#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Mouse BAZ1A (ACF1) Is Dispensable for Double-Strand Break Repair but Is Essential for Averting Improper Gene Expression during Spermatogenesis


ATP-dependent chromatin remodelers control DNA access for transcription, recombination, and other processes. Acf1 (also known as BAZ1A in mammals) is a defining subunit of the conserved ISWI-family chromatin remodelers ACF and CHRAC, first purified over 15 years ago from Drosophila melanogaster embryos. Much is known about biochemical properties of ACF and CHRAC, which move nucleosomes in vitro and in vivo to establish ordered chromatin arrays. Genetic studies in yeast, flies and cultured human cells clearly implicate these complexes in transcriptional repression via control of chromatin structures. RNAi experiments in transformed mammalian cells in culture also implicate ACF and CHRAC in DNA damage checkpoints and double-strand break repair. However, their essential in vivo roles in mammals are unknown. Here, we show that Baz1a-knockout mice are viable and able to repair developmentally programmed DNA double-strand breaks in the immune system and germ line, I-SceI endonuclease-induced breaks in primary fibroblasts via homologous recombination, and DNA damage from mitomycin C exposure in vivo. However, Baz1a deficiency causes male-specific sterility in accord with its high expression in male germ cells, where it displays dynamic, stage-specific patterns of chromosomal localization. Sterility is caused by pronounced defects in sperm development, most likely a consequence of massively perturbed gene expression in spermatocytes and round spermatids in the absence of BAZ1A: the normal spermiogenic transcription program is largely intact but more than 900 other genes are mis-regulated, primarily reflecting inappropriate up-regulation. We propose that large-scale changes in chromatin composition that occur during spermatogenesis create a window of vulnerability to promiscuous transcription changes, with an essential function of ACF and/or CHRAC chromatin remodeling activities being to safeguard against these alterations.


Vyšlo v časopise: Mouse BAZ1A (ACF1) Is Dispensable for Double-Strand Break Repair but Is Essential for Averting Improper Gene Expression during Spermatogenesis. PLoS Genet 9(11): e32767. doi:10.1371/journal.pgen.1003945
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003945

Souhrn

ATP-dependent chromatin remodelers control DNA access for transcription, recombination, and other processes. Acf1 (also known as BAZ1A in mammals) is a defining subunit of the conserved ISWI-family chromatin remodelers ACF and CHRAC, first purified over 15 years ago from Drosophila melanogaster embryos. Much is known about biochemical properties of ACF and CHRAC, which move nucleosomes in vitro and in vivo to establish ordered chromatin arrays. Genetic studies in yeast, flies and cultured human cells clearly implicate these complexes in transcriptional repression via control of chromatin structures. RNAi experiments in transformed mammalian cells in culture also implicate ACF and CHRAC in DNA damage checkpoints and double-strand break repair. However, their essential in vivo roles in mammals are unknown. Here, we show that Baz1a-knockout mice are viable and able to repair developmentally programmed DNA double-strand breaks in the immune system and germ line, I-SceI endonuclease-induced breaks in primary fibroblasts via homologous recombination, and DNA damage from mitomycin C exposure in vivo. However, Baz1a deficiency causes male-specific sterility in accord with its high expression in male germ cells, where it displays dynamic, stage-specific patterns of chromosomal localization. Sterility is caused by pronounced defects in sperm development, most likely a consequence of massively perturbed gene expression in spermatocytes and round spermatids in the absence of BAZ1A: the normal spermiogenic transcription program is largely intact but more than 900 other genes are mis-regulated, primarily reflecting inappropriate up-regulation. We propose that large-scale changes in chromatin composition that occur during spermatogenesis create a window of vulnerability to promiscuous transcription changes, with an essential function of ACF and/or CHRAC chromatin remodeling activities being to safeguard against these alterations.


Zdroje

1. FlausA, Owen-HughesT (2011) Mechanisms for ATP-dependent chromatin remodelling: the means to the end. The FEBS Journal 278: 3579–3595.

