#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Yemanuclein and HIRA Cooperate for Assembly of H3.3-Containing Nucleosomes in the Male Pronucleus


The differentiation of post-meiotic spermatids in animals is characterized by a unique reorganization of their nuclear architecture and chromatin composition. In many species, the formation of sperm nuclei involves the massive replacement of nucleosomes with protamines, followed by a phase of extreme nuclear compaction. At fertilization, the reconstitution of a nucleosome-based paternal chromatin after the removal of protamines requires the deposition of maternally provided histones before the first round of DNA replication. This process exclusively uses the histone H3 variant H3.3 and constitutes a unique case of genome-wide replication-independent (RI) de novo chromatin assembly. We had previously shown that the histone H3.3 chaperone HIRA plays a central role for paternal chromatin assembly in Drosophila. Although several conserved HIRA-interacting proteins have been identified from yeast to human, their conservation in Drosophila, as well as their actual implication in this highly peculiar RI nucleosome assembly process, is an open question. Here, we show that Yemanuclein (YEM), the Drosophila member of the Hpc2/Ubinuclein family, is essential for histone deposition in the male pronucleus. yem loss of function alleles affect male pronucleus formation in a way remarkably similar to Hira mutants and abolish RI paternal chromatin assembly. In addition, we demonstrate that HIRA and YEM proteins interact and are mutually dependent for their targeting to the decondensing male pronucleus. Finally, we show that the alternative ATRX/XNP-dependent H3.3 deposition pathway is not involved in paternal chromatin assembly, thus underlining the specific implication of the HIRA/YEM complex for this essential step of zygote formation.


Vyšlo v časopise: Yemanuclein and HIRA Cooperate for Assembly of H3.3-Containing Nucleosomes in the Male Pronucleus. PLoS Genet 9(2): e32767. doi:10.1371/journal.pgen.1003285
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003285

Souhrn

The differentiation of post-meiotic spermatids in animals is characterized by a unique reorganization of their nuclear architecture and chromatin composition. In many species, the formation of sperm nuclei involves the massive replacement of nucleosomes with protamines, followed by a phase of extreme nuclear compaction. At fertilization, the reconstitution of a nucleosome-based paternal chromatin after the removal of protamines requires the deposition of maternally provided histones before the first round of DNA replication. This process exclusively uses the histone H3 variant H3.3 and constitutes a unique case of genome-wide replication-independent (RI) de novo chromatin assembly. We had previously shown that the histone H3.3 chaperone HIRA plays a central role for paternal chromatin assembly in Drosophila. Although several conserved HIRA-interacting proteins have been identified from yeast to human, their conservation in Drosophila, as well as their actual implication in this highly peculiar RI nucleosome assembly process, is an open question. Here, we show that Yemanuclein (YEM), the Drosophila member of the Hpc2/Ubinuclein family, is essential for histone deposition in the male pronucleus. yem loss of function alleles affect male pronucleus formation in a way remarkably similar to Hira mutants and abolish RI paternal chromatin assembly. In addition, we demonstrate that HIRA and YEM proteins interact and are mutually dependent for their targeting to the decondensing male pronucleus. Finally, we show that the alternative ATRX/XNP-dependent H3.3 deposition pathway is not involved in paternal chromatin assembly, thus underlining the specific implication of the HIRA/YEM complex for this essential step of zygote formation.


Zdroje

1. PoloSE, AlmouzniG (2006) Chromatin assembly: a basic recipe with various flavours. Curr Opin Genet Dev 16: 104–111.

2. AhmadK, HenikoffS (2002) Histone H3 variants specify modes of chromatin assembly. Proc Natl Acad Sci U S A 99 Suppl 4: 16477–16484.

3. TalbertPB, AhmadK, AlmouzniG, AusioJ, BergerF, et al. (2012) A unified phylogeny-based nomenclature for histone variants. Epigenetics Chromatin 5: 7.

4. CorpetA, AlmouzniG (2009) Making copies of chromatin: the challenge of nucleosomal organization and epigenetic information. Trends Cell Biol 19: 29–41.

