#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Synergistic Interaction of Rnf8 and p53 in the Protection against Genomic Instability and Tumorigenesis


Rnf8 is an E3 ubiquitin ligase that plays a key role in the DNA damage response as well as in the maintenance of telomeres and chromatin remodeling. Rnf8−/− mice exhibit developmental defects and increased susceptibility to tumorigenesis. We observed that levels of p53, a central regulator of the cellular response to DNA damage, increased in Rnf8−/− mice in a tissue- and cell type–specific manner. To investigate the role of the p53-pathway inactivation on the phenotype observed in Rnf8−/− mice, we have generated Rnf8−/−p53−/− mice. Double-knockout mice showed similar growth retardation defects and impaired class switch recombination compared to Rnf8−/− mice. In contrast, loss of p53 fully rescued the increased apoptosis and reduced number of thymocytes and splenocytes in Rnf8−/− mice. Similarly, the senescence phenotype of Rnf8−/− mouse embryonic fibroblasts was rescued in p53 null background. Rnf8−/−p53−/− cells displayed defective cell cycle checkpoints and DNA double-strand break repair. In addition, Rnf8−/−p53−/− mice had increased levels of genomic instability and a remarkably elevated tumor incidence compared to either Rnf8−/− or p53−/− mice. Altogether, the data in this study highlight the importance of p53-pathway activation upon loss of Rnf8, suggesting that Rnf8 and p53 functionally interact to protect against genomic instability and tumorigenesis.


Vyšlo v časopise: Synergistic Interaction of Rnf8 and p53 in the Protection against Genomic Instability and Tumorigenesis. PLoS Genet 9(1): e32767. doi:10.1371/journal.pgen.1003259
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003259

Souhrn

Rnf8 is an E3 ubiquitin ligase that plays a key role in the DNA damage response as well as in the maintenance of telomeres and chromatin remodeling. Rnf8−/− mice exhibit developmental defects and increased susceptibility to tumorigenesis. We observed that levels of p53, a central regulator of the cellular response to DNA damage, increased in Rnf8−/− mice in a tissue- and cell type–specific manner. To investigate the role of the p53-pathway inactivation on the phenotype observed in Rnf8−/− mice, we have generated Rnf8−/−p53−/− mice. Double-knockout mice showed similar growth retardation defects and impaired class switch recombination compared to Rnf8−/− mice. In contrast, loss of p53 fully rescued the increased apoptosis and reduced number of thymocytes and splenocytes in Rnf8−/− mice. Similarly, the senescence phenotype of Rnf8−/− mouse embryonic fibroblasts was rescued in p53 null background. Rnf8−/−p53−/− cells displayed defective cell cycle checkpoints and DNA double-strand break repair. In addition, Rnf8−/−p53−/− mice had increased levels of genomic instability and a remarkably elevated tumor incidence compared to either Rnf8−/− or p53−/− mice. Altogether, the data in this study highlight the importance of p53-pathway activation upon loss of Rnf8, suggesting that Rnf8 and p53 functionally interact to protect against genomic instability and tumorigenesis.


Zdroje

1. CicciaA, ElledgeSJ (2010) The DNA damage response: making it safe to play with knives. Mol Cell 40: 179–204.

2. BohgakiT, BohgakiM, HakemR (2010) DNA double-strand break signaling and human disorders. Genome Integr 1: 15.

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

4. 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.

5. 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.

6. WuJ, HuenMS, LuLY, YeL, DouY, et al. (2009) Histone ubiquitination associates with BRCA1-dependent DNA damage response. Mol Cell Biol 29: 849–860.

7. 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.

8. 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.

9. WangB, ElledgeSJ (2007) Ubc13/Rnf8 ubiquitin ligases control foci formation of the Rap80/Abraxas/Brca1/Brcc36 complex in response to DNA damage. Proc Natl Acad Sci U S A 104: 20759–20763.

10. Bekker-JensenS, MailandN (2011) The ubiquitin- and SUMO-dependent signaling response to DNA double-strand breaks. FEBS Lett 585: 2914–2919.

11. MalletteFA, MattiroliF, CuiG, YoungLC, HendzelMJ, et al. (2012) RNF8- and RNF168-dependent degradation of KDM4A/JMJD2A triggers 53BP1 recruitment to DNA damage sites. EMBO J 31: 1865–1878.

12. FengL, ChenJ (2012) The E3 ligase RNF8 regulates KU80 removal and NHEJ repair. Nat Struct Mol Biol 19: 201–206.

13. RaiR, LiJM, ZhengH, LokGT, DengY, et al. (2011) The E3 ubiquitin ligase Rnf8 stabilizes Tpp1 to promote telomere end protection. Nat Struct Mol Biol 18: 1400–1407.

