#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Conditional Inactivation of the DNA Damage Response Gene in Mouse Testis Reveals Separable Roles for Components of the RAD9-RAD1-HUS1 Complex in Meiotic Chromosome Maintenance


The RAD9-RAD1-HUS1 (9-1-1) complex is a heterotrimeric PCNA-like clamp that responds to DNA damage in somatic cells by promoting DNA repair as well as ATR-dependent DNA damage checkpoint signaling. In yeast, worms, and flies, the 9-1-1 complex is also required for meiotic checkpoint function and efficient completion of meiotic recombination; however, since Rad9, Rad1, and Hus1 are essential genes in mammals, little is known about their functions in mammalian germ cells. In this study, we assessed the meiotic functions of 9-1-1 by analyzing mice with germ cell-specific deletion of Hus1 as well as by examining the localization of RAD9 and RAD1 on meiotic chromosomes during prophase I. Hus1 loss in testicular germ cells resulted in meiotic defects, germ cell depletion, and severely compromised fertility. Hus1-deficient primary spermatocytes exhibited persistent autosomal γH2AX and RAD51 staining indicative of unrepaired meiotic DSBs, synapsis defects, an extended XY body domain often encompassing partial or whole autosomes, and an increase in structural chromosome abnormalities such as end-to-end X chromosome-autosome fusions and ruptures in the synaptonemal complex. Most of these aberrations persisted in diplotene-stage spermatocytes. Consistent with a role for the 9-1-1 complex in meiotic DSB repair, RAD9 localized to punctate, RAD51-containing foci on meiotic chromosomes in a Hus1-dependent manner. Interestingly, RAD1 had a broader distribution that only partially overlapped with RAD9, and localization of both RAD1 and the ATR activator TOPBP1 to the XY body and to unsynapsed autosomes was intact in Hus1 conditional knockouts. We conclude that mammalian HUS1 acts as a component of the canonical 9-1-1 complex during meiotic prophase I to promote DSB repair and further propose that RAD1 and TOPBP1 respond to unsynapsed chromatin through an alternative mechanism that does not require RAD9 or HUS1.


Vyšlo v časopise: Conditional Inactivation of the DNA Damage Response Gene in Mouse Testis Reveals Separable Roles for Components of the RAD9-RAD1-HUS1 Complex in Meiotic Chromosome Maintenance. PLoS Genet 9(2): e32767. doi:10.1371/journal.pgen.1003320
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003320

Souhrn

The RAD9-RAD1-HUS1 (9-1-1) complex is a heterotrimeric PCNA-like clamp that responds to DNA damage in somatic cells by promoting DNA repair as well as ATR-dependent DNA damage checkpoint signaling. In yeast, worms, and flies, the 9-1-1 complex is also required for meiotic checkpoint function and efficient completion of meiotic recombination; however, since Rad9, Rad1, and Hus1 are essential genes in mammals, little is known about their functions in mammalian germ cells. In this study, we assessed the meiotic functions of 9-1-1 by analyzing mice with germ cell-specific deletion of Hus1 as well as by examining the localization of RAD9 and RAD1 on meiotic chromosomes during prophase I. Hus1 loss in testicular germ cells resulted in meiotic defects, germ cell depletion, and severely compromised fertility. Hus1-deficient primary spermatocytes exhibited persistent autosomal γH2AX and RAD51 staining indicative of unrepaired meiotic DSBs, synapsis defects, an extended XY body domain often encompassing partial or whole autosomes, and an increase in structural chromosome abnormalities such as end-to-end X chromosome-autosome fusions and ruptures in the synaptonemal complex. Most of these aberrations persisted in diplotene-stage spermatocytes. Consistent with a role for the 9-1-1 complex in meiotic DSB repair, RAD9 localized to punctate, RAD51-containing foci on meiotic chromosomes in a Hus1-dependent manner. Interestingly, RAD1 had a broader distribution that only partially overlapped with RAD9, and localization of both RAD1 and the ATR activator TOPBP1 to the XY body and to unsynapsed autosomes was intact in Hus1 conditional knockouts. We conclude that mammalian HUS1 acts as a component of the canonical 9-1-1 complex during meiotic prophase I to promote DSB repair and further propose that RAD1 and TOPBP1 respond to unsynapsed chromatin through an alternative mechanism that does not require RAD9 or HUS1.