5. ElsaesserSJ, GoldbergAD, AllisCD (2010) New functions for an old variant: no substitute for histone H3.3. Curr Opin Genet Dev 20: 110–117.

6. GoldbergAD, BanaszynskiLA, NohKM, LewisPW, ElsaesserSJ, et al. (2010) Distinct factors control histone variant H3.3 localization at specific genomic regions. Cell 140: 678–691.

7. SzenkerE, Ray-GalletD, AlmouzniG (2011) The double face of the histone variant H3.3. Cell Res 21: 421–434.

8. TagamiH, Ray-GalletD, AlmouzniG, NakataniY (2004) Histone H3.1 and H3.3 complexes mediate nucleosome assembly pathways dependent or independent of DNA synthesis. Cell 116: 51–61.

9. AhmadK, HenikoffS (2002) The histone variant H3.3 marks active chromatin by replication-independent nucleosome assembly. Mol Cell 9: 1191–1200.

10. MitoY, HenikoffJG, HenikoffS (2005) Genome-scale profiling of histone H3.3 replacement patterns. Nat Genet 37: 1090–1097.

11. SchwartzBE, AhmadK (2005) Transcriptional activation triggers deposition and removal of the histone variant H3.3. Genes Dev 19: 804–814.

12. WirbelauerC, BellO, SchubelerD (2005) Variant histone H3.3 is deposited at sites of nucleosomal displacement throughout transcribed genes while active histone modifications show a promoter-proximal bias. Genes Dev 19: 1761–1766.

13. ChowCM, GeorgiouA, SzutoriszH, Maia e SilvaA, PomboA, et al. (2005) Variant histone H3.3 marks promoters of transcriptionally active genes during mammalian cell division. EMBO Rep 6: 354–360.

14. DealRB, HenikoffJG, HenikoffS (2010) Genome-wide kinetics of nucleosome turnover determined by metabolic labeling of histones. Science 328: 1161–1164.

15. MitoY, HenikoffJG, HenikoffS (2007) Histone replacement marks the boundaries of cis-regulatory domains. Science 315: 1408–1411.

16. SchneidermanJI, SakaiA, GoldsteinS, AhmadK (2009) The XNP remodeler targets dynamic chromatin in Drosophila. Proc Natl Acad Sci U S A 106: 14472–14477.

17. WongLH, RenH, WilliamsE, McGhieJ, AhnS, et al. (2009) Histone H3.3 incorporation provides a unique and functionally essential telomeric chromatin in embryonic stem cells. Genome Res 19: 404–414.

18. NakayamaT, NishiokaK, DongYX, ShimojimaT, HiroseS (2007) Drosophila GAGA factor directs histone H3.3 replacement that prevents the heterochromatin spreading. Genes & development 21: 552–561.

19. TalbertPB, HenikoffS (2010) Histone variants–ancient wrap artists of the epigenome. Nat Rev Mol Cell Biol 11: 264–275.

20. OoiSL, HenikoffS (2007) Germline histone dynamics and epigenetics. Curr Opin Cell Biol 19: 257–265.

21. OrsiGA, CoubleP, LoppinB (2009) Epigenetic and replacement roles of histone variant H3.3 in reproduction and development. Int J Dev Biol 53: 231–243.

22. BanaszynskiLA, AllisCD, LewisPW (2010) Histone variants in metazoan development. Dev Cell 19: 662–674.

23. van der HeijdenGW, DerijckAA, PosfaiE, GieleM, PelczarP, et al. (2007) Chromosome-wide nucleosome replacement and H3.3 incorporation during mammalian meiotic sex chromosome inactivation. Nat Genet 39: 251–258.

24. CouldreyC, CarltonMB, NolanPM, ColledgeWH, EvansMJ (1999) A retroviral gene trap insertion into the histone 3.3A gene causes partial neonatal lethality, stunted growth, neuromuscular deficits and male sub-fertility in transgenic mice. Hum Mol Genet 8: 2489–2495.

