#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Components of a Fanconi-Like Pathway Control Pso2-Independent DNA Interstrand Crosslink Repair in Yeast


Fanconi anemia (FA) is a devastating genetic disease, associated with genomic instability and defects in DNA interstrand cross-link (ICL) repair. The FA repair pathway is not thought to be conserved in budding yeast, and although the yeast Mph1 helicase is a putative homolog of human FANCM, yeast cells disrupted for MPH1 are not sensitive to ICLs. Here, we reveal a key role for Mph1 in ICL repair when the Pso2 exonuclease is inactivated. We find that the yeast FANCM ortholog Mph1 physically and functionally interacts with Mgm101, a protein previously implicated in mitochondrial DNA repair, and the MutSα mismatch repair factor (Msh2-Msh6). Co-disruption of MPH1, MGM101, MSH6, or MSH2 with PSO2 produces a lesion-specific increase in ICL sensitivity, the elevation of ICL-induced chromosomal rearrangements, and persistence of ICL-associated DNA double-strand breaks. We find that Mph1-Mgm101-MutSα directs the ICL-induced recruitment of Exo1 to chromatin, and we propose that Exo1 is an alternative 5′-3′ exonuclease utilised for ICL repair in the absence of Pso2. Moreover, ICL-induced Rad51 chromatin loading is delayed when both Pso2 and components of the Mph1-Mgm101-MutSα and Exo1 pathway are inactivated, demonstrating that the homologous recombination stages of ICL repair are inhibited. Finally, the FANCJ- and FANCP-related factors Chl1 and Slx4, respectively, are also components of the genetic pathway controlled by Mph1-Mgm101-MutSα. Together this suggests that a prototypical FA–related ICL repair pathway operates in budding yeast, which acts redundantly with the pathway controlled by Pso2, and is required for the targeting of Exo1 to chromatin to execute ICL repair.


Vyšlo v časopise: Components of a Fanconi-Like Pathway Control Pso2-Independent DNA Interstrand Crosslink Repair in Yeast. PLoS Genet 8(8): e32767. doi:10.1371/journal.pgen.1002884
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1002884

Souhrn

Fanconi anemia (FA) is a devastating genetic disease, associated with genomic instability and defects in DNA interstrand cross-link (ICL) repair. The FA repair pathway is not thought to be conserved in budding yeast, and although the yeast Mph1 helicase is a putative homolog of human FANCM, yeast cells disrupted for MPH1 are not sensitive to ICLs. Here, we reveal a key role for Mph1 in ICL repair when the Pso2 exonuclease is inactivated. We find that the yeast FANCM ortholog Mph1 physically and functionally interacts with Mgm101, a protein previously implicated in mitochondrial DNA repair, and the MutSα mismatch repair factor (Msh2-Msh6). Co-disruption of MPH1, MGM101, MSH6, or MSH2 with PSO2 produces a lesion-specific increase in ICL sensitivity, the elevation of ICL-induced chromosomal rearrangements, and persistence of ICL-associated DNA double-strand breaks. We find that Mph1-Mgm101-MutSα directs the ICL-induced recruitment of Exo1 to chromatin, and we propose that Exo1 is an alternative 5′-3′ exonuclease utilised for ICL repair in the absence of Pso2. Moreover, ICL-induced Rad51 chromatin loading is delayed when both Pso2 and components of the Mph1-Mgm101-MutSα and Exo1 pathway are inactivated, demonstrating that the homologous recombination stages of ICL repair are inhibited. Finally, the FANCJ- and FANCP-related factors Chl1 and Slx4, respectively, are also components of the genetic pathway controlled by Mph1-Mgm101-MutSα. Together this suggests that a prototypical FA–related ICL repair pathway operates in budding yeast, which acts redundantly with the pathway controlled by Pso2, and is required for the targeting of Exo1 to chromatin to execute ICL repair.