Zdroje

1. EichingerCS, JentschS (2011) 9-1-1: PCNA's specialized cousin. Trends Biochem Sci 36: 563–568.

2. WeissRS, LederP, VaziriC (2003) Critical role for mouse Hus1 in an S-phase DNA damage cell cycle checkpoint. Mol Cell Biol 23: 791–803.

3. ZouL, CortezD, ElledgeSJ (2002) Regulation of ATR substrate selection by Rad17-dependent loading of Rad9 complexes onto chromatin. Genes Dev 16: 198–208.

4. Navadgi-PatilVM, BurgersPM (2009) A tale of two tails: Activation of DNA damage checkpoint kinase Mec1/ATR by the 9-1-1 clamp and by Dpb11/TopBP1. DNA Repair (Amst) 8: 996–1003.

5. XuY-j, LeffakM (2010) ATRIP from TopBP1 to ATR—in vitro activation of a DNA damage checkpoint. Proc Natl Acad Sci USA 107: 13561–13562.

6. Parrilla-CastellarER, ArlanderSJH, KarnitzL (2004) Dial 9-1-1 for DNA damage: the Rad9-Hus1-Rad1 (9-1-1) clamp complex. DNA Repair (Amst) 3: 1009–1014.

7. PanditaRK, SharmaGG, LaszloA, HopkinsKM, DaveyS, et al. (2006) Mammalian Rad9 plays a role in telomere stability, S- and G2-phase-specific cell survival, and homologous recombinational repair. Mol Cell Biol 26: 1850–1864.

8. SabbionedaS, MinesingerBK, GiannattasioM, PlevaniP, Muzi-FalconiM, et al. (2005) The 9-1-1 checkpoint clamp physically interacts with Polzeta and is partially required for spontaneous Polzeta-dependent mutagenesis in Saccharomyces cerevisiae. J Biol Chem 38657–38665.

9. ToueilleM, El-AndaloussiN, FrouinI, FreireR, FunkD, et al. (2004) The human Rad9/Rad1/Hus1 damage sensor clamp interacts with DNA polymerase β and increases its DNA substrate utilisation efficiency: implications for DNA repair. Nucleic Acids Res 32: 3316–3324.

10. GembkaA, ToueilleM, SmirnovaE, PoltzR, FerrariE, et al. (2007) The checkpoint clamp, Rad9-Rad1-Hus1 complex, preferentially stimulates the activity of apurinic/apyrimidinic endonuclease 1 and DNA polymerase β in long patch base excision repair. Nucleic Acids Res 35: 2596–2608.

11. WangW, BrandtP, RossiML, Lindsey-BoltzL, PodustV, et al. (2004) The human Rad9-Rad1-Hus1 checkpoint complex stimulates flap endonuclease 1. Proc Natl Acad Sci USA 101: 16762–16767.

12. Friedrich-HeinekenE, ToueilleM, TännlerB, BürkiC, FerrariE, et al. (2005) The two DNA clamps Rad9/Rad1/Hus1 complex and Proliferating Cell Nuclear Antigen differentially regulate Flap Endonuclease 1 activity. J Mol Biol 353: 980–989.

13. SongW, PascalJM, EllenbergerT, TomkinsonAE (2009) The DNA binding domain of human DNA ligase I interacts with both nicked DNA and the DNA sliding clamps, PCNA and hRad9-hRad1-hHus1. DNA Repair (Amst) 8: 912–919.

14. GuanX, MadabushiA, ChangD-Y, FitzgeraldME, ShiG, et al. (2007) The human checkpoint sensor Rad9 Rad1 Hus1 interacts with and stimulates DNA repair enzyme TDG glycosylase. Nucleic Acids Res 35: 6207–6218.

15. GuanX, BaiH, ShiG, TheriotCA, HazraTK, et al. (2007) The human checkpoint sensor Rad9-Rad1-Hus1 interacts with and stimulates NEIL1 glycosylase. Nucleic Acids Res 35: 2463–2472.