25. SantenardA, Ziegler-BirlingC, KochM, ToraL, BannisterAJ, et al. (2010) Heterochromatin formation in the mouse embryo requires critical residues of the histone variant H3.3. Nat Cell Biol 12: 853–862.

26. SzenkerE, LacosteN, AlmouzniG (2012) A Developmental Requirement for HIRA-Dependent H3.3 Deposition Revealed at Gastrulation in Xenopus. Cell Rep 1: 730–740.

27. HodlM, BaslerK (2009) Transcription in the absence of histone H3.3. Curr Biol 19: 1221–1226.

28. SakaiA, SchwartzBE, GoldsteinS, AhmadK (2009) Transcriptional and developmental functions of the H3.3 histone variant in Drosophila. Curr Biol 19: 1816–1820.

29. AkhmanovaA, MiedemaK, WangY, van BruggenM, BerdenJH, et al. (1997) The localization of histone H3.3 in germ line chromatin of Drosophila males as established with a histone H3.3-specific antiserum. Chromosoma 106: 335–347.

30. CarrellDT (2012) Epigenetics of the male gamete. Fertil Steril 97: 267–274.

31. Eirin-LopezJM, AusioJ (2009) Origin and evolution of chromosomal sperm proteins. Bioessays 31: 1062–1070.

32. MillerD, BrinkworthM, IlesD (2010) Paternal DNA packaging in spermatozoa: more than the sum of its parts? DNA, histones, protamines and epigenetics. Reproduction 139: 287–301.

33. AshburnerM, MisraS, RooteJ, LewisSE, BlazejR, et al. (1999) An exploration of the sequence of a 2.9-Mb region of the genome of Drosophila melanogaster: the Adh region. Genetics 153: 179–219.

34. Jayaramaiah RajaS, Renkawitz-PohlR (2005) Replacement by Drosophila melanogaster protamines and Mst77F of histones during chromatin condensation in late spermatids and role of sesame in the removal of these proteins from the male pronucleus. Mol Cell Biol 25: 6165–6177.

35. RathkeC, BaarendsWM, Jayaramaiah-RajaS, BartkuhnM, RenkawitzR, et al. (2007) Transition from a nucleosome-based to a protamine-based chromatin configuration during spermiogenesis in Drosophila. J Cell Sci 120: 1689–1700.

36. BonnefoyE, OrsiGA, CoubleP, LoppinB (2007) The essential role of Drosophila HIRA for de novo assembly of paternal chromatin at fertilization. PLoS Genet 3: e182 doi:10.1371/journal.pgen.0030182.

37. LoppinB, BonnefoyE, AnselmeC, LaurenconA, KarrTL, et al. (2005) The histone H3.3 chaperone HIRA is essential for chromatin assembly in the male pronucleus. Nature 437: 1386–1390.

38. van der HeijdenGW, DiekerJW, DerijckAA, MullerS, BerdenJH, et al. (2005) Asymmetry in histone H3 variants and lysine methylation between paternal and maternal chromatin of the early mouse zygote. Mech Dev 122: 1008–1022.

39. Torres-PadillaME, BannisterAJ, HurdPJ, KouzaridesT, Zernicka-GoetzM (2006) Dynamic distribution of the replacement histone variant H3.3 in the mouse oocyte and preimplantation embryos. Int J Dev Biol 50: 455–461.

40. ZhaoZK, LiW, WangMY, ZhouL, WangJL, et al. (2011) The role of HIRA and maternal histones in sperm nucleus decondensation in the gibel carp and color crucian carp. Mol Reprod Dev 78: 139–147.

41. GreenEM, AntczakAJ, BaileyAO, FrancoAA, WuKJ, et al. (2005) Replication-independent histone deposition by the HIR complex and Asf1. Curr Biol 15: 2044–2049.