14. LiL, HalabyMJ, HakemA, CardosoR, El GhamrasniS, et al. (2010) Rnf8 deficiency impairs class switch recombination, spermatogenesis, and genomic integrity and predisposes for cancer. J Exp Med 207: 983–997.

15. LuLY, WuJ, YeL, GavrilinaGB, SaundersTL, et al. (2010) RNF8-dependent histone modifications regulate nucleosome removal during spermatogenesis. Dev Cell 18: 371–384.

16. SantosMA, HuenMS, JankovicM, ChenHT, Lopez-ContrerasAJ, et al. (2010) Class switching and meiotic defects in mice lacking the E3 ubiquitin ligase RNF8. J Exp Med 207: 973–981.

17. LevineAJ, OrenM (2009) The first 30 years of p53: growing ever more complex. Nat Rev Cancer 9: 749–758.

18. BradyCA, AttardiLD (2010) p53 at a glance. J Cell Sci 123: 2527–2532.

19. GreenblattMS, BennettWP, HollsteinM, HarrisCC (1994) Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res 54: 4855–4878.

20. BiegingKT, AttardiLD (2012) Deconstructing p53 transcriptional networks in tumor suppression. Trends Cell Biol 22: 97–106.

21. DonehowerLA, LozanoG (2009) 20 years studying p53 functions in genetically engineered mice. Nat Rev Cancer 9: 831–841.

22. FrankKM, SharplessNE, GaoY, SekiguchiJM, FergusonDO, et al. (2000) DNA ligase IV deficiency in mice leads to defective neurogenesis and embryonic lethality via the p53 pathway. Mol Cell 5: 993–1002.

23. GaoY, FergusonDO, XieW, ManisJP, SekiguchiJ, et al. (2000) Interplay of p53 and DNA-repair protein XRCC4 in tumorigenesis, genomic stability and development. Nature 404: 897–900.

24. XuX, WagnerKU, LarsonD, WeaverZ, LiC, et al. (1999) Conditional mutation of Brca1 in mammary epithelial cells results in blunted ductal morphogenesis and tumour formation. Nat Genet 22: 37–43.

25. MakTW, HakemA, McPhersonJP, ShehabeldinA, ZablockiE, et al. (2000) Brca1 required for T cell lineage development but not TCR loci rearrangement. Nat Immunol 1: 77–82.

26. McPhersonJP, LemmersB, HiraoA, HakemA, AbrahamJ, et al. (2004) Collaboration of Brca1 and Chk2 in tumorigenesis. Genes Dev 18: 1144–1153.

27. JonkersJ, MeuwissenR, van der GuldenH, PeterseH, van der ValkM, et al. (2001) Synergistic tumor suppressor activity of BRCA2 and p53 in a conditional mouse model for breast cancer. Nat Genet 29: 418–425.

28. BohgakiT, BohgakiM, CardosoR, PanierS, ZeegersD, et al. (2011) Genomic instability, defective spermatogenesis, immunodeficiency, and cancer in a mouse model of the RIDDLE syndrome. PLoS Genet 7: e1001381 doi:10.1371/journal.pgen.1001381.

29. MeekDW (2009) Tumour suppression by p53: a role for the DNA damage response? Nat Rev Cancer 9: 714–723.

30. Reina-San-MartinB, DifilippantonioS, HanitschL, MasilamaniRF, NussenzweigA, et al. (2003) H2AX is required for recombination between immunoglobulin switch regions but not for intra-switch region recombination or somatic hypermutation. J Exp Med 197: 1767–1778.

31. WardIM, Reina-San-MartinB, OlaruA, MinnK, TamadaK, et al. (2004) 53BP1 is required for class switch recombination. J Cell Biol 165: 459–464.

32. ManisJP, MoralesJC, XiaZ, KutokJL, AltFW, et al. (2004) 53BP1 links DNA damage-response pathways to immunoglobulin heavy chain class-switch recombination. Nat Immunol 5: 481–487.

33. GuikemaJE, SchraderCE, BrodskyMH, LinehanEK, RichardsA, et al. (2010) p53 represses class switch recombination to IgG2a through its antioxidant function. J Immunol 184: 6177–6187.

34. KuilmanT, MichaloglouC, MooiWJ, PeeperDS (2010) The essence of senescence. Genes Dev 24: 2463–2479.

35. ZindyF, EischenCM, RandleDH, KamijoT, ClevelandJL, et al. (1998) Myc signaling via the ARF tumor suppressor regulates p53-dependent apoptosis and immortalization. Genes Dev 12: 2424–2433.