2. ClapierCR, CairnsBR (2009) The biology of chromatin remodeling complexes. Annu Rev Biochem 78: 273–304.

3. ItoT, BulgerM, PazinMJ, KobayashiR, KadonagaJT (1997) ACF, an ISWI-containing and ATP-utilizing chromatin assembly and remodeling factor. Cell 90: 145–155.

4. Varga-WeiszPD, WilmM, BonteE, DumasK, MannM, et al. (1997) Chromatin-remodelling factor CHRAC contains the ATPases ISWI and topoisomerase II. Nature 388: 598–602.

5. ItoT, LevensteinME, FyodorovDV, KutachAK, KobayashiR, et al. (1999) ACF consists of two subunits, Acf1 and ISWI, that function cooperatively in the ATP-dependent catalysis of chromatin assembly. Genes Dev 13: 1529–1539.

6. BocharDA, SavardJ, WangW, LafleurDW, MooreP, et al. (2000) A family of chromatin remodeling factors related to Williams syndrome transcription factor. Proc Natl Acad Sci USA 97: 1038–1043.

7. LeRoyG, LoyolaA, LaneWS, ReinbergD (2000) Purification and characterization of a human factor that assembles and remodels chromatin. J Biol Chem 275: 14787–14790.

8. EberharterA, FerrariS, LängstG, StraubT, ImhofA, et al. (2001) Acf1, the largest subunit of CHRAC, regulates ISWI-induced nucleosome remodelling. EMBO J 20: 3781–3788.

9. CoronaDF, EberharterA, BuddeA, DeuringR, FerrariS, et al. (2000) Two histone fold proteins, CHRAC-14 and CHRAC-16, are developmentally regulated subunits of chromatin accessibility complex (CHRAC). EMBO J 19: 3049–3059.

10. PootRA, DellaireG, HülsmannBB, GrimaldiMA, CoronaDF, et al. (2000) HuCHRAC, a human ISWI chromatin remodelling complex contains hACF1 and two novel histone-fold proteins. EMBO J 19: 3377–3387.

11. JonesMH, HamanaN, NezuJ-i, ShimaneM (2000) A Novel Family of Bromodomain Genes. Genomics 63: 40–45.

12. NarlikarGJ (2010) A proposal for kinetic proof reading by ISWI family chromatin remodeling motors. Curr Opin Chem Biol 14: 660–665.

13. HotaSK, BartholomewB (2011) Diversity of operation in ATP-dependent chromatin remodelers. Biochim Biophys Acta 1809: 476–487.

14. HopfnerKP, GerholdCB, LakomekK, WollmannP (2012) Swi2/Snf2 remodelers: hybrid views on hybrid molecular machines. Curr Opin Struct Biol 22: 225–233.

15. HeX, FanHY, GarlickJD, KingstonRE (2008) Diverse regulation of SNF2h chromatin remodeling by noncatalytic subunits. Biochemistry 47: 7025–7033.

16. HeX, FanHY, NarlikarGJ, KingstonRE (2006) Human ACF1 alters the remodeling strategy of SNF2h. J Biol Chem 281: 28636–28647.

17. BouazouneK, MirandaTB, JonesPA, KingstonRE (2009) Analysis of individual remodeled nucleosomes reveals decreased histone-DNA contacts created by hSWI/SNF. Nucleic Acids Res 37: 5279–5294.

18. KagalwalaMN, GlausBJ, DangW, ZofallM, BartholomewB (2004) Topography of the ISW2-nucleosome complex: insights into nucleosome spacing and chromatin remodeling. EMBO J 23: 2092–2104.

19. KukimotoI, ElderkinS, GrimaldiM, OelgeschlägerT, Varga-WeiszPD (2004) The histone-fold protein complex CHRAC-15/17 enhances nucleosome sliding and assembly mediated by ACF. Molecular Cell 13: 265–277.