16. ChangD-Y, LuA-L (2005) Interaction of checkpoint proteins Hus1/Rad1/Rad9 with DNA base excision repair enzyme MutY homolog in fission yeast, Schizosaccharomyces pombe. J Biol Chem 280: 408–417.

17. ShiG, ChangD-Y, ChengC-C, GuanX, VenclovasÄ, et al. (2006) Physical and functional interactions between MutY glycosylase homologue (MYH) and checkpoint proteins Rad9-Rad1-Hus1. Biochem J 400: 53–62.

18. ParkMJ, ParkJ-H, HahmS-H, KoSI, LeeYR, et al. (2009) Repair activities of human 8-oxoguanine DNA glycosylase are stimulated by the interaction with human checkpoint sensor Rad9–Rad1–Hus1 complex. DNA Repair (Amst) 8: 1190–1200.

19. CaiRL, Yan-NealeY, CuetoMA, XuH, CohenD (2000) HDAC1, a histone deacetylase, forms a complex with Hus1 and Rad9, two G2/M checkpoint Rad proteins. J Biol Chem 275: 27909–27916.

20. PichierriP, NicolaiS, CignoloL, BignamiM, FranchittoA (2011) The RAD9-RAD1-HUS1 (9.1.1) complex interacts with WRN and is crucial to regulate its response to replication fork stalling. Oncogene doi:10.1038/onc.2011.1468

21. HeW, ZhaoY, ZhangC, AnL, HuZ, et al. (2008) Rad9 plays an important role in DNA mismatch repair through physical interaction with MLH1. Nucleic Acids Res 36: 6406–6417.

22. BaiH, MadabushiA, GuanX, LuAL (2010) Interaction between human mismatch repair recognition proteins and checkpoint sensor Rad9-Rad1-Hus1. DNA Repair (Amst) 9: 478–487.

23. Cotta-RamusinoC, McDonaldER, HurovK, SowaME, HarperJW, et al. (2011) A DNA damage response screen identifies RHINO, a 9-1-1 and TopBP1 interacting protein required for ATR signaling. Science 332: 1313–1317.

24. PereraD, Perez-HidalgoL, MoensPB, ReiniK, LakinN, et al. (2004) TopBP1 and ATR colocalization at meiotic chromosomes: Role of TopBP1/Cut5 in the meiotic recombination checkpoint. Mol Biol Cell 15: 1568–1579.

25. MoensPB, TarsounasM, MoritaT, HabuT, RottinghausST, et al. (1999) The association of ATR protein with mouse meiotic chromosome cores. Chromosoma 108: 95–102.

26. TurnerJMA, AprelikovaO, XuX, WangR, KimS, et al. (2004) BRCA1, histone H2AX phosphorylation, and male meiotic sex chromosome inactivation. Curr Biol 14: 2135–2142.

27. HandelMA (2004) The XY body: a specialized meiotic chromatin domain. Exp Cell Res 296: 57–63.

28. Hoyer-FenderS (2004) Molecular aspects of XY body formation. Cytogenet Genome Res 103: 245–255.

29. BellaniMA, RomanienkoPJ, CairattiDA, Camerini-OteroRD (2005) SPO11 is required for sex-body formation, and Spo11 heterozygosity rescues the prophase arrest of Atm-/- spermatocytes. J Cell Sci 118: 3233–3245.

30. TurnerJMA (2007) Meiotic sex chromosome inactivation. Development 134: 1823–1831.

31. RoyoH, PolikiewiczG, MahadevaiahSK, ProsserH, MitchellM, et al. (2010) Evidence that meiotic sex chromosome inactivation is essential for male fertility. Curr Biol 20: 2117–2123.

32. TurnerJMA, MahadevaiahSK, Fernandez-CapetilloO, NussenzweigA, XuX, et al. (2005) Silencing of unsynapsed meiotic chromosomes in the mouse. Nat Genet 37: 41–47.