36. KastanMB, BartekJ (2004) Cell-cycle checkpoints and cancer. Nature 432: 316–323.

37. BoldersonE, RichardDJ, ZhouBB, KhannaKK (2009) Recent advances in cancer therapy targeting proteins involved in DNA double-strand break repair. Clin Cancer Res 15: 6314–6320.

38. WuJ, ChenY, LuLY, WuY, PaulsenMT, et al. (2011) Chfr and RNF8 synergistically regulate ATM activation. Nat Struct Mol Biol 18: 761–768.

39. YuanJ, ChenJ (2010) MRE11-RAD50-NBS1 complex dictates DNA repair independent of H2AX. J Biol Chem 285: 1097–1104.

40. MoynahanME, ChiuJW, KollerBH, JasinM (1999) Brca1 controls homology-directed DNA repair. Mol Cell 4: 511–518.

41. RoyR, ChunJ, PowellSN (2011) BRCA1 and BRCA2: different roles in a common pathway of genome protection. Nat Rev Cancer 12: 68–78.

42. BartekJ, LukasJ (2007) DNA damage checkpoints: from initiation to recovery or adaptation. Curr Opin Cell Biol 19: 238–245.

43. AttardiLD, JacksT (1999) The role of p53 in tumour suppression: lessons from mouse models. Cell Mol Life Sci 55: 48–63.

44. VenkatachalamS, ShiYP, JonesSN, VogelH, BradleyA, et al. (1998) Retention of wild-type p53 in tumors from p53 heterozygous mice: reduction of p53 dosage can promote cancer formation. EMBO J 17: 4657–4667.

45. FrenchJE, LacksGD, TrempusC, DunnickJK, FoleyJ, et al. (2001) Loss of heterozygosity frequency at the Trp53 locus in p53-deficient (+/−) mouse tumors is carcinogen-and tissue-dependent. Carcinogenesis 22: 99–106.

46. HuangTT, D'AndreaAD (2006) Regulation of DNA repair by ubiquitylation. Nat Rev Mol Cell Biol 7: 323–334.

47. HakemR, de la PompaJL, EliaA, PotterJ, MakTW (1997) Partial rescue of Brca1 (5–6) early embryonic lethality by p53 or p21 null mutation. Nat Genet 16: 298–302.

48. CaoL, LiW, KimS, BrodieSG, DengCX (2003) Senescence, aging, and malignant transformation mediated by p53 in mice lacking the Brca1 full-length isoform. Genes Dev 17: 201–213.

49. DifilippantonioS, GapudE, WongN, HuangCY, MahowaldG, et al. (2008) 53BP1 facilitates long-range DNA end-joining during V(D)J recombination. Nature 456: 529–533.

50. HinmanRM, NicholsWA, DiazTM, GallardoTD, CastrillonDH, et al. (2009) Foxo3−/− mice demonstrate reduced numbers of pre-B and recirculating B cells but normal splenic B cell sub-population distribution. Int Immunol 21: 831–842.

51. ZlotoffDA, SambandamA, LoganTD, BellJJ, SchwarzBA, et al. (2010) CCR7 and CCR9 together recruit hematopoietic progenitors to the adult thymus. Blood 115: 1897–1905.

52. StewartGS, StankovicT, ByrdPJ, WechslerT, MillerES, et al. (2007) RIDDLE immunodeficiency syndrome is linked to defects in 53BP1-mediated DNA damage signaling. Proc Natl Acad Sci U S A 104: 16910–16915.

53. ManisJP, DudleyD, KaylorL, AltFW (2002) IgH class switch recombination to IgG1 in DNA-PKcs-deficient B cells. Immunity 16: 607–617.

54. FrancoS, MurphyMM, LiG, BorjesonT, BoboilaC, et al. (2008) DNA-PKcs and Artemis function in the end-joining phase of immunoglobulin heavy chain class switch recombination. J Exp Med 205: 557–564.

55. HakemR (2008) DNA-damage repair; the good, the bad, and the ugly. EMBO J 27: 589–605.

56. MeerangM, RitzD, PaliwalS, GarajovaZ, BosshardM, et al. (2011) The ubiquitin-selective segregase VCP/p97 orchestrates the response to DNA double-strand breaks. Nat Cell Biol 13: 1376–1382.

57. AttardiLD, DonehowerLA (2005) Probing p53 biological functions through the use of genetically engineered mouse models. Mutat Res 576: 4–21.

58. DonehowerLA, HarveyM, SlagleBL, McArthurMJ, MontgomeryCAJr, et al. (1992) Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumors. Nature 356: 215–221.

59. TheunissenJW, PetriniJH (2006) Methods for studying the cellular response to DNA damage: influence of the Mre11 complex on chromosome metabolism. Methods Enzymol 409: 251–284.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 1
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#