#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

H2A.Z Acidic Patch Couples Chromatin Dynamics to Regulation of Gene Expression Programs during ESC Differentiation


The histone H2A variant H2A.Z is essential for embryonic development and for proper control of developmental gene expression programs in embryonic stem cells (ESCs). Divergent regions of amino acid sequence of H2A.Z likely determine its functional specialization compared to core histone H2A. For example, H2A.Z contains three divergent residues in the essential C-terminal acidic patch that reside on the surface of the histone octamer as an uninterrupted acidic patch domain; however, we know little about how these residues contribute to chromatin structure and function. Here, we show that the divergent amino acids Gly92, Asp97, and Ser98 in the H2A.Z C-terminal acidic patch (H2A.ZAP3) are critical for lineage commitment during ESC differentiation. H2A.Z is enriched at most H3K4me3 promoters in ESCs including poised, bivalent promoters that harbor both activating and repressive marks, H3K4me3 and H3K27me3 respectively. We found that while H2A.ZAP3 interacted with its deposition complex and displayed a highly similar distribution pattern compared to wild-type H2A.Z, its enrichment levels were reduced at target promoters. Further analysis revealed that H2A.ZAP3 was less tightly associated with chromatin, suggesting that the mutant is more dynamic. Notably, bivalent genes in H2A.ZAP3 ESCs displayed significant changes in expression compared to active genes. Moreover, bivalent genes in H2A.ZAP3 ESCs gained H3.3, a variant associated with higher nucleosome turnover, compared to wild-type H2A.Z. We next performed single cell imaging to measure H2A.Z dynamics. We found that H2A.ZAP3 displayed higher mobility in chromatin compared to wild-type H2A.Z by fluorescent recovery after photobleaching (FRAP). Moreover, ESCs treated with the transcriptional inhibitor flavopiridol resulted in a decrease in the H2A.ZAP3 mobile fraction and an increase in its occupancy at target genes indicating that the mutant can be properly incorporated into chromatin. Collectively, our work suggests that the divergent residues in the H2A.Z acidic patch comprise a unique domain that couples control of chromatin dynamics to the regulation of developmental gene expression patterns during lineage commitment.


Vyšlo v časopise: H2A.Z Acidic Patch Couples Chromatin Dynamics to Regulation of Gene Expression Programs during ESC Differentiation. PLoS Genet 9(8): e32767. doi:10.1371/journal.pgen.1003725
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003725

Souhrn

The histone H2A variant H2A.Z is essential for embryonic development and for proper control of developmental gene expression programs in embryonic stem cells (ESCs). Divergent regions of amino acid sequence of H2A.Z likely determine its functional specialization compared to core histone H2A. For example, H2A.Z contains three divergent residues in the essential C-terminal acidic patch that reside on the surface of the histone octamer as an uninterrupted acidic patch domain; however, we know little about how these residues contribute to chromatin structure and function. Here, we show that the divergent amino acids Gly92, Asp97, and Ser98 in the H2A.Z C-terminal acidic patch (H2A.ZAP3) are critical for lineage commitment during ESC differentiation. H2A.Z is enriched at most H3K4me3 promoters in ESCs including poised, bivalent promoters that harbor both activating and repressive marks, H3K4me3 and H3K27me3 respectively. We found that while H2A.ZAP3 interacted with its deposition complex and displayed a highly similar distribution pattern compared to wild-type H2A.Z, its enrichment levels were reduced at target promoters. Further analysis revealed that H2A.ZAP3 was less tightly associated with chromatin, suggesting that the mutant is more dynamic. Notably, bivalent genes in H2A.ZAP3 ESCs displayed significant changes in expression compared to active genes. Moreover, bivalent genes in H2A.ZAP3 ESCs gained H3.3, a variant associated with higher nucleosome turnover, compared to wild-type H2A.Z. We next performed single cell imaging to measure H2A.Z dynamics. We found that H2A.ZAP3 displayed higher mobility in chromatin compared to wild-type H2A.Z by fluorescent recovery after photobleaching (FRAP). Moreover, ESCs treated with the transcriptional inhibitor flavopiridol resulted in a decrease in the H2A.ZAP3 mobile fraction and an increase in its occupancy at target genes indicating that the mutant can be properly incorporated into chromatin. Collectively, our work suggests that the divergent residues in the H2A.Z acidic patch comprise a unique domain that couples control of chromatin dynamics to the regulation of developmental gene expression patterns during lineage commitment.


