#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

An Essential Role of Variant Histone H3.3 for Ectomesenchyme Potential of the Cranial Neural Crest


The neural crest (NC) is a vertebrate-specific cell population that exhibits remarkable multipotency. Although derived from the neural plate border (NPB) ectoderm, cranial NC (CNC) cells contribute not only to the peripheral nervous system but also to the ectomesenchymal precursors of the head skeleton. To date, the developmental basis for such broad potential has remained elusive. Here, we show that the replacement histone H3.3 is essential during early CNC development for these cells to generate ectomesenchyme and head pigment precursors. In a forward genetic screen in zebrafish, we identified a dominant D123N mutation in h3f3a, one of five zebrafish variant histone H3.3 genes, that eliminates the CNC–derived head skeleton and a subset of pigment cells yet leaves other CNC derivatives and trunk NC intact. Analyses of nucleosome assembly indicate that mutant D123N H3.3 interferes with H3.3 nucleosomal incorporation by forming aberrant H3 homodimers. Consistent with CNC defects arising from insufficient H3.3 incorporation into chromatin, supplying exogenous wild-type H3.3 rescues head skeletal development in mutants. Surprisingly, embryo-wide expression of dominant mutant H3.3 had little effect on embryonic development outside CNC, indicating an unexpectedly specific sensitivity of CNC to defects in H3.3 incorporation. Whereas previous studies had implicated H3.3 in large-scale histone replacement events that generate totipotency during germ line development, our work has revealed an additional role of H3.3 in the broad potential of the ectoderm-derived CNC, including the ability to make the mesoderm-like ectomesenchymal precursors of the head skeleton.


Vyšlo v časopise: An Essential Role of Variant Histone H3.3 for Ectomesenchyme Potential of the Cranial Neural Crest. PLoS Genet 8(9): e32767. doi:10.1371/journal.pgen.1002938
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1002938

Souhrn

The neural crest (NC) is a vertebrate-specific cell population that exhibits remarkable multipotency. Although derived from the neural plate border (NPB) ectoderm, cranial NC (CNC) cells contribute not only to the peripheral nervous system but also to the ectomesenchymal precursors of the head skeleton. To date, the developmental basis for such broad potential has remained elusive. Here, we show that the replacement histone H3.3 is essential during early CNC development for these cells to generate ectomesenchyme and head pigment precursors. In a forward genetic screen in zebrafish, we identified a dominant D123N mutation in h3f3a, one of five zebrafish variant histone H3.3 genes, that eliminates the CNC–derived head skeleton and a subset of pigment cells yet leaves other CNC derivatives and trunk NC intact. Analyses of nucleosome assembly indicate that mutant D123N H3.3 interferes with H3.3 nucleosomal incorporation by forming aberrant H3 homodimers. Consistent with CNC defects arising from insufficient H3.3 incorporation into chromatin, supplying exogenous wild-type H3.3 rescues head skeletal development in mutants. Surprisingly, embryo-wide expression of dominant mutant H3.3 had little effect on embryonic development outside CNC, indicating an unexpectedly specific sensitivity of CNC to defects in H3.3 incorporation. Whereas previous studies had implicated H3.3 in large-scale histone replacement events that generate totipotency during germ line development, our work has revealed an additional role of H3.3 in the broad potential of the ectoderm-derived CNC, including the ability to make the mesoderm-like ectomesenchymal precursors of the head skeleton.


Zdroje

1. GansC, NorthcuttRG (1983) Neural Crest and the Origin of Vertebrates: A New Head. Science 220: 268–273.

2. BaroffioA, DupinE, Le DouarinNM (1991) Common precursors for neural and mesectodermal derivatives in the cephalic neural crest. Development 112: 301–305.

3. Bronner-FraserM, FraserSE (1988) Cell lineage analysis reveals multipotency of some avian neural crest cells. Nature 335: 161–164.

4. PlattJB (1893) Ectodermic origin of the cartilages of the head. Anat Anz 8: 506–509.

5. SchillingTF, KimmelCB (1994) Segment and cell type lineage restrictions during pharyngeal arch development in the zebrafish embryo. Development 120: 483–494.

6. BreauMA, PietriT, StemmlerMP, ThieryJP, WestonJA (2008) A nonneural epithelial domain of embryonic cranial neural folds gives rise to ectomesenchyme. Proc Natl Acad Sci U S A 105: 7750–7755.

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

8. VastenhouwNL, ZhangY, WoodsIG, ImamF, RegevA, et al. (2010) Chromatin signature of embryonic pluripotency is established during genome activation. Nature 464: 922–926.

9. Rada-IglesiasA, BajpaiR, SwigutT, BrugmannSA, FlynnRA, et al. (2011) A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470: 279–283.

10. GargiuloG, LevyS, BucciG, RomanenghiM, FornasariL, et al. (2009) NA-Seq: a discovery tool for the analysis of chromatin structure and dynamics during differentiation. Dev Cell 16: 466–481.