Zdroje

1. McHughPJ, SpanswickVJ, HartleyJA (2001) Repair of DNA interstrand crosslinks: molecular mechanisms and clinical relevance. Lancet Oncol 2: 483–490.

2. DronkertML, KanaarR (2001) Repair of DNA interstrand cross-links. Mutat Res 486: 217–247.

3. LehoczkyP, McHughPJ, ChovanecM (2007) DNA interstrand cross-link repair in Saccharomyces cerevisiae. FEMS Microbiol Rev 31: 109–133.

4. RuhlandA, BrendelM (1979) Mutagenesis by cytostatic alkylating agents in yeast strains of differing repair capacities. Genetics 92: 83–97.

5. MillerRD, PrakashL, PrakashS (1982) Genetic control of excision of Saccharomyces cerevisiae interstrand DNA cross-links induced by psoralen plus near-UV light. Mol Cell Biol 2: 939–948.

6. McHughPJ, SonesWR, HartleyJA (2000) Repair of intermediate structures produced at DNA interstrand cross-links in Saccharomyces cerevisiae. Mol Cell Biol 20: 3425–3433.

7. JachymczykWJ, von BorstelRC, MowatMR, HastingsPJ (1981) Repair of interstrand cross-links in DNA of Saccharomyces cerevisiae requires two systems for DNA repair: the RAD3 system and the RAD51 system. Mol Gen Genet 182: 196–205.

8. ZhengH, WangX, WarrenAJ, LegerskiRJ, NairnRS, et al. (2003) Nucleotide excision repair- and polymerase eta-mediated error-prone removal of mitomycin C interstrand cross-links. Mol Cell Biol 23: 754–761.

9. NiedernhoferLJ, OdijkH, BudzowskaM, van DrunenE, MaasA, et al. (2004) The structure-specific endonuclease Ercc1-Xpf is required to resolve DNA interstrand cross-link-induced double-strand breaks. Mol Cell Biol 24: 5776–5787.

10. SarkarS, DaviesAA, UlrichHD, McHughPJ (2006) DNA interstrand crosslink repair during G1 involves nucleotide excision repair and DNA polymerase zeta. Embo J 25: 1285–1294.

11. RaschleM, KnipscheerP, EnoiuM, AngelovT, SunJ, et al. (2008) Mechanism of replication-coupled DNA interstrand crosslink repair. Cell 134: 969–980.

12. NiedzwiedzW, MosedaleG, JohnsonM, OngCY, PaceP, et al. (2004) The Fanconi anaemia gene FANCC promotes homologous recombination and error-prone DNA repair. Mol Cell 15: 607–620.

13. KnipscheerP, RaschleM, SmogorzewskaA, EnoiuM, HoTV, et al. (2009) The Fanconi anemia pathway promotes replication-dependent DNA interstrand cross-link repair. Science 326: 1698–1701.

14. LongDT, RaschleM, JoukovV, WalterJC (2011) Mechanism of RAD51-dependent DNA interstrand cross-link repair. Science 333: 84–87.

15. JoenjeH, PatelKJ (2001) The emerging genetic and molecular basis of Fanconi anaemia. Nat Rev Genet 2: 446–457.

16. MeeteiAR, MedhurstAL, LingC, XueY, SinghTR, et al. (2005) A human ortholog of archaeal DNA repair protein Hef is defective in Fanconi anemia complementation group M. Nat Genet 37: 958–963.

17. MosedaleG, NiedzwiedzW, AlpiA, PerrinaF, Pereira-LealJB, et al. (2005) The vertebrate Hef ortholog is a component of the Fanconi anemia tumor-suppressor pathway. Nat Struct Mol Biol 12: 763–771.

18. EntianKD, SchusterT, HegemannJH, BecherD, FeldmannH, et al. (1999) Functional analysis of 150 deletion mutants in Saccharomyces cerevisiae by a systematic approach. Mol Gen Genet 262: 683–702.