Zdroje

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

2. HenikoffS, AhmadK (2005) Assembly of variant histones into chromatin. Annu Rev Cell Dev Biol 21: 133–153.

3. LugerK, DechassaML, TremethickDJ (2012) New insights into nucleosome and chromatin structure: an ordered state or a disordered affair? Nat Rev Mol Cell Biol 13: 436–447.

4. GuillemetteB, GaudreauL (2006) Reuniting the contrasting functions of H2A.Z. Biochem Cell Biol 84: 528–535.

5. FaastR, ThonglairoamV, SchulzTC, BeallJ, WellsJR, et al. (2001) Histone variant H2A.Z is required for early mammalian development. Curr Biol 11: 1183–1187.

6. RidgwayP, BrownKD, RangasamyD, SvenssonU, TremethickDJ (2004) Unique residues on the H2A.Z containing nucleosome surface are important for Xenopus laevis development. J Biol Chem 279: 43815–43820.

7. DrakerR, CheungP (2009) Transcriptional and epigenetic functions of histone variant H2A.Z. Biochem Cell Biol 87: 19–25.

8. GuillemetteB, BatailleAR, GevryN, AdamM, BlanchetteM, et al. (2005) Variant histone H2A.Z is globally localized to the promoters of inactive yeast genes and regulates nucleosome positioning. PLoS Biol 3: e384.

9. SarmaK, ReinbergD (2005) Histone variants meet their match. Nat Rev Mol Cell Biol 6: 139–149.

10. CreyghtonMP, MarkoulakiS, LevineSS, HannaJ, LodatoMA, et al. (2008) H2AZ is enriched at polycomb complex target genes in ES cells and is necessary for lineage commitment. Cell 135: 649–661.

11. KuM, JaffeJD, KocheRP, RheinbayE, EndohM, et al. (2012) H2A.Z landscapes and dual modifications in pluripotent and multipotent stem cells underlie complex genome regulatory functions. Genome Biol 13: R85.

12. RaisnerRM, HartleyPD, MeneghiniMD, BaoMZ, LiuCL, et al. (2005) Histone variant H2A.Z marks the 5′ ends of both active and inactive genes in euchromatin. Cell 123: 233–248.

13. AzuaraV, PerryP, SauerS, SpivakovM, JorgensenHF, et al. (2006) Chromatin signatures of pluripotent cell lines. Nat Cell Biol 8: 532–538.

14. BernsteinBE, MikkelsenTS, XieX, KamalM, HuebertDJ, et al. (2006) A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125: 315–326.

15. KumarSV, WiggePA (2010) H2A.Z-containing nucleosomes mediate the thermosensory response in Arabidopsis. Cell 140: 136–147.

16. SutcliffeEL, ParishIA, HeYQ, JuelichT, TierneyML, et al. (2009) Dynamic histone variant exchange accompanies gene induction in T cells. Mol Cell Biol 29: 1972–1986.

17. HardyS, JacquesPE, GevryN, ForestA, FortinME, et al. (2009) The euchromatic and heterochromatic landscapes are shaped by antagonizing effects of transcription on H2A.Z deposition. PLoS Genet 5: e1000687.

18. CuiK, ZangC, RohTY, SchonesDE, ChildsRW, et al. (2009) Chromatin signatures in multipotent human hematopoietic stem cells indicate the fate of bivalent genes during differentiation. Cell Stem Cell 4: 80–93.

19. BernsteinBE, KamalM, Lindblad-TohK, BekiranovS, BaileyDK, et al. (2005) Genomic maps and comparative analysis of histone modifications in human and mouse. Cell 120: 169–181.

20. AbbottDW, IvanovaVS, WangX, BonnerWM, AusioJ (2001) Characterization of the stability and folding of H2A.Z chromatin particles: implications for transcriptional activation. J Biol Chem 276: 41945–41949.

21. ParkYJ, DyerPN, TremethickDJ, LugerK (2004) A new fluorescence resonance energy transfer approach demonstrates that the histone variant H2AZ stabilizes the histone octamer within the nucleosome. J Biol Chem 279: 24274–24282.

22. ParkYJ, LugerK (2008) Histone chaperones in nucleosome eviction and histone exchange. Curr Opin Struct Biol 18: 282–289.

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

24. JinC, ZangC, WeiG, CuiK, PengW, et al. (2009) H3.3/H2A.Z double variant-containing nucleosomes mark ‘nucleosome-free regions’ of active promoters and other regulatory regions. Nat Genet 41: 941–945.