33. WojtaszL, CloutierJM, BaumannM, DanielK, VargaJ, et al. (2012) Meiotic DNA double-strand breaks and chromosome asynapsis in mice are monitored by distinct HORMAD2-independent and -dependent mechanisms. Genes Dev 26: 958–973.

34. LydallD, NikolskyY, BishopDK, WeinertT (1996) A meiotic recombination checkpoint controlled by mitotic checkpoint genes. Nature 383: 840–843.

35. HongE-JE, RoederGS (2002) A role for Ddc1 in signaling meiotic double-strand breaks at the pachytene checkpoint. Genes Dev 16: 363–376.

36. AbduU, KlovstadM, Butin-IsraeliV, BakhratA, SchüpbachT (2007) An essential role for Drosophila hus1 in somatic and meiotic DNA damage responses. J Cell Sci 120: 1042–1049.

37. ShinoharaM, SakaiK, OgawaT, ShinoharaA (2003) The mitotic DNA damage checkpoint proteins Rad17 and Rad24 are required for repair of double-strand breaks during meiosis in yeast. Genetics 164: 855–865.

38. GrushcowJM, HolzenTM, ParkKJ, WeinertT, LichtenM, et al. (1999) Saccharomyces cerevisiae checkpoint genes MEC1, RAD17 and RAD24 are required for normal meiotic recombination partner choice. Genetics 153: 607–620.

39. PeretzG, ArieLG, BakhratA, AbduU (2009) The Drosophila hus1 gene is required for homologous recombination repair during meiosis. Mech Dev 126: 677–686.

40. SmirnovaNA, RomanienkoPJ, KhilPP, Camerini-OteroRD (2006) Gene expression profiles of Spo11−/− mouse testes with spermatocytes arrested in meiotic prophase I. Reproduction 132: 67–77.

41. FreireR, MurguiaJR, TarsounasM, LowndesNF, MoensPB, et al. (1998) Human and mouse homologs of Schizosaccharomyces pombe rad1+ and Saccharomyces cerevisiae RAD17: linkage to checkpoint control and mammalian meiosis. Genes Dev 12: 2560–2573.

42. BrownEJ, BaltimoreD (2000) ATR disruption leads to chromosomal fragmentation and early embryonic lethality. Genes Dev 14: 397–402.

43. HopkinsKM, AuerbachW, WangXY, HandeMP, HangH, et al. (2004) Deletion of mouse Rad9 causes abnormal cellular responses to DNA damage, genomic instability, and embryonic lethality. Mol Cell Biol 24: 7235–7248.

44. WeissRS, EnochT, LederP (2000) Inactivation of mouse Hus1 results in genomic instability and impaired responses to genotoxic stress. Genes Dev 14: 1886–1898.

45. HanL, HuZ, LiuY, WangX, HopkinsK, et al. (2010) Mouse Rad1 deletion enhances susceptibility for skin tumor development. Mol Cancer 9: 67.

46. LevittPS, LiuH, ManningC, WeissRS (2005) Conditional inactivation of the mouse Hus1 cell cycle checkpoint gene. Genomics 86: 212–224.

47. Sadate-NgatchouPI, PayneCJ, DearthAT, Robert EBraun (2008) Cre recombinase activity specific to postnatal, premeiotic male germ cells in transgenic mice. Genesis 46: 738–742.

48. WangX, GuanJ, HuB, WeissRS, IliakisG, et al. (2004) Involvement of Hus1 in the chain elongation step of DNA replication after exposure to camptothecin or ionizing radiation. Nucleic Acids Res 32: 767–775.

49. LevittPS, ZhuM, CassanoA, YazinskiSA, LiuH, et al. (2007) Genome maintenance defects in cultured cells and mice following partial inactivation of the essential cell cycle checkpoint gene Hus1. Mol Cell Biol 27: 2189–2201.

50. ZhuM, WeissRS (2007) Increased common fragile site expression, cell proliferation defects, and apoptosis following conditional inactivation of mouse Hus1 in primary cultured cells. Mol Biol Cell 18: 1044–1055.

51. WardIM, ChenJ (2001) Histone H2AX is phosphorylated in an ATR-dependent manner in response to replicational stress. J Biol Chem 276: 47759–47762.