42. ProchassonP, FlorensL, SwansonSK, WashburnMP, WorkmanJL (2005) The HIR corepressor complex binds to nucleosomes generating a distinct protein/DNA complex resistant to remodeling by SWI/SNF. Genes Dev 19: 2534–2539.

43. AminAD, VishnoiN, ProchassonP (2011) A global requirement for the HIR complex in the assembly of chromatin. Biochim Biophys Acta 1819: 264–276.

44. BalajiS, IyerLM, AravindL (2009) HPC2 and ubinuclein define a novel family of histone chaperones conserved throughout eukaryotes. Mol Biosyst 5: 269–275.

45. RaiTS, PuriA, McBryanT, HoffmanJ, TangY, et al. (2011) Human CABIN1 is a functional member of the human HIRA/UBN1/ASF1a histone H3.3 chaperone complex. Mol Cell Biol 31: 4107–4118.

46. AndersonHE, KaganskyA, WardleJ, RappsilberJ, AllshireRC, et al. (2010) Silencing mediated by the Schizosaccharomyces pombe HIRA complex is dependent upon the Hpc2-like protein, Hip4. PLoS ONE 5: e13488 doi:10.1371/journal.pone.0013488.

47. AhoS, BuissonM, PajunenT, RyooYW, GiotJF, et al. (2000) Ubinuclein, a novel nuclear protein interacting with cellular and viral transcription factors. J Cell Biol 148: 1165–1176.

48. BanumathyG, SomaiahN, ZhangR, TangY, HoffmannJ, et al. (2009) Human UBN1 is an ortholog of yeast Hpc2p and has an essential role in the HIRA/ASF1a chromatin-remodeling pathway in senescent cells. Mol Cell Biol 29: 758–770.

49. TangY, PuriA, RickettsMD, RaiTS, HoffmannJ, et al. (2012) Identification of an ubinuclein 1 region required for stability and function of the human HIRA/UBN1/CABIN1/ASF1a histone H3.3 chaperone complex. Biochemistry 51: 2366–2377.

50. Ait-AhmedO, Thomas-CavallinM, RossetR (1987) Isolation and characterization of a region of the Drosophila genome which contains a cluster of differentially expressed maternal genes (yema gene region). Dev Biol 122: 153–162.

51. Ait-AhmedO, BellonB, CapriM, JobletC, Thomas-DelaageM (1992) The yemanuclein-alpha: a new Drosophila DNA binding protein specific for the oocyte nucleus. Mech Dev 37: 69–80.

52. MeyerRE, DelaageM, RossetR, CapriM, Ait-AhmedO (2010) A single mutation results in diploid gamete formation and parthenogenesis in a Drosophila yemanuclein-alpha meiosis I defective mutant. BMC Genet 11: 104.

53. MoshkinYM, KanTW, GoodfellowH, BezstarostiK, MaedaRK, et al. (2009) Histone chaperones ASF1 and NAP1 differentially modulate removal of active histone marks by LID-RPD3 complexes during NOTCH silencing. Mol Cell 35: 782–793.

54. KonevAY, TribusM, ParkSY, PodhraskiV, LimCY, et al. (2007) CHD1 motor protein is required for deposition of histone variant H3.3 into chromatin in vivo. Science 317: 1087–1090.

55. Fuller M (1993) Spermatogenesis. In: Bate M, Martinez Arias A, editors. The development of Drosophila melanogaster. 1 ed. Cold Spring Harbor, NY. Cold Spring Harbor Laboratory Press: 71–148.

56. LoppinB, DocquierM, BonnetonF, CoubleP (2000) The maternal effect mutation sesame affects the formation of the male pronucleus in Drosophila melanogaster. Dev Biol 222: 392–404.

57. LoppinB, BergerF, CoubleP (2001) The Drosophila maternal gene sesame is required for sperm chromatin remodeling at fertilization. Chromosoma 110: 430–440.

58. DraneP, OuararhniK, DepauxA, ShuaibM, HamicheA (2010) The death-associated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3.3. Genes Dev 24: 1253–1265.