25. JinC, FelsenfeldG (2007) Nucleosome stability mediated by histone variants H3.3 and H2A.Z. Genes Dev 21: 1519–1529.

26. ThakarA, GuptaP, IshibashiT, FinnR, Silva-MorenoB, et al. (2009) H2A.Z and H3.3 histone variants affect nucleosome structure: biochemical and biophysical studies. Biochemistry 48: 10852–10857.

27. SutoRK, ClarksonMJ, TremethickDJ, LugerK (2000) Crystal structure of a nucleosome core particle containing the variant histone H2A.Z. Nat Struct Biol 7: 1121–1124.

28. ClarksonMJ, WellsJR, GibsonF, SaintR, TremethickDJ (1999) Regions of variant histone His2AvD required for Drosophila development. Nature 399: 694–697.

29. JensenK, SantistebanMS, UrekarC, SmithMM Histone H2A.Z acid patch residues required for deposition and function. Mol Genet Genomics 285: 287–296.

30. WangAY, AristizabalMJ, RyanC, KroganNJ, KoborMS (2011) Key functional regions in the histone variant H2A.Z C-terminal docking domain. Mol Cell Biol 31: 3871–3884.

31. WrattingD, ThistlethwaiteA, HarrisM, ZeefLA, MillarCB A conserved function for the H2A.Z C terminus. J Biol Chem 287: 19148–19157.

32. JaenischR, YoungR (2008) Stem cells, the molecular circuitry of pluripotency and nuclear reprogramming. Cell 132: 567–582.

33. MurryCE, KellerG (2008) Differentiation of embryonic stem cells to clinically relevant populations: lessons from embryonic development. Cell 132: 661–680.

34. DryhurstD, IshibashiT, RoseKL, Eirin-LopezJM, McDonaldD, et al. (2009) Characterization of the histone H2A.Z-1 and H2A.Z-2 isoforms in vertebrates. BMC Biol 7: 86.

35. Eirin-LopezJM, Gonzalez-RomeroR, DryhurstD, IshibashiT, AusioJ (2009) The evolutionary differentiation of two histone H2A.Z variants in chordates (H2A.Z-1 and H2A.Z-2) is mediated by a stepwise mutation process that affects three amino acid residues. BMC Evol Biol 9: 31.

36. HuG, CuiK, NorthrupD, LiuC, WangC, et al. (2013) H2A.Z facilitates access of active and repressive complexes to chromatin in embryonic stem cell self-renewal and differentiation. Cell Stem Cell 12: 180–192.

37. BarskiA, CuddapahS, CuiK, RohTY, SchonesDE, et al. (2007) High-resolution profiling of histone methylations in the human genome. Cell 129: 823–837.

38. WamstadJA, AlexanderJM, TrutyRM, ShrikumarA, LiF, et al. (2012) Dynamic and coordinated epigenetic regulation of developmental transitions in the cardiac lineage. Cell 151: 206–220.

39. JiangC, PughBF (2009) Nucleosome positioning and gene regulation: advances through genomics. Nat Rev Genet 10: 161–172.

40. NockA, AscanoJM, BarreroMJ, MalikS (2012) Mediator-regulated transcription through the +1 nucleosome. Mol Cell 48: 837–848.

41. NekrasovM, AmrichovaJ, ParkerBJ, SobolevaTA, JackC, et al. (2012) Histone H2A.Z inheritance during the cell cycle and its impact on promoter organization and dynamics. Nat Struct Mol Biol 19: 1076–1083.

42. RuhlDD, JinJ, CaiY, SwansonS, FlorensL, et al. (2006) Purification of a human SRCAP complex that remodels chromatin by incorporating the histone variant H2A.Z into nucleosomes. Biochemistry 45: 5671–5677.

43. WongMM, CoxLK, ChriviaJC (2007) The chromatin remodeling protein, SRCAP, is critical for deposition of the histone variant H2A.Z at promoters. J Biol Chem 282: 26132–26139.

44. LukE, VuND, PattesonK, MizuguchiG, WuWH, et al. (2007) Chz1, a nuclear chaperone for histone H2AZ. Mol Cell 25: 357–368.

45. ThakarA, GuptaP, McAllisterWT, ZlatanovaJ (2010) Histone variant H2A.Z inhibits transcription in reconstituted nucleosomes. Biochemistry 49: 4018–4026.

46. ZhouJ, FanJY, RangasamyD, TremethickDJ (2007) The nucleosome surface regulates chromatin compaction and couples it with transcriptional repression. Nat Struct Mol Biol 14: 1070–1076.

47. MeshorerE, YellajoshulaD, GeorgeE, ScamblerPJ, BrownDT, et al. (2006) Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells. Dev Cell 10: 105–116.