52. ZhaoH, Piwnica-WormsH (2001) ATR-mediated checkpoint pathways regulate phosphorylation and activation of human Chk1. Molecular and Cellular Biology 21: 4129–4139.

53. ChicheporticheA, Bernardino-SgherriJ, de MassyB, DutrillauxB (2007) Characterization of Spo11-dependent and independent phospho-H2AX foci during meiotic prophase I in the male mouse. J Cell Sci 120: 1733–1742.

54. LiXC, SchimentiJC (2007) Mouse pachytene checkpoint 2 (Trip13) is required for completing meiotic recombination but not synapsis. PLoS Genet 3: e130 doi:10.1371/journal.pgen.0030130.

55. HollowayJK, MohanS, BalmusG, SunX, ModzelewskiA, et al. (2011) Mammalian BTBD12 (SLX4) protects against genomic instability during mammalian spermatogenesis. PLoS Genet 7: e1002094 doi:10.1371/journal.pgen.1002094.

56. HollowayJK, MorelliMA, BorstPL, CohenPE (2010) Mammalian BLM helicase is critical for integrating multiple pathways of meiotic recombination. J Cell Biol 188: 779–789.

57. HollowayJK, BoothJ, EdelmannW, McGowanCH, CohenPE (2008) MUS81 generates a subset of MLH1-MLH3–independent crossovers in mammalian meiosis. PLoS Genet 4: e1000186 doi:10.1371/journal.pgen.1000186.

58. KolasNK, SvetlanovA, LenziML, MacalusoFP, LipkinSM, et al. (2005) Localization of MMR proteins on meiotic chromosomes in mice indicates distinct functions during prophase I. J Cell Biol 171: 447–458.

59. BaudatF, de MassyB (2007) Regulating double-stranded DNA break repair towards crossover or non-crossover during mammalian meiosis. Chromosome Res 15: 565–577.

60. ReiniK, UittoL, PereraD, MoensP, FreireR, et al. (2004) TopBP1 localises to centrosomes in mitosis and to chromosome cores in meiosis. Chromosoma 112: 323–330.

61. Lindsey-BoltzLA, SancarA (2011) Tethering DNA damage checkpoint mediator proteins Topoisomerase IIβ-binding Protein 1 (TopBP1) and Claspin to DNA activates Ataxia-Telangiectasia Mutated and RAD3-related (ATR) phosphorylation of Checkpoint Kinase 1 (Chk1). J Biol Chem 286: 19229–19236.

62. NamEA, CortezD (2011) ATR signalling: more than meeting at the fork. Biochem J 436: 527–536.

63. DelacroixS, WagnerJM, KobayashiM, YamamotoK-i, KarnitzLM (2007) The Rad9-Hus1-Rad1 (9-1-1) clamp activates checkpoint signaling via TopBP1. Genes Dev 21: 1472–1477.

64. RomanienkoPJ, Camerini-OteroRD (2000) The mouse Spo11 gene is required for meiotic chromosome synapsis. Molecular Cell 6: 975–987.

65. PittmanDL, CobbJ, SchimentiKJ, WilsonLA, CooperDM, et al. (1998) Meiotic prophase arrest with failure of chromosome synapsis in mice deficient for Dmc1, a germline-specific RecA homolog. Mol Cell 1: 697–705.

66. KneitzB, CohenPE, AvdievichE, ZhuL, KaneMF, et al. (2000) MutS homolog 4 localization to meiotic chromosomes is required for chromosome pairing during meiosis in male and female mice. Genes & Development 14: 1085–1097.

67. HangH, ZhangY, DunbrackRLJr, WangC, LiebermanHB (2002) Identification and characterization of a paralog of human cell cycle checkpoint gene HUS1. Genomics 79: 487–492.

68. DufaultVM, OestreichAJ, VromanBT, KarnitzLM (2003) Identification and characterization of RAD9B, a paralog of the RAD9 checkpoint gene. Genomics 82: 644–651.