11. BajpaiR, ChenDA, Rada-IglesiasA, ZhangJ, XiongY, et al. (2010) CHD7 cooperates with PBAF to control multipotent neural crest formation. Nature 463: 958–962.

12. ErogluB, WangG, TuN, SunX, MivechiNF (2006) Critical role of Brg1 member of the SWI/SNF chromatin remodeling complex during neurogenesis and neural crest induction in zebrafish. Dev Dyn 235: 2722–2735.

13. Strobl-MazzullaPH, Sauka-SpenglerT, Bronner-FraserM (2010) Histone demethylase JmjD2A regulates neural crest specification. Dev Cell 19: 460–468.

14. TakeuchiT, WatanabeY, Takano-ShimizuT, KondoS (2006) Roles of jumonji and jumonji family genes in chromatin regulation and development. Dev Dyn 235: 2449–2459.

15. PattenSA, Jacobs-McDanielsNL, ZaouterC, DrapeauP, AlbertsonRC, et al. (2012) Role of Chd7 in zebrafish: a model for CHARGE syndrome. PLoS ONE 7: e31650 doi:10.1371/journal.pone.0031650.

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

17. HajkovaP, AncelinK, WaldmannT, LacosteN, LangeUC, et al. (2008) Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 452: 877–881.

18. 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.

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

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

21. 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.

22. LawsonND, WeinsteinBM (2002) In vivo imaging of embryonic vascular development using transgenic zebrafish. Dev Biol 248: 307–318.

23. CoulyGF, ColteyPM, Le DouarinNM (1993) The triple origin of skull in higher vertebrates: a study in quail-chick chimeras. Development 117: 409–429.

24. KimmelCB, MillerCT, MoensCB (2001) Specification and morphogenesis of the zebrafish larval head skeleton. Dev Biol 233: 239–257.

25. ArduiniBL, BosseKM, HenionPD (2009) Genetic ablation of neural crest cell diversification. Development 136: 1987–1994.

26. LiYX, ZdanowiczM, YoungL, KumiskiD, LeatherburyL, et al. (2003) Cardiac neural crest in zebrafish embryos contributes to myocardial cell lineage and early heart function. Dev Dyn 226: 540–550.

27. ThisseB, HeyerV, LuxA, AlunniV, DegraveA, et al. (2004) Spatial and temporal expression of the zebrafish genome by large-scale in situ hybridization screening. Methods Cell Biol 77: 505–519.

28. PaulsS, Geldmacher-VossB, Campos-OrtegaJA (2001) A zebrafish histone variant H2A.F/Z and a transgenic H2A.F/Z:GFP fusion protein for in vivo studies of embryonic development. Dev Genes Evol 211: 603–610.

29. LugerK, MaderAW, RichmondRK, SargentDF, RichmondTJ (1997) Crystal structure of the nucleosome core particle at 2.8 A resolution. Nature 389: 251–260.

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

31. MeulemansD, Bronner-FraserM (2004) Gene-regulatory interactions in neural crest evolution and development. Dev Cell 7: 291–299.

32. NgRK, GurdonJB (2008) Epigenetic memory of an active gene state depends on histone H3.3 incorporation into chromatin in the absence of transcription. Nat Cell Biol 10: 102–109.

33. McKittrickE, GafkenPR, AhmadK, HenikoffS (2004) Histone H3.3 is enriched in covalent modifications associated with active chromatin. Proc Natl Acad Sci U S A 101: 1525–1530.

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

35. Ray-GalletD, QuivyJP, ScampsC, MartiniEM, LipinskiM, et al. (2002) HIRA is critical for a nucleosome assembly pathway independent of DNA synthesis. Mol Cell 9: 1091–1100.

36. 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.

37. 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.

38. SawatsubashiS, MurataT, LimJ, FujikiR, ItoS, et al. (2010) A histone chaperone, DEK, transcriptionally coactivates a nuclear receptor. Genes Dev 24: 159–170.

39. 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.

40. Wise-DraperTM, Mintz-ColeRA, MorrisTA, SimpsonDS, Wikenheiser-BrokampKA, et al. (2009) Overexpression of the cellular DEK protein promotes epithelial transformation in vitro and in vivo. Cancer research 69: 1792–1799.

41. MichaelsonJS, BaderD, KuoF, KozakC, LederP (1999) Loss of Daxx, a promiscuously interacting protein, results in extensive apoptosis in early mouse development. Genes & development 13: 1918–1923.

42. SchnetzMP, BartelsCF, ShastriK, BalasubramanianD, ZentnerGE, et al. (2009) Genomic distribution of CHD7 on chromatin tracks H3K4 methylation patterns. Genome Res 19: 590–601.

43. BakerCV, Bronner-FraserM, Le DouarinNM, TeilletMA (1997) Early- and late-migrating cranial neural crest cell populations have equivalent developmental potential in vivo. Development 124: 3077–3087.

44. WestonJA, YoshidaH, RobinsonV, NishikawaS, FraserST (2004) Neural crest and the origin of ectomesenchyme: Neural fold heterogeneity suggests an alternative hypothesis. Dev Dyn 229: 118–130.