19. SchellerJ, SchurerA, RudolphC, HettwerS, KramerW (2000) MPH1, a yeast gene encoding a DEAH protein, plays a role in protection of the genome from spontaneous and chemically induced damage. Genetics 155: 1069–1081.

20. SchurerKA, RudolphC, UlrichHD, KramerW (2004) Yeast MPH1 gene functions in an error-free DNA damage bypass pathway that requires genes from Homologous recombination, but not from postreplicative repair. Genetics 166: 1673–1686.

21. SunW, NandiS, OsmanF, AhnJS, JakovleskaJ, et al. (2008) The FANCM ortholog Fml1 promotes recombination at stalled replication forks and limits crossing over during DNA double-strand break repair. Mol Cell 32: 118–128.

22. BanerjeeS, SmithS, OumJH, LiawHJ, HwangJY, et al. (2008) Mph1p promotes gross chromosomal rearrangement through partial inhibition of homologous recombination. J Cell Biol 181: 1083–1093.

23. PrakashR, SatoryD, DrayE, PapushaA, SchellerJ, et al. (2009) Yeast Mph1 helicase dissociates Rad51-made D-loops: implications for crossover control in mitotic recombination. Genes Dev 23: 67–79.

24. PrakashR, KrejciL, Van KomenS, Anke SchurerK, KramerW, et al. (2005) Saccharomyces cerevisiae MPH1 gene, required for homologous recombination-mediated mutation avoidance, encodes a 3′ to 5′ DNA helicase. J Biol Chem 280: 7854–7860.

25. GariK, DecailletC, StasiakAZ, StasiakA, ConstantinouA (2008) The Fanconi anemia protein FANCM can promote branch migration of Holliday junctions and replication forks. Mol Cell 29: 141–148.

26. XueY, LiY, GuoR, LingC, WangW (2008) FANCM of the Fanconi anemia core complex is required for both monoubiquitination and DNA repair. Hum Mol Genet

27. RosadoIV, NiedzwiedzW, AlpiAF, PatelKJ (2009) The Walker B motif in avian FANCM is required to limit sister chromatid exchanges but is dispensable for DNA crosslink repair. Nucleic Acids Res 37: 4360–4370.

28. SchwabRA, BlackfordAN, NiedzwiedzW (2010) ATR activation and replication fork restart are defective in FANCM-deficient cells. Embo J 29: 806–818.

29. CollisSJ, CicciaA, DeansAJ, HorejsiZ, MartinJS, et al. (2008) FANCM and FAAP24 function in ATR-mediated checkpoint signaling independently of the Fanconi anemia core complex. Mol Cell 32: 313–324.

30. Luke-GlaserS, LukeB, GrossiS, ConstantinouA (2010) FANCM regulates DNA chain elongation and is stabilized by S-phase checkpoint signalling. Embo J 29: 795–805.

31. HenriquesJA, MoustacchiE (1980) Isolation and characterization of pso mutants sensitive to photo-addition of psoralen derivatives in Saccharomyces cerevisiae. Genetics 95: 273–288.

32. RuhlandA, HaaseE, SiedeW, BrendelM (1981) Isolation of yeast mutants sensitive to the bifunctional alkylating agent nitrogen mustard. Mol Gen Genet 181: 346–351.

33. RuhlandA, KircherM, WilbornF, BrendelM (1981) A yeast mutant specifically sensitive to bifunctional alkylation. Mutat Res 91: 457–462.

34. GrossmannKF, WardAM, MosesRE (2000) Saccharomyces cerevisiae lacking Snm1, Rev3 or Rad51 have a normal S-phase but arrest permanently in G2 after cisplatin treatment. Mutat Res 461: 1–13.

35. Magana-SchwenckeN, HenriquesJA, ChanetR, MoustacchiE (1982) The fate of 8-methoxypsoralen photoinduced crosslinks in nuclear and mitochondrial yeast DNA: comparison of wild-type and repair-deficient strains. Proc Natl Acad Sci U S A 79: 1722–1726.

