivo that this nucleosome structure is critical for histone H2A and H2AX ubiquitinations and the DNA damage response in cells. In addition, we engineered a novel biological tool that blocked the nucleosome acidic patch of all histone H2A species leading to the repression of the DNA damage response in cells. Collectively, DNA damage factors elicit their response not only through histone modifications such as ubiquitin but also through interactions within nucleosome surface structures to activate DNA damage signaling.


Zdroje

1. KouzaridesT (2007) Chromatin modifications and their function. Cell 128: 693–705.

2. MillerKM, JacksonSP (2012) Histone marks: repairing DNA breaks within the context of chromatin. Biochem Soc Trans 40: 370–376.

3. RogakouEP, PilchDR, OrrAH, IvanovaVS, BonnerWM (1998) DNA double-stranded breaks induce histone H2AX phosphorylation on serine 139. J Biol Chem 273: 5858–5868.

4. PaullTT, RogakouEP, YamazakiV, KirchgessnerCU, GellertM, et al. (2000) A critical role for histone H2AX in recruitment of repair factors to nuclear foci after DNA damage. Curr Biol 10: 886–895.

5. RogakouEP, BoonC, RedonC, BonnerWM (1999) Megabase chromatin domains involved in DNA double-strand breaks in vivo. J Cell Biol 146: 905–916.

6. JacksonSP, DurocherD (2013) Regulation of DNA damage responses by ubiquitin and SUMO. Mol Cell 49: 795–807.

7. PanierS, DurocherD (2013) Push back to respond better: regulatory inhibition of the DNA double-strand break response. Nat Rev Mol Cell Biol 14(10): 661–72.

8. JacksonSP, BartekJ (2009) The DNA-damage response in human biology and disease. Nature 461: 1071–1078.

9. PoloSE, JacksonSP (2011) Dynamics of DNA damage response proteins at DNA breaks: a focus on protein modifications. Genes Dev 25: 409–433.

10. Fradet-TurcotteA, CannyMD, Escribano-DiazC, OrthweinA, LeungCC, et al. (2013) 53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark. Nature 499: 50–54.

11. MattiroliF, VissersJH, van DijkWJ, IkpaP, CitterioE, et al. (2012) RNF168 ubiquitinates K13–15 on H2A/H2AX to drive DNA damage signaling. Cell 150: 1182–1195.

12. GattiM, PinatoS, MasperoE, SoffientiniP, PoloS, et al. (2012) A novel ubiquitin mark at the N-terminal tail of histone H2As targeted by RNF168 ubiquitin ligase. Cell Cycle 11: 2538–2544.

13. ChagraouiJ, HebertJ, GirardS, SauvageauG (2011) An anticlastogenic function for the Polycomb Group gene Bmi1. Proc Natl Acad Sci U S A 108: 5284–5289.

14. FacchinoS, AbdouhM, ChatooW, BernierG (2010) BMI1 confers radioresistance to normal and cancerous neural stem cells through recruitment of the DNA damage response machinery. J Neurosci 30: 10096–10111.

15. GinjalaV, NacerddineK, KulkarniA, OzaJ, HillSJ, et al. (2011) BMI1 is recruited to DNA breaks and contributes to DNA damage-induced H2A ubiquitination and repair. Mol Cell Biol 31: 1972–1982.

16. IsmailIH, AndrinC, McDonaldD, HendzelMJ (2010) BMI1-mediated histone ubiquitylation promotes DNA double-strand break repair. J Cell Biol 191: 45–60.

17. Fradet-TurcotteA, CannyMD, Escribano-DiazC, OrthweinA, LeungCC, et al. (2013) 53BP1 is a reader of the DNA-damage-induced H2A Lys 15 ubiquitin mark. Nature 499(7456): 50–4.

18. SobhianB, ShaoG, LilliDR, CulhaneAC, MoreauLA, et al. (2007) RAP80 targets BRCA1 to specific ubiquitin structures at DNA damage sites. Science 316: 1198–1202.

19. LukasJ, LukasC, BartekJ (2011) More than just a focus: The chromatin response to DNA damage and its role in genome integrity maintenance. Nat Cell Biol 13: 1161–1169.