69. BurtelowMA, Roos-MattjusPMK, RauenM, BabendureJR, KarnitzLM (2001) Reconstitution and molecular analysis of the hRad9-hHus1-hRad1 (9-1-1) DNA damage responsive checkpoint complex. Journal of Biological Chemistry 276: 25903–25909.

70. DanielK, LangeJ, HachedK, FuJ, AnastassiadisK, et al. (2011) Meiotic homologue alignment and its quality surveillance are controlled by mouse HORMAD1. Nat Cell Biol 13: 599–610.

71. OhSD, LaoJP, HwangPY-H, TaylorAF, SmithGR, et al. (2007) BLM ortholog, Sgs1, prevents aberrant crossing-over by suppressing formation of multichromatid joint molecules. Cell 130: 259–272.

72. De MuytA, JessopL, KolarE, SourirajanA, ChenJ, et al. (2012) BLM helicase ortholog Sgs1 is a central regulator of meiotic recombination intermediate metabolism. Molecular Cell 46: 43–53.

73. RoigI, DowdleJA, TothA, de RooijDG, JasinM, et al. (2010) Mouse TRIP13/PCH2 is required for recombination and normal higher-order chromosome structure during meiosis. PLoS Genet 6: e1001062 doi:10.1371/journal.pgen.1001062

74. BrandtPD, HeltCE, KengPC, BambaraRA (2006) The Rad9 protein enhances survival and promotes DNA repair following exposure to ionizing radiation. Biochem Biophys Res Comm 347: 232–237.

75. WangX, HuB, WeissRS, WangY (2006) The effect of Hus1 on ionizing radiation sensitivity is associated with homologous recombination repair but is independent of nonhomologous end-joining. Oncogene 25: 1980–1983.

76. KidaneD, JonasonAS, GortonTS, MihaylovI, PanJ, et al. (2010) DNA polymerase [beta] is critical for mouse meiotic synapsis. EMBO J 29: 410–423.

77. PlugAW, ClairmontCA, SapiE, AshleyT, SweasyJB (1997) Evidence for a role for DNA polymerase β in mammalian meiosis. Proc Natl Acad Sci USA 94: 1327–1331.

78. KawamotoT, ArakiK, SonodaE, YamashitaYM, HaradaK, et al. (2005) Dual roles for DNA polymerase η in homologous DNA recombination and translesion DNA synthesis. Mol Cell 20: 793–799.

79. McIlwraithMJ, VaismanA, LiuY, FanningE, WoodgateR, et al. (2005) Human DNA polymerase η promotes DNA synthesis from strand invasion intermediates of homologous recombination. Mol Cell 20: 783–792.

80. SharmaS, HicksJK, ChuteCL, BrennanJR, AhnJ-Y, et al. (2012) REV1 and polymerase ζ facilitate homologous recombination repair. Nucleic Acids Res 40: 682–691.

81. GoedeckeW, EijpeM, OffenbergHH, AalderenMv, HeytingC (1999) Mre11 and Ku70 interact in somatic cells, but are differentially expressed in early meiosis. Nat Genet 23: 194–198.

82. EndersGC, MayJJ (1994) Developmentally regulated expression of a mouse germ cell nuclear antigen examined from embryonic day 11 to adult in male and female mice. Dev Biol 163: 331–340.

83. LenziML, SmithJ, SnowdenT, KimM, FishelR, et al. (2005) Extreme heterogeneity in the molecular events leading to the establishment of chiasmata during meiosis I in human oocytes. Am J Hum Genet 76: 112–127.

84. DanielsenJMR, LarsenDH, SchouKB, FreireR, FalckJ, et al. (2009) HCLK2 is required for activity of the DNA damage response kinase ATR. J Biol Chem 284: 4140–4147.

85. LipkinSM, MoensPB, WangV, LenziM, ShanmugarajahD, et al. (2002) Meiotic arrest and aneuploidy in MLH3-deficient mice. Nat Genet 31: 385–390.

86. KanR, SunX, KolasNK, AvdievichE, KneitzB, et al. (2008) Comparative analysis of meiotic progression in female mice bearing mutations in genes of the DNA mismatch repair pathway. Biology of Reproduction 78: 462–471.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 2
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#