36. WilbornF, BrendelM (1989) Formation and stability of interstrand cross-links induced by cis- and trans-diamminedichloroplatinum (II) in the DNA of Saccharomyces cerevisiae strains differing in repair capacity. Curr Genet 16: 331–338.

37. LiX, MosesRE (2003) The beta-lactamase motif in Snm1 is required for repair of DNA double-strand breaks caused by interstrand crosslinks in S. cerevisiae. DNA Repair (Amst) 2: 121–129.

38. BarberLJ, WardTA, HartleyJA, McHughPJ (2005) DNA interstrand cross-link repair in the Saccharomyces cerevisiae cell cycle: overlapping roles for PSO2 (SNM1) with MutS factors and EXO1 during S phase. Mol Cell Biol 25: 2297–2309.

39. De SilvaIU, McHughPJ, ClingenPH, HartleyJA (2000) Defining the roles of nucleotide excision repair and recombination in the repair of DNA interstrand cross-links in mammalian cells. Mol Cell Biol 20: 7980–7990.

40. AkkariYM, BatemanRL, ReifsteckCA, OlsonSB, GrompeM (2000) DNA replication is required To elicit cellular responses to psoralen-induced DNA interstrand cross-links. Mol Cell Biol 20: 8283–8289.

41. BesshoT (2003) Induction of DNA replication-mediated double strand breaks by psoralen DNA interstrand cross-links. J Biol Chem 278: 5250–5254.

42. HanadaK, BudzowskaM, ModestiM, MaasA, WymanC, et al. (2006) The structure-specific endonuclease Mus81-Eme1 promotes conversion of interstrand DNA crosslinks into double-strands breaks. Embo J 25: 4921–4932.

43. WangAT, SengerovaB, CattellE, InagawaT, HartleyJM, et al. (2011) Human SNM1A and XPF-ERCC1 collaborate to initiate DNA interstrand cross-link repair. Genes Dev 25: 1859–1870.

44. ZhangN, LuX, ZhangX, PetersonCA, LegerskiRJ (2002) hMutSbeta is required for the recognition and uncoupling of psoralen interstrand cross-links in vitro. Mol Cell Biol 22: 2388–2397.

45. WuQ, ChristensenLA, LegerskiRJ, VasquezKM (2005) Mismatch repair participates in error-free processing of DNA interstrand crosslinks in human cells. EMBO Rep 6: 551–557.

46. LanL, HayashiT, RabeyaRM, NakajimaS, KannoS, et al. (2004) Functional and physical interactions between ERCC1 and MSH2 complexes for resistance to cis-diamminedichloroplatinum(II) in mammalian cells. DNA Repair (Amst) 3: 135–143.

47. HoY, GruhlerA, HeilbutA, BaderGD, MooreL, et al. (2002) Systematic identification of protein complexes in Saccharomyces cerevisiae by mass spectrometry. Nature 415: 180–183.

48. ChenXJ, GuanMX, Clark-WalkerGD (1993) MGM101, a nuclear gene involved in maintenance of the mitochondrial genome in Saccharomyces cerevisiae. Nucleic Acids Res 21: 3473–3477.

49. MeeusenS, TieuQ, WongE, WeissE, SchieltzD, et al. (1999) Mgm101p is a novel component of the mitochondrial nucleoid that binds DNA and is required for the repair of oxidatively damaged mitochondrial DNA. J Cell Biol 145: 291–304.

50. MeeusenS, NunnariJ (2003) Evidence for a two membrane-spanning autonomous mitochondrial DNA replisome. J Cell Biol 163: 503–510.

51. MbantenkhuM, WangX, NardozziJD, WilkensS, HoffmanE, et al. (2011) Mgm101 is a Rad52-related protein required for mitochondrial DNA recombination. J Biol Chem

52. ZuoX, XueD, LiN, Clark-WalkerGD (2007) A functional core of the mitochondrial genome maintenance protein Mgm101p in Saccharomyces cerevisiae determined with a temperature-conditional allele. FEMS Yeast Res 7: 131–140.