20. HartleppKF, Fernández-TorneroC, EberharterA, GrüneT, MüllerCW, et al. (2005) The histone fold subunits of Drosophila CHRAC facilitate nucleosome sliding through dynamic DNA interactions. Mol Cell Biol 25: 9886–9896.

21. LangstG, BonteEJ, CoronaDF, BeckerPB (1999) Nucleosome movement by CHRAC and ISWI without disruption or trans-displacement of the histone octamer. Cell 97: 843–852.

22. ErdelF, RippeK (2011) Chromatin remodeling in mammalian cells by ISWI type complexes - where, when and why? FEBS J 278: 3608–3618.

23. StopkaT, SkoultchiAI (2003) The ISWI ATPase Snf2h is required for early mouse development. Proc Natl Acad Sci USA 100: 14097–14102.

24. DeuringR, FantiL, ArmstrongJA, SarteM, PapoulasO, et al. (2000) The ISWI chromatin-remodeling protein is required for gene expression and the maintenance of higher order chromatin structure in vivo. Molecular Cell 5: 355–365.

25. FyodorovDV, BlowerMD, KarpenGH, KadonagaJT (2004) Acf1 confers unique activities to ACF/CHRAC and promotes the formation rather than disruption of chromatin in vivo. Genes Dev 18: 170–183.

26. ChiodaM, VengadasalamS, KremmerE, EberharterA, BeckerPB (2010) Developmental role for ACF1-containing nucleosome remodellers in chromatin organisation. Development 137: 3513–3522.

27. LiuYI, ChangMV, LiHE, BaroloS, ChangJL, et al. (2008) The chromatin remodelers ISWI and ACF1 directly repress Wingless transcriptional targets. Dev Biol 323: 41–52.

28. TsukiyamaT, PalmerJ, LandelCC, ShiloachJ, WuC (1999) Characterization of the imitation switch subfamily of ATP-dependent chromatin-remodeling factors in Saccharomyces cerevisiae. Genes Dev 13: 686–697.

29. CollinsN, PootRA, KukimotoI, García-JiménezC, DellaireG, et al. (2002) An ACF1-ISWI chromatin-remodeling complex is required for DNA replication through heterochromatin. Nat Genet 32: 627–632.

30. EwingAK, AttnerM, ChakravartiD (2007) Novel regulatory role for human Acf1 in transcriptional repression of vitamin D3 receptor-regulated genes. Mol Endocrinol 21: 1791–1806.

31. LanL, UiA, NakajimaS, HatakeyamaK, HoshiM, et al. (2010) The ACF1 Complex Is Required for DNA Double-Strand Break Repair in Human Cells. Molecular Cell 40: 976–987.

32. Sanchez-MolinaS, MortusewiczO, BieberB, AuerS, EckeyM, et al. (2011) Role for hACF1 in the G2/M damage checkpoint. Nucleic Acids Research 39: 8445–8456.

33. GovinJ, CaronC, LestratC, RousseauxS, KhochbinS (2004) The role of histones in chromatin remodelling during mammalian spermiogenesis. Eur J Biochem 271: 3459–3469.

34. KimminsS, Sassone-CorsiP (2005) Chromatin remodelling and epigenetic features of germ cells. Nature 434: 583–589.

35. GaucherJ, BoussouarF, MontellierE, CurtetS, BuchouT, et al. (2012) Bromodomain-dependent stage-specific male genome programming by Brdt. EMBO J 31: 3809–3820.

36. MacleanJA2nd (2005) Gene regulation in spermatogenesis. Curr Top Dev Biol 71: 131–197.

37. Bolcun-FilasE, BannisterLA, BarashA, SchimentiKJ, HartfordSA, et al. (2011) A-MYB (MYBL1) transcription factor is a master regulator of male meiosis. Development 138: 3319–3330.

38. TateP, LeeM, TweedieS, SkarnesWC, BickmoreWA (1998) Capturing novel mouse genes encoding chromosomal and other nuclear proteins. J Cell Sci 111: 2575–2585.