59. LewisPW, ElsaesserSJ, NohKM, StadlerSC, AllisCD (2010) Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres. Proc Natl Acad Sci U S A 107: 14075–14080.

60. WongLH, McGhieJD, SimM, AndersonMA, AhnS, et al. (2010) ATRX interacts with H3.3 in maintaining telomere structural integrity in pluripotent embryonic stem cells. Genome Res 20: 351–360.

61. BassettAR, CooperSE, RagabA, TraversAA (2008) The chromatin remodelling factor dATRX is involved in heterochromatin formation. PLoS ONE 3: e2099 doi:10.1371/journal.pone.0002099.

62. RobertsC, SutherlandHF, FarmerH, KimberW, HalfordS, et al. (2002) Targeted mutagenesis of the Hira gene results in gastrulation defects and patterning abnormalities of mesoendodermal derivatives prior to early embryonic lethality. Mol Cell Biol 22: 2318–2328.

63. Ray-GalletD, WoolfeA, VassiasI, PellentzC, LacosteN, et al. (2011) Dynamics of histone H3 deposition in vivo reveal a nucleosome gap-filling mechanism for H3.3 to maintain chromatin integrity. Mol Cell 44: 928–941.

64. KennedyAL, McBryanT, EndersGH, JohnsonFB, ZhangR, et al. (2010) Senescent mouse cells fail to overtly regulate the HIRA histone chaperone and do not form robust Senescence Associated Heterochromatin Foci. Cell Div 5: 16.

65. MoussonF, OchsenbeinF, MannC (2007) The histone chaperone Asf1 at the crossroads of chromatin and DNA checkpoint pathways. Chromosoma 116: 79–93.

66. Ray-GalletD, QuivyJP, SilljeHW, NiggEA, AlmouzniG (2007) The histone chaperone Asf1 is dispensable for direct de novo histone deposition in Xenopus egg extracts. Chromosoma 116: 487–496.

67. YeX, ZerlankoB, ZhangR, SomaiahN, LipinskiM, et al. (2007) Definition of pRB- and p53-dependent and -independent steps in HIRA/ASF1a-mediated formation of senescence-associated heterochromatin foci. Mol Cell Biol 27: 2452–2465.

68. SingerAB, GallJG (2011) An inducible nuclear body in the Drosophila germinal vesicle. Nucleus 2: 403–409.

69. DubruilleR, OrsiGA, DelabaereL, CortierE, CoubleP, et al. (2010) Specialization of a Drosophila capping protein essential for the protection of sperm telomeres. Curr Biol 20: 2090–2099.

70. SchuhM, LehnerCF, HeidmannS (2007) Incorporation of Drosophila CID/CENP-A and CENP-C into centromeres during early embryonic anaphase. Curr Biol 17: 237–243.

71. ThummelCS, BouletAM, LipshitzHD (1988) Vectors for Drosophila P-element-mediated transformation and tissue culture transfection. Gene 74: 445–456.

72. BischofJ, MaedaRK, HedigerM, KarchF, BaslerK (2007) An optimized transgenesis system for Drosophila using germ-line-specific phiC31 integrases. Proc Natl Acad Sci U S A 104: 3312–3317.

73. GrothAC, FishM, NusseR, CalosMP (2004) Construction of transgenic Drosophila by using the site-specific integrase from phage phiC31. Genetics 166: 1775–1782.

74. CapriM, SantoniMJ, Thomas-DelaageM, Ait-AhmedO (1997) Implication of a 5′ coding sequence in targeting maternal mRNA to the Drosophila oocyte. Mech Dev 68: 91–100.

75. AhoS, LupoJ, ColyPA, SabineA, CastellazziM, et al. (2009) Characterization of the ubinuclein protein as a new member of the nuclear and adhesion complex components (NACos). Biol Cell 101: 319–334.

76. JagerH, HerzigA, LehnerCF, HeidmannS (2001) Drosophila separase is required for sister chromatid separation and binds to PIM and THR. Genes Dev 15: 2572–2584.

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#