53. DrummondJT, GenschelJ, WolfE, ModrichP (1997) DHFR/MSH3 amplification in methotrexate-resistant cells alters the hMutSalpha/hMutSbeta ratio and reduces the efficiency of base-base mismatch repair. Proc Natl Acad Sci U S A 94: 10144–10149.

54. ContamineV, PicardM (2000) Maintenance and integrity of the mitochondrial genome: a plethora of nuclear genes in the budding yeast. Microbiol Mol Biol Rev 64: 281–315.

55. ReenanRA, KolodnerRD (1992) Isolation and characterization of two Saccharomyces cerevisiae genes encoding homologs of the bacterial HexA and MutS mismatch repair proteins. Genetics 132: 963–973.

56. ChenC, KolodnerRD (1999) Gross chromosomal rearrangements in Saccharomyces cerevisiae replication and recombination defective mutants. Nat Genet 23: 81–85.

57. KolodnerRD, PutnamCD, MyungK (2002) Maintenance of genome stability in Saccharomyces cerevisiae. Science 297: 552–557.

58. MyungK, DattaA, ChenC, KolodnerRD (2001) SGS1, the Saccharomyces cerevisiae homologue of BLM and WRN, suppresses genome instability and homeologous recombination. Nat Genet 27: 113–116.

59. SmithS, HwangJY, BanerjeeS, MajeedA, GuptaA, et al. (2004) Mutator genes for suppression of gross chromosomal rearrangements identified by a genome-wide screening in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A 101: 9039–9044.

60. TishkoffDX, BoergerAL, BertrandP, FilosiN, GaidaGM, et al. (1997) Identification and characterization of Saccharomyces cerevisiae EXO1, a gene encoding an exonuclease that interacts with MSH2. Proc Natl Acad Sci U S A 94: 7487–7492.

61. TranPT, ErdenizN, DudleyS, LiskayRM (2002) Characterization of nuclease-dependent functions of Exo1p in Saccharomyces cerevisiae. DNA Repair (Amst) 1: 895–912.

62. SymingtonLS, GautierJ (2010) Double-Strand Break End Resection and Repair Pathway Choice. Annu Rev Genet

63. WhiteMF (2009) Structure, function and evolution of the XPD family of iron-sulfur-containing 5′→3′ DNA helicases. Biochem Soc Trans 37: 547–551.

64. StoepkerC, HainK, SchusterB, Hilhorst-HofsteeY, RooimansMA, et al. (2011) SLX4, a coordinator of structure-specific endonucleases, is mutated in a new Fanconi anemia subtype. Nat Genet 43: 138–141.

65. CrossanGP, van der WeydenL, RosadoIV, LangevinF, GaillardPH, et al. (2011) Disruption of mouse Slx4, a regulator of structure-specific nucleases, phenocopies Fanconi anemia. Nat Genet 43: 147–152.

66. KimY, LachFP, DesettyR, HanenbergH, AuerbachAD, et al. (2011) Mutations of the SLX4 gene in Fanconi anemia. Nat Genet 43: 142–146.

67. ZuoXM, Clark-WalkerGD, ChenXJ (2002) The mitochondrial nucleoid protein, Mgm101p, of Saccharomyces cerevisiae is involved in the maintenance of rho(+) and ori/rep-devoid petite genomes but is not required for hypersuppressive rho(−) mtDNA. Genetics 160: 1389–1400.

68. De SilvaIU, McHughPJ, ClingenPH, HartleyJA (2002) Defects in interstrand cross-link uncoupling do not account for the extreme sensitivity of ERCC1 and XPF cells to cisplatin. Nucleic Acids Res 30: 3848–3856.

