#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Tissue Specific Roles for the Ribosome Biogenesis Factor Wdr43 in Zebrafish Development


During vertebrate craniofacial development, neural crest cells (NCCs) contribute to most of the craniofacial pharyngeal skeleton. Defects in NCC specification, migration and differentiation resulting in malformations in the craniofacial complex are associated with human craniofacial disorders including Treacher-Collins Syndrome, caused by mutations in TCOF1. It has been hypothesized that perturbed ribosome biogenesis and resulting p53 mediated neuroepithelial apoptosis results in NCC hypoplasia in mouse Tcof1 mutants. However, the underlying mechanisms linking ribosome biogenesis and NCC development remain poorly understood. Here we report a new zebrafish mutant, fantome (fan), which harbors a point mutation and predicted premature stop codon in zebrafish wdr43, the ortholog to yeast UTP5. Although wdr43 mRNA is widely expressed during early zebrafish development, and its deficiency triggers early neural, eye, heart and pharyngeal arch defects, later defects appear fairly restricted to NCC derived craniofacial cartilages. Here we show that the C-terminus of Wdr43, which is absent in fan mutant protein, is both necessary and sufficient to mediate its nucleolar localization and protein interactions in metazoans. We demonstrate that Wdr43 functions in ribosome biogenesis, and that defects observed in fan mutants are mediated by a p53 dependent pathway. Finally, we show that proper localization of a variety of nucleolar proteins, including TCOF1, is dependent on that of WDR43. Together, our findings provide new insight into roles for Wdr43 in development, ribosome biogenesis, and also ribosomopathy-induced craniofacial phenotypes including Treacher-Collins Syndrome.


Vyšlo v časopise: Tissue Specific Roles for the Ribosome Biogenesis Factor Wdr43 in Zebrafish Development. PLoS Genet 10(1): e32767. doi:10.1371/journal.pgen.1004074
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004074

Souhrn

During vertebrate craniofacial development, neural crest cells (NCCs) contribute to most of the craniofacial pharyngeal skeleton. Defects in NCC specification, migration and differentiation resulting in malformations in the craniofacial complex are associated with human craniofacial disorders including Treacher-Collins Syndrome, caused by mutations in TCOF1. It has been hypothesized that perturbed ribosome biogenesis and resulting p53 mediated neuroepithelial apoptosis results in NCC hypoplasia in mouse Tcof1 mutants. However, the underlying mechanisms linking ribosome biogenesis and NCC development remain poorly understood. Here we report a new zebrafish mutant, fantome (fan), which harbors a point mutation and predicted premature stop codon in zebrafish wdr43, the ortholog to yeast UTP5. Although wdr43 mRNA is widely expressed during early zebrafish development, and its deficiency triggers early neural, eye, heart and pharyngeal arch defects, later defects appear fairly restricted to NCC derived craniofacial cartilages. Here we show that the C-terminus of Wdr43, which is absent in fan mutant protein, is both necessary and sufficient to mediate its nucleolar localization and protein interactions in metazoans. We demonstrate that Wdr43 functions in ribosome biogenesis, and that defects observed in fan mutants are mediated by a p53 dependent pathway. Finally, we show that proper localization of a variety of nucleolar proteins, including TCOF1, is dependent on that of WDR43. Together, our findings provide new insight into roles for Wdr43 in development, ribosome biogenesis, and also ribosomopathy-induced craniofacial phenotypes including Treacher-Collins Syndrome.


Zdroje

1. CorderoDR, BrugmannS, ChuY, BajpaiR, JameM, et al. (2011) Cranial neural crest cells on the move: their roles in craniofacial development. Am J Med Genet A 155A: 270–279.

2. MinouxM, RijliFM (2010) Molecular mechanisms of cranial neural crest cell migration and patterning in craniofacial development. Development 137: 2605–2621.

3. Gorlin RJ, Cohen MM, Levin LS (1990) Syndromes of the head and neck. New York; Oxford: Oxford University Press. xxi, 977.

4. TrainorPA (2010) Craniofacial birth defects: The role of neural crest cells in the etiology and pathogenesis of Treacher Collins syndrome and the potential for prevention. Am J Med Genet A 152A: 2984–2994.

5. Positional cloning of a gene involved in the pathogenesis of Treacher Collins syndrome. The Treacher Collins Syndrome Collaborative Group. Nat Genet 12: 130–136.