39. ThompsonPJ, NortonKA, NiriFH, DaweCE, McDermidHE (2012) CECR2 is involved in spermatogenesis and forms a complex with SNF2H in the testis. J Mol Biol 415: 793–806.

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

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

42. ErdelF, RippeK (2011) Binding kinetics of human ISWI chromatin-remodelers to DNA repair sites elucidate their target location mechanism. Nucleus 2: 105–112.

43. KeeneyS (2008) Spo11 and the Formation of DNA Double-Strand Breaks in Meiosis. Genome Dyn Stab 2: 81–123.

44. BarchiM, MahadevaiahS, Di GiacomoM, BaudatF, de RooijDG, et al. (2005) Surveillance of different recombination defects in mouse spermatocytes yields distinct responses despite elimination at an identical developmental stage. Mol Cell Biol 25: 7203–7215.

45. 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.

46. Di GiacomoM, BarchiM, BaudatF, EdelmannW, KeeneyS, et al. (2005) Distinct DNA damage-dependent and independent responses drive the loss of oocytes in recombination-defective mouse mutants. Proc Natl Acad Sci USA 102: 737–742.

47. LiXC, SchimentiJC (2007) Mouse pachytene checkpoint 2 (trip13) is required for completing meiotic recombination but not synapsis. PLoS Genet 3: e130.

48. BassingCH, SwatW, AltFW (2002) The mechanism and regulation of chromosomal V(D)J recombination. Cell 109 Suppl: S45–55.

49. ManisJP, TianM, AltFW (2002) Mechanism and control of class-switch recombination. Trends Immunol 23: 31–39.

50. LiL, SalidoE, ZhouY, BhattacharyyaS, YannoneSM, et al. (2005) Targeted disruption of the Artemis murine counterpart results in SCID and defective V(D)J recombination that is partially corrected with bone marrow transplantation. J Immunol 174: 2420–2428.

51. KassEM, HelgadottirHR, ChenCC, BarberaM, WangR, et al. (2013) Double-strand break repair by homologous recombination in primary mouse somatic cells requires BRCA1 but not the ATM kinase. Proc Natl Acad Sci U S A 110: 5564–5569.

52. OlivaR, Bazett-JonesD, MezquitaC, DixonGH (1987) Factors affecting nucleosome disassembly by protamines in vitro. Histone hyperacetylation and chromatin structure, time dependence, and the size of the sperm nuclear proteins. J Biol Chem 262: 17016–17025.

53. ZhaoM, ShirleyCR, YuYE, MohapatraB, ZhangY, et al. (2001) Targeted disruption of the transition protein 2 gene affects sperm chromatin structure and reduces fertility in mice. Mol Cell Biol 21: 7243–7255.

54. LeeK, HaugenHS, CleggCH, BraunRE (1995) Premature translation of protamine 1 mRNA causes precocious nuclear condensation and arrests spermatid differentiation in mice. Proc Natl Acad Sci USA 92: 12451–12455.

55. ChoC, WillisWD, GouldingEH, Jung-HaH, ChoiYC, et al. (2001) Haploinsufficiency of protamine-1 or -2 causes infertility in mice. Nat Genet 28: 82–86.

56. EskelandR, EberharterA, ImhofA (2006) HP1 Binding to Chromatin Methylated at H3K9 Is Enhanced by Auxiliary Factors. Molecular and Cellular Biology 27: 453–465.

57. NamekawaSH, ParkPJ, ZhangLF, ShimaJE, McCarreyJR, et al. (2006) Postmeiotic sex chromatin in the male germline of mice. Curr Biol 16: 660–667.

58. ChalmelF, RollandAD, Niederhauser-WiederkehrC, ChungSS, DemouginP, et al. (2007) The conserved transcriptome in human and rodent male gametogenesis. Proc Natl Acad Sci U S A 104: 8346–8351.