69. LiX, HejnaJ, MosesRE (2005) The yeast Snm1 protein is a DNA 5′-exonuclease. DNA Repair (Amst) 4: 163–170.

70. HazratiA, Ramis-CastelltortM, SarkarS, BarberLJ, SchofieldCJ, et al. (2007) Human SNM1A rescues the DNA repair defects of yeast pso2 mutants. DNA Repair (Amst) in press.

71. PengM, LitmanR, XieJ, SharmaS, BroshRMJr, et al. (2007) The FANCJ/MutLalpha interaction is required for correction of the cross-link response in FA-J cells. Embo J 26: 3238–3249.

72. WilliamsSA, WilsonJB, ClarkAP, Mitson-SalazarA, TomashevskiA, et al. (2011) Functional and physical interaction between the mismatch repair and FA-BRCA pathways. Hum Mol Genet

73. HuangM, KennedyR, AliAM, MoreauLA, MeeteiAR, et al. (2011) Human MutS and FANCM complexes function as redundant DNA damage sensors in the Fanconi Anemia pathway. DNA Repair (Amst)

74. HemphillAW, BruunD, ThrunL, AkkariY, TorimaruY, et al. (2008) Mammalian SNM1 is required for genome stability. Mol Genet Metab 94: 38–45.

75. YangK, MoldovanGL, D'AndreaAD (2010) RAD18-dependent recruitment of SNM1A to DNA repair complexes by a ubiquitin-binding zinc finger. J Biol Chem 285: 19085–19091.

76. MarsitCJ, LiuM, NelsonHH, PosnerM, SuzukiM, et al. (2004) Inactivation of the Fanconi anemia/BRCA pathway in lung and oral cancers: implications for treatment and survival. Oncogene 23: 1000–1004.

77. WreesmannVB, EstiloC, EiseleDW, SinghB, WangSJ (2007) Downregulation of Fanconi anemia genes in sporadic head and neck squamous cell carcinoma. ORL J Otorhinolaryngol Relat Spec 69: 218–225.

78. TaniguchiT, TischkowitzM, AmezianeN, HodgsonSV, MathewCG, et al. (2003) Disruption of the Fanconi anemia-BRCA pathway in cisplatin-sensitive ovarian tumors. Nat Med 9: 568–574.

79. JiricnyJ (1998) Eukaryotic mismatch repair: an update. Mutat Res 409: 107–121.

80. WachA, BrachatA, PohlmannR, PhilippsenP (1994) New heterologous modules for classical or PCR-based gene disruptions in Saccharomyces cerevisiae. Yeast 10: 1793–1808.

81. LongtineMS, McKenzieA3rd, DemariniDJ, ShahNG, WachA, et al. (1998) Additional modules for versatile and economical PCR-based gene deletion and modification in Saccharomyces cerevisiae. Yeast 14: 953–961.

82. GoldsteinAL, McCuskerJH (1999) Three new dominant drug resistance cassettes for gene disruption in Saccharomyces cerevisiae. Yeast 15: 1541–1553.

83. TongAH, EvangelistaM, ParsonsAB, XuH, BaderGD, et al. (2001) Systematic genetic analysis with ordered arrays of yeast deletion mutants. Science 294: 2364–2368.

84. SuckauD, ResemannA, SchuerenbergM, HufnagelP, FranzenJ, et al. (2003) A novel MALDI LIFT-TOF/TOF mass spectrometer for proteomics. Anal Bioanal Chem 376: 952–965.

85. ZinserE, DaumG (1995) Isolation and biochemical characterization of organelles from the yeast, Saccharomyces cerevisiae. Yeast 11: 493–536.

86. LiangC, StillmanB (1997) Persistent initiation of DNA replication and chromatin-bound MCM proteins during the cell cycle in cdc6 mutants. Genes Dev 11: 3375–3386.

87. McHughPJ, GillRD, WatersR, HartleyJA (1999) Excision repair of nitrogen mustard-DNA adducts in Saccharomyces cerevisiae. Nucleic Acids Res 27: 3259–3266.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 8
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#