20. ChenWT, AlpertA, LeiterC, GongF, JacksonSP, et al. (2013) Systematic identification of functional residues in mammalian histone H2AX. Mol Cell Biol 33: 111–126.

21. WangH, WangL, Erdjument-BromageH, VidalM, TempstP, et al. (2004) Role of histone H2A ubiquitination in Polycomb silencing. Nature 431: 873–878.

22. GudjonssonT, AltmeyerM, SavicV, ToledoL, DinantC, et al. (2012) TRIP12 and UBR5 suppress spreading of chromatin ubiquitylation at damaged chromosomes. Cell 150: 697–709.

23. DoilC, MailandN, Bekker-JensenS, MenardP, LarsenDH, et al. (2009) RNF168 binds and amplifies ubiquitin conjugates on damaged chromosomes to allow accumulation of repair proteins. Cell 136: 435–446.

24. StuckiM, ClappertonJA, MohammadD, YaffeMB, SmerdonSJ, et al. (2005) MDC1 directly binds phosphorylated histone H2AX to regulate cellular responses to DNA double-strand breaks. Cell 123: 1213–1226.

25. HuenMS, GrantR, MankeI, MinnK, YuX, et al. (2007) RNF8 transduces the DNA-damage signal via histone ubiquitylation and checkpoint protein assembly. Cell 131: 901–914.

26. KolasNK, ChapmanJR, NakadaS, YlankoJ, ChahwanR, et al. (2007) Orchestration of the DNA-damage response by the RNF8 ubiquitin ligase. Science 318: 1637–1640.

27. MailandN, Bekker-JensenS, FaustrupH, MelanderF, BartekJ, et al. (2007) RNF8 ubiquitylates histones at DNA double-strand breaks and promotes assembly of repair proteins. Cell 131: 887–900.

28. FangJ, FengQ, KetelCS, WangH, CaoR, et al. (2002) Purification and functional characterization of SET8, a nucleosomal histone H4-lysine 20-specific methyltransferase. Curr Biol 12: 1086–1099.

29. IsmailIH, McDonaldD, StrickfadenH, XuZ, HendzelMJ (2013) A small molecule inhibitor of Polycomb repressive complex 1 inhibits ubiquitin signaling at DNA double-strand breaks. J Biol Chem 288(37): 26944–54.

30. PanMR, PengG, HungWC, LinSY (2011) Monoubiquitination of H2AX protein regulates DNA damage response signaling. J Biol Chem 286: 28599–28607.

31. WuCY, KangHY, YangWL, WuJ, JeongYS, et al. (2011) Critical role of monoubiquitination of histone H2AX protein in histone H2AX phosphorylation and DNA damage response. J Biol Chem 286: 30806–30815.

32. BarberaAJ, ChodaparambilJV, Kelley-ClarkeB, JoukovV, WalterJC, et al. (2006) The nucleosomal surface as a docking station for Kaposi's sarcoma herpesvirus LANA. Science 311: 856–861.

33. KatoH, van IngenH, ZhouBR, FengH, BustinM, et al. (2011) Architecture of the high mobility group nucleosomal protein 2-nucleosome complex as revealed by methyl-based NMR. Proc Natl Acad Sci U S A 108: 12283–12288.

34. LugerK, MaderAW, RichmondRK, SargentDF, RichmondTJ (1997) Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 389: 251–260.

35. MakdeRD, EnglandJR, YennawarHP, TanS (2010) Structure of RCC1 chromatin factor bound to the nucleosome core particle. Nature 467: 562–566.

36. RousselL, ErardM, CayrolC, GirardJP (2008) Molecular mimicry between IL-33 and KSHV for attachment to chromatin through the H2A-H2B acidic pocket. EMBO Rep 9: 1006–1012.

37. StewartGS, PanierS, TownsendK, Al-HakimAK, KolasNK, et al. (2009) The RIDDLE syndrome protein mediates a ubiquitin-dependent signaling cascade at sites of DNA damage. Cell 136: 420–434.

38. ZimmermannM, de LangeT (2013) 53BP1: pro choice in DNA repair. Trends Cell Biol pii: S0962-8924(13)00155-4.

39. ChapmanJR, BarralP, VannierJB, BorelV, StegerM, et al. (2013) RIF1 is essential for 53BP1-dependent nonhomologous end joining and suppression of DNA double-strand break resection. Mol Cell 49: 858–871.