59. MuellerJL, MahadevaiahSK, ParkPJ, WarburtonPE, PageDC, et al. (2008) The mouse X chromosome is enriched for multicopy testis genes showing postmeiotic expression. Nat Genet 40: 794–799.

60. BaoY, ShenX (2007) Chromatin remodeling in DNA double-strand break repair. Curr Opin Genet Dev 17: 126–131.

61. XiaoA, LiH, ShechterD, AhnSH, FabrizioLA, et al. (2009) WSTF regulates the H2A.X DNA damage response via a novel tyrosine kinase activity. Nature 457: 57–62.

62. KosirR, JuvanP, PerseM, BudefeldT, MajdicG, et al. (2012) Novel insights into the downstream pathways and targets controlled by transcription factors CREM in the testis. PLoS One 7: e31798.

63. LiXZ, RoyCK, DongX, Bolcun-FilasE, WangJ, et al. (2013) An ancient transcription factor initiates the burst of piRNA production during early meiosis in mouse testes. Mol Cell 50: 67–81.

64. ShangE, NickersonHD, WenD, WangX, WolgemuthDJ (2007) The first bromodomain of Brdt, a testis-specific member of the BET sub-family of double-bromodomain-containing proteins, is essential for male germ cell differentiation. Development 134: 3507–3515.

65. MatzukMM, McKeownMR, FilippakopoulosP, LiQ, MaL, et al. (2012) Small-molecule inhibition of BRDT for male contraception. Cell 150: 673–684.

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

67. SinHS, BarskiA, ZhangF, KartashovAV, NussenzweigA, et al. (2012) RNF8 regulates active epigenetic modifications and escape gene activation from inactive sex chromosomes in post-meiotic spermatids. Genes Dev 26: 2737–2748.

68. WangJ, GuH, LinH, ChiT (2012) Essential roles of the chromatin remodeling factor BRG1 in spermatogenesis in mice. Biol Reprod 86: 186.

69. KimY, FedoriwAM, MagnusonT (2012) An essential role for a mammalian SWI/SNF chromatin-remodeling complex during male meiosis. Development 139: 1133–1140.

70. BantingGS, BarakO, AmesTM, BurnhamAC, KardelMD, et al. (2005) CECR2, a protein involved in neurulation, forms a novel chromatin remodeling complex with SNF2L. Hum Mol Genet 14: 513–524.

71. GoldmarkJP, FazzioTG, EstepPW, ChurchGM, TsukiyamaT (2000) The Isw2 chromatin remodeling complex represses early meiotic genes upon recruitment by Ume6p. Cell 103: 423–433.

72. FazzioTG, KooperbergC, GoldmarkJP, NealC, BasomR, et al. (2001) Widespread collaboration of Isw2 and Sin3-Rpd3 chromatin remodeling complexes in transcriptional repression. Mol Cell Biol 21: 6450–6460.

73. CoronaDF, TamkunJW (2004) Multiple roles for ISWI in transcription, chromosome organization and DNA replication. Biochim Biophys Acta 1677: 113–119.

74. BorgCL, WolskiKM, GibbsGM, O'BryanMK (2010) Phenotyping male infertility in the mouse: how to get the most out of a ‘non-performer’. Hum Reprod Update 16: 205–224.

75. BerkovitsBD, WolgemuthDJ (2011) The first bromodomain of the testis-specific double bromodomain protein Brdt is required for chromocenter organization that is modulated by genetic background. Dev Biol 360: 358–368.

76. BrownJP, BullwinkelJ, Baron-LuhrB, BillurM, SchneiderP, et al. (2010) HP1gamma function is required for male germ cell survival and spermatogenesis. Epigenetics Chromatin 3: 9.

77. PetersAH, O'CarrollD, ScherthanH, MechtlerK, SauerS, et al. (2001) Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell 107: 323–337.

78. WuS, HuYC, LiuH, ShiY (2009) Loss of YY1 impacts the heterochromatic state and meiotic double-strand breaks during mouse spermatogenesis. Mol Cell Biol 29: 6245–6256.