48. HagerGL, McNallyJG, MisteliT (2009) Transcription dynamics. Mol Cell 35: 741–753.

49. ThirietC, HayesJJ (2006) Histone dynamics during transcription: exchange of H2A/H2B dimers and H3/H4 tetramers during pol II elongation. Results Probl Cell Differ 41: 77–90.

50. BhattacharyaD, TalwarS, MazumderA, ShivashankarGV (2009) Spatio-temporal plasticity in chromatin organization in mouse cell differentiation and during Drosophila embryogenesis. Biophys J 96: 3832–3839.

51. KimuraH (2005) Histone dynamics in living cells revealed by photobleaching. DNA Repair (Amst) 4: 939–950.

52. KimuraH, CookPR (2001) Kinetics of core histones in living human cells: little exchange of H3 and H4 and some rapid exchange of H2B. J Cell Biol 153: 1341–1353.

53. FanJY, GordonF, LugerK, HansenJC, TremethickDJ (2002) The essential histone variant H2A.Z regulates the equilibrium between different chromatin conformational states. Nat Struct Biol 9: 172–176.

54. ThambirajahAA, LiA, IshibashiT, AusioJ (2009) New developments in post-translational modifications and functions of histone H2A variants. Biochem Cell Biol 87: 7–17.

55. FanJY, RangasamyD, LugerK, TremethickDJ (2004) H2A.Z alters the nucleosome surface to promote HP1alpha-mediated chromatin fiber folding. Mol Cell 16: 655–661.

56. SeefeldtB, KasperR, SeidelT, TinnefeldP, DietzKJ, et al. (2008) Fluorescent proteins for single-molecule fluorescence applications. J Biophotonics 1: 74–82.

57. FlynnRA, AlmadaAE, ZamudioJR, SharpPA Antisense RNA polymerase II divergent transcripts are P-TEFb dependent and substrates for the RNA exosome. Proc Natl Acad Sci U S A 108: 10460–10465.

58. RahlPB, LinCY, SeilaAC, FlynnRA, McCuineS, et al. c-Myc regulates transcriptional pause release. Cell 141: 432–445.

59. SpragueBL, PegoRL, StavrevaDA, McNallyJG (2004) Analysis of binding reactions by fluorescence recovery after photobleaching. Biophys J 86: 3473–3495.

60. RangasamyD, GreavesI, TremethickDJ (2004) RNA interference demonstrates a novel role for H2A.Z in chromosome segregation. Nat Struct Mol Biol 11: 650–655.

61. MavrichTN, JiangC, IoshikhesIP, LiX, VentersBJ, et al. (2008) Nucleosome organization in the Drosophila genome. Nature 453: 358–362.

62. TevesSS, HenikoffS (2011) Heat shock reduces stalled RNA polymerase II and nucleosome turnover genome-wide. Genes Dev 25: 2387–2397.

63. WhittleCM, McClinicKN, ErcanS, ZhangX, GreenRD, et al. (2008) The genomic distribution and function of histone variant HTZ-1 during C. elegans embryogenesis. PLoS Genet 4: e1000187.

64. SurfaceLE, ThorntonSR, BoyerLA Polycomb group proteins set the stage for early lineage commitment. Cell Stem Cell 7: 288–298.

65. HuG, CuiK, NorthrupD, LiuC, WangC, TangQ, GeK, LevensD, Crane-RobinsonC, ZhaoK (2013) H2A.Z Facilitates Access of Active and Repressive Complexes to Chromatin in Embryonic Stem Cell Self-Renewal and Differentiation. Cell Stem Cell 12: 1–13.

66. CuadradoA, CorradoN, PerdigueroE, LafargaV, Munoz-CanovesP, et al. (2010) Essential role of p18Hamlet/SRCAP-mediated histone H2A.Z chromatin incorporation in muscle differentiation. Embo J 29: 2014–2025.

67. FazzioTG, HuffJT, PanningB (2008) An RNAi screen of chromatin proteins identifies Tip60-p400 as a regulator of embryonic stem cell identity. Cell 134: 162–174.

68. Papamichos-ChronakisM, KrebsJE, PetersonCL (2006) Interplay between Ino80 and Swr1 chromatin remodeling enzymes regulates cell cycle checkpoint adaptation in response to DNA damage. Genes Dev 20: 2437–2449.

69. Papamichos-ChronakisM, WatanabeS, RandoOJ, PetersonCL (2011) Global regulation of H2A.Z localization by the INO80 chromatin-remodeling enzyme is essential for genome integrity. Cell 144: 200–213.