45. DorskyRI, MoonRT, RaibleDW (1998) Control of neural crest cell fate by the Wnt signalling pathway. Nature 396: 370–373.

46. StreisingerG, WalkerC, DowerN, KnauberD, SingerF (1981) Production of clones of homozygous diploid zebra fish (Brachydanio rerio). Nature 291: 293–296.

47. KimmelCB, BallardWW, KimmelSR, UllmannB, SchillingTF (1995) Stages of embryonic development of the zebrafish. Dev Dyn 203: 253–310.

48. WalkerMB, KimmelCB (2007) A two-color acid-free cartilage and bone stain for zebrafish larvae. Biotech Histochem 82: 23–28.

49. ZunigaE, RippenM, AlexanderC, SchillingTF, CrumpJG (2011) Gremlin 2 regulates distinct roles of BMP and Endothelin 1 signaling in dorsoventral patterning of the facial skeleton. Development 138: 5147–5156.

50. PhillipsBT, KwonHJ, MeltonC, HoughtalingP, FritzA, et al. (2006) Zebrafish msxB, msxC and msxE function together to refine the neural-nonneural border and regulate cranial placodes and neural crest development. Dev Biol 294: 376–390.

51. SeoHC, SaetreBO, HavikB, EllingsenS, FjoseA (1998) The zebrafish Pax3 and Pax7 homologues are highly conserved, encode multiple isoforms and show dynamic segment-like expression in the developing brain. Mech Dev 70: 49–63.

52. ToyamaR, GomezDM, ManaMD, DawidIB (2004) Sequence relationships and expression patterns of zebrafish zic2 and zic5 genes. Gene Expr Patterns 4: 345–350.

53. ThisseC, ThisseB, SchillingTF, PostlethwaitJH (1993) Structure of the zebrafish snail1 gene and its expression in wild-type, spadetail and no tail mutant embryos. Development 119: 1203–1215.

54. DuttonKA, PaulinyA, LopesSS, ElworthyS, CarneyTJ, et al. (2001) Zebrafish colourless encodes sox10 and specifies non-ectomesenchymal neural crest fates. Development 128: 4113–4125.

55. FurthauerM, ThisseC, ThisseB (1997) A role for FGF-8 in the dorsoventral patterning of the zebrafish gastrula. Development 124: 4253–4264.

56. YanYL, WilloughbyJ, LiuD, CrumpJG, WilsonC, et al. (2005) A pair of Sox: distinct and overlapping functions of zebrafish sox9 co-orthologs in craniofacial and pectoral fin development. Development 132: 1069–1083.

57. AkimenkoMA, EkkerM, WegnerJ, LinW, WesterfieldM (1994) Combinatorial expression of three zebrafish genes related to distal-less: part of a homeobox gene code for the head. J Neurosci 14: 3475–3486.

58. KraussS, JohansenT, KorzhV, FjoseA (1991) Expression of the zebrafish paired box gene pax[zf-b] during early neurogenesis. Development 113: 1193–1206.

59. OxtobyE, JowettT (1993) Cloning of the zebrafish krox-20 gene (krx-20) and its expression during hindbrain development. Nucleic Acids Res 21: 1087–1095.

60. LuoR, AnM, ArduiniBL, HenionPD (2001) Specific pan-neural crest expression of zebrafish Crestin throughout embryonic development. Dev Dyn 220: 169–174.

61. GermanguzI, LevD, WaismanT, KimCH, GitelmanI (2007) Four twist genes in zebrafish, four expression patterns. Dev Dyn 236: 2615–2626.

62. IgnatiusMS, MooseHE, El-HodiriHM, HenionPD (2008) colgate/hdac1 Repression of foxd3 expression is required to permit mitfa-dependent melanogenesis. Dev Biol 313: 568–583.

63. MavesL, JackmanW, KimmelCB (2002) FGF3 and FGF8 mediate a rhombomere 4 signaling activity in the zebrafish hindbrain. Development 129: 3825–3837.

64. KwanKM, FujimotoE, GrabherC, MangumBD, HardyME, et al. (2007) The Tol2kit: a multisite gateway-based construction kit for Tol2 transposon transgenesis constructs. Dev Dyn 236: 3088–3099.

65. HeckmanKL, PeaseLR (2007) Gene splicing and mutagenesis by PCR-driven overlap extension. Nat Protoc 2: 924–932.

66. OoiSL, PriessJR, HenikoffS (2006) Histone H3.3 variant dynamics in the germline of Caenorhabditis elegans. PLoS Genet 2: e97 doi:10.1371/journal.pgen.0020097.

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

68. JungSY, MalovannayaA, WeiJ, O'MalleyBW, QinJ (2005) Proteomic analysis of steady-state nuclear hormone receptor coactivator complexes. Mol Endocrinol 19: 2451–2465.

69. CrumpJG, SwartzME, KimmelCB (2004) An Integrin-Dependent Role of Pouch Endoderm in Hyoid Cartilage Development. PLoS Biol 2: e244 doi:10.1371/journal.pbio.0020244.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 9
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#