40. Di VirgilioM, CallenE, YamaneA, ZhangW, JankovicM, et al. (2013) Rif1 prevents resection of DNA breaks and promotes immunoglobulin class switching. Science 339: 711–715.

41. Escribano-DiazC, OrthweinA, Fradet-TurcotteA, XingM, YoungJT, et al. (2013) A cell cycle-dependent regulatory circuit composed of 53BP1-RIF1 and BRCA1-CtIP controls DNA repair pathway choice. Mol Cell 49: 872–883.

42. FengL, FongKW, WangJ, WangW, ChenJ (2013) RIF1 counteracts BRCA1-mediated end resection during DNA repair. J Biol Chem 288: 11135–11143.

43. ZimmermannM, LottersbergerF, BuonomoSB, SfeirA, de LangeT (2013) 53BP1 regulates DSB repair using Rif1 to control 5′ end resection. Science 339: 700–704.

44. BuntingSF, CallenE, WongN, ChenHT, PolatoF, et al. (2010) 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell 141: 243–254.

45. ChaurushiyaMS, LilleyCE, AslanianA, MeisenhelderJ, ScottDC, et al. (2012) Viral E3 ubiquitin ligase-mediated degradation of a cellular E3: viral mimicry of a cellular phosphorylation mark targets the RNF8 FHA domain. Mol Cell 46: 79–90.

46. LilleyCE, ChaurushiyaMS, BoutellC, LandryS, SuhJ, et al. (2010) A viral E3 ligase targets RNF8 and RNF168 to control histone ubiquitination and DNA damage responses. EMBO J 29: 943–955.

47. WeitzmanMD, LilleyCE, ChaurushiyaMS (2010) Genomes in conflict: maintaining genome integrity during virus infection. Annu Rev Microbiol 64: 61–81.

48. KalashnikovaAA, Porter-GoffME, MuthurajanUM, LugerK, HansenJC (2013) The role of the nucleosome acidic patch in modulating higher order chromatin structure. J R Soc Interface 10: 20121022.

49. PanierS, IchijimaY, Fradet-TurcotteA, LeungCC, KaustovL, et al. (2012) Tandem protein interaction modules organize the ubiquitin-dependent response to DNA double-strand breaks. Mol Cell 47: 383–395.

50. BentleyML, CornJE, DongKC, PhungQ, CheungTK, et al. (2011) Recognition of UbcH5c and the nucleosome by the Bmi1/Ring1b ubiquitin ligase complex. EMBO J 30: 3285–3297.

51. TjeertesJV, MillerKM, JacksonSP (2009) Screen for DNA-damage-responsive histone modifications identifies H3K9Ac and H3K56Ac in human cells. EMBO J 28: 1878–1889.

52. BarberaAJ, BallestasME, KayeKM (2004) The Kaposi's sarcoma-associated herpesvirus latency-associated nuclear antigen 1 N terminus is essential for chromosome association, DNA replication, and episome persistence. J Virol 78: 294–301.

53. SheeC, CoxBD, GuF, LuengasEM, JoshiMC, et al. (2013) Engineered proteins detect spontaneous DNA breakage in human and bacterial cells. Elife 2: e01222.

54. MillerKM, TjeertesJV, CoatesJ, LegubeG, PoloSE, et al. (2010) Human HDAC1 and HDAC2 function in the DNA-damage response to promote DNA nonhomologous end-joining. Nat Struct Mol Biol 17: 1144–1151.

55. BartkeT, VermeulenM, XhemalceB, RobsonSC, MannM, et al. (2010) Nucleosome-interacting proteins regulated by DNA and histone methylation. Cell 143: 470–484.

56. DyerPN, EdayathumangalamRS, WhiteCL, BaoY, ChakravarthyS, et al. (2004) Reconstitution of nucleosome core particles from recombinant histones and DNA. Methods Enzymol 375: 23–44.

57. LeeJY, GreeneEC (2011) Assembly of recombinant nucleosomes on nanofabricated DNA curtains for single-molecule imaging. Methods Mol Biol 778: 243–258.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 3
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#