70. WuWH, AlamiS, LukE, WuCH, SenS, et al. (2005) Swc2 is a widely conserved H2AZ-binding module essential for ATP-dependent histone exchange. Nat Struct Mol Biol 12: 1064–1071.

71. WatanabeS, Radman-LivajaM, RandoOJ, PetersonCL (2013) A histone acetylation switch regulates H2A.Z deposition by the SWR-C remodeling enzyme. Science 340: 195–199.

72. DrakerR, NgMK, SarcinellaE, IgnatchenkoV, KislingerT, et al. (2012) A combination of H2A.Z and H4 acetylation recruits Brd2 to chromatin during transcriptional activation. PLoS Genet 8: e1003047.

73. HalleyJE, KaplanT, WangAY, KoborMS, RineJ Roles for H2A.Z and its acetylation in GAL1 transcription and gene induction, but not GAL1-transcriptional memory. PLoS Biol 8: e1000401.

74. KimHS, VanoosthuyseV, FillinghamJ, RoguevA, WattS, et al. (2009) An acetylated form of histone H2A.Z regulates chromosome architecture in Schizosaccharomyces pombe. Nat Struct Mol Biol 16: 1286–1293.

75. KroganNJ, BaetzK, KeoghMC, DattaN, SawaC, et al. (2004) Regulation of chromosome stability by the histone H2A variant Htz1, the Swr1 chromatin remodeling complex, and the histone acetyltransferase NuA4. Proc Natl Acad Sci U S A 101: 13513–13518.

76. Valdes-MoraF, SongJZ, StathamAL, StrbenacD, RobinsonMD, et al. Acetylation of H2A.Z is a key epigenetic modification associated with gene deregulation and epigenetic remodeling in cancer. Genome Res 22: 307–321.

77. KalocsayM, HillerNJ, JentschS (2009) Chromosome-wide Rad51 spreading and SUMO-H2A.Z-dependent chromosome fixation in response to a persistent DNA double-strand break. Mol Cell 33: 335–343.

78. SarcinellaE, ZuzartePC, LauPN, DrakerR, CheungP (2007) Monoubiquitylation of H2A.Z distinguishes its association with euchromatin or facultative heterochromatin. Mol Cell Biol 27: 6457–6468.

79. BoyerLA, PlathK, ZeitlingerJ, BrambrinkT, MedeirosLA, et al. (2006) Polycomb complexes repress developmental regulators in murine embryonic stem cells. Nature 441: 349–353.

80. BeardC, HochedlingerK, PlathK, WutzA, JaenischR (2006) Efficient method to generate single-copy transgenic mice by site-specific integration in embryonic stem cells. Genesis 44: 23–28.

81. IuchiS, DabelsteenS, EasleyK, RheinwaldJG, GreenH (2006) Immortalized keratinocyte lines derived from human embryonic stem cells. Proc Natl Acad Sci U S A 103: 1792–1797.

82. MarsonA, LevineSS, ColeMF, FramptonGM, BrambrinkT, et al. (2008) Connecting microRNA genes to the core transcriptional regulatory circuitry of embryonic stem cells. Cell 134: 521–533.

83. BendallSC, HughesC, StewartMH, DobleB, BhatiaM, et al. (2008) Prevention of amino acid conversion in SILAC experiments with embryonic stem cells. Mol Cell Proteomics 7: 1587–1597.

84. ThomasCE, KelleherNL, MizzenCA (2006) Mass spectrometric characterization of human histone H3: a bird's eye view. J Proteome Res 5: 240–247.

85. Sherman MKaNE (2000) Protein Sequencing and Identification Using Tandem Mass Spectrometry Sons JWa, editor: John Wiley and Sons.

86. GarciaBA, MollahS, UeberheideBM, BusbySA, MuratoreTL, et al. (2007) Chemical derivatization of histones for facilitated analysis by mass spectrometry. Nat Protoc 2: 933–938.

87. JaffeJD, KeshishianH, ChangB, AddonaTA, GilletteMA, et al. (2008) Accurate inclusion mass screening: a bridge from unbiased discovery to targeted assay development for biomarker verification. Mol Cell Proteomics 7: 1952–1962.

88. GoshimaG, KiyomitsuT, YodaK, YanagidaM (2003) Human centromere chromatin protein hMis12, essential for equal segregation, is independent of CENP-A loading pathway. J Cell Biol 160: 25–39.

89. ToyodaY, YanagidaM (2006) Coordinated requirements of human topo II and cohesin for metaphase centromere alignment under Mad2-dependent spindle checkpoint surveillance. Mol Biol Cell 17: 2287–2302.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 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#