79. YoshimuraK, KitagawaH, FujikiR, TanabeM, TakezawaS, et al. (2009) Distinct function of 2 chromatin remodeling complexes that share a common subunit, Williams syndrome transcription factor (WSTF). Proc Natl Acad Sci U S A 106: 9280–9285.

80. SteilmannC, CavalcantiMCO, BergmannM, KlieschS, WeidnerW, et al. (2010) Aberrant mRNA expression of chromatin remodelling factors in round spermatid maturation arrest compared with normal human spermatogenesis. Mol Hum Reprod 16: 726–733.

81. LiuP, JenkinsNA, CopelandNG (2003) A highly efficient recombineering-based method for generating conditional knockout mutations. Genome Res 13: 476–484.

82. LeeEC, YuD, Martinez de VelascoJ, TessarolloL, SwingDA, et al. (2001) A highly efficient Escherichia coli-based chromosome engineering system adapted for recombinogenic targeting and subcloning of BAC DNA. Genomics 73: 56–65.

83. BastosH, LassalleB, ChicheporticheA, RiouL, TestartJ, et al. (2005) Flow cytometric characterization of viable meiotic and postmeiotic cells by Hoechst 33342 in mouse spermatogenesis. Cytometry A 65: 40–49.

84. Smyth GK (2005) Limma: linear models for microarray data. In: R. Gentleman VC, S Dudoit, R Irizarry, W Huber, editor. Bioinformatics and Computational Biology Solutions using R and Bioconductor. New York: Springer. pp. 397–420.

85. RitchieME, DunningMJ, SmithML, ShiW, LynchAG (2011) BeadArray expression analysis using bioconductor. PLoS Comput Biol 7: e1002276.

86. DunningM, ShiW, LynchA, SmithM, RitchieM (2012) Analysing Illumina BeadArray expression data using Bioconductor. BeadArrayUseCases

87. Barbosa-MoraisNL, DunningMJ, SamarajiwaSA, DarotJF, RitchieME, et al. (2010) A re-annotation pipeline for Illumina BeadArrays: improving the interpretation of gene expression data. Nucleic Acids Res 38: e17.

88. DuncanDT, ProdduturiN, ZhangB (2010) WebGestalt2: an updated and expanded version of the Web-based Gene Set Analysis Toolkit. BMC Bioinformatics 11: 10.

89. YabutaY, OhtaH, AbeT, KurimotoK, ChumaS, et al. (2011) TDRD5 is required for retrotransposon silencing, chromatoid body assembly, and spermiogenesis in mice. J Cell Biol 192: 781–795.

90. GirardA, SachidanandamR, HannonGJ, CarmellMA (2006) A germline-specific class of small RNAs binds mammalian Piwi proteins. Nature 442: 199–202.

91. PetersAH, PlugAW, van VugtMJ, de BoerP (1997) A drying-down technique for the spreading of mammalian meiocytes from the male and female germline. Chromosome Res 5: 66–68.

92. PageJ, SujaJA, SantosJL, RufasJS (1998) Squash procedure for protein immunolocalization in meiotic cells. Chromosome Res 6: 639–642.

93. GavrieliY, ShermanY, Ben-SassonSA (1992) Identification of programmed cell death in situ via specific labeling of nuclear DNA fragmentation. J Cell Biol 119: 493–501.

94. MendezJ, StillmanB (2000) Chromatin association of human origin recognition complex, cdc6, and minichromosome maintenance proteins during the cell cycle: assembly of prereplication complexes in late mitosis. Mol Cell Biol 20: 8602–8612.

95. YadonAN, TsukiyamaT (2011) SnapShot: Chromatin remodeling: ISWI. Cell 144: 453–453 e451.

96. AhmedEA, de RooijDG (2009) Staging of mouse seminiferous tubule cross-sections. Methods Mol Biol 558: 263–277.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 11
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#