6. WiseCA, ChiangLC, PaznekasWA, SharmaM, MusyMM, et al. (1997) TCOF1 gene encodes a putative nucleolar phosphoprotein that exhibits mutations in Treacher Collins Syndrome throughout its coding region. Proc Natl Acad Sci U S A 94: 3110–3115.

7. HayanoT, YanagidaM, YamauchiY, ShinkawaT, IsobeT, et al. (2003) Proteomic analysis of human Nop56p-associated pre-ribosomal ribonucleoprotein complexes. Possible link between Nop56p and the nucleolar protein treacle responsible for Treacher Collins syndrome. J Biol Chem 278: 34309–34319.

8. ValdezBC, HenningD, SoRB, DixonJ, DixonMJ (2004) The Treacher Collins syndrome (TCOF1) gene product is involved in ribosomal DNA gene transcription by interacting with upstream binding factor. Proc Natl Acad Sci U S A 101: 10709–10714.

9. GonzalesB, HenningD, SoRB, DixonJ, DixonMJ, et al. (2005) The Treacher Collins syndrome (TCOF1) gene product is involved in pre-rRNA methylation. Hum Mol Genet 14: 2035–2043.

10. DixonJ, JonesNC, SandellLL, JayasingheSM, CraneJ, et al. (2006) Tcof1/Treacle is required for neural crest cell formation and proliferation deficiencies that cause craniofacial abnormalities. Proc Natl Acad Sci U S A 103: 13403–13408.

11. JonesNC, LynnML, GaudenzK, SakaiD, AotoK, et al. (2008) Prevention of the neurocristopathy Treacher Collins syndrome through inhibition of p53 function. Nat Med 14: 125–133.

12. CisternaB, BiggiogeraM (2010) Ribosome biogenesis: from structure to dynamics. Int Rev Cell Mol Biol 284: 67–111.

13. KresslerD, HurtE, BasslerJ (2010) Driving ribosome assembly. Biochim Biophys Acta 1803: 673–683.

14. DragonF, GallagherJE, Compagnone-PostPA, MitchellBM, PorwancherKA, et al. (2002) A large nucleolar U3 ribonucleoprotein required for 18S ribosomal RNA biogenesis. Nature 417: 967–970.

15. KroganNJ, PengWT, CagneyG, RobinsonMD, HawR, et al. (2004) High-definition macromolecular composition of yeast RNA-processing complexes. Mol Cell 13: 225–239.

16. GallagherJE, DunbarDA, GrannemanS, MitchellBM, OsheimY, et al. (2004) RNA polymerase I transcription and pre-rRNA processing are linked by specific SSU processome components. Genes Dev 18: 2506–2517.

17. FreedEF, BasergaSJ (2010) The C-terminus of Utp4, mutated in childhood cirrhosis, is essential for ribosome biogenesis. Nucleic Acids Res 38: 4798–4806.

18. WeryM, RuidantS, SchillewaertS, LeporeN, LafontaineDL (2009) The nuclear poly(A) polymerase and Exosome cofactor Trf5 is recruited cotranscriptionally to nucleolar surveillance. RNA 15: 406–419.

19. PrietoJL, McStayB (2007) Recruitment of factors linking transcription and processing of pre-rRNA to NOR chromatin is UBF-dependent and occurs independent of transcription in human cells. Genes Dev 21: 2041–2054.

20. FreedEF, PrietoJL, McCannKL, McStayB, BasergaSJ (2012) NOL11, implicated in the pathogenesis of North American Indian childhood cirrhosis, is required for pre-rRNA transcription and processing. PLoS Genet 8: e1002892.

21. McCannKL, BasergaSJ (2013) Genetics. Mysterious ribosomopathies. Science 341: 849–850.

22. ChagnonP, MichaudJ, MitchellG, MercierJ, MarionJF, et al. (2002) A missense mutation (R565W) in cirhin (FLJ14728) in North American Indian childhood cirrhosis. Am J Hum Genet 71: 1443–1449.

23. AzumaM, ToyamaR, LaverE, DawidIB (2006) Perturbation of rRNA synthesis in the bap28 mutation leads to apoptosis mediated by p53 in the zebrafish central nervous system. J Biol Chem 281: 13309–13316.

24. GallenbergerM, MeinelDM, KroeberM, WegnerM, MilkereitP, et al. (2011) Lack of WDR36 leads to preimplantation embryonic lethality in mice and delays the formation of small subunit ribosomal RNA in human cells in vitro. Hum Mol Genet 20: 422–435.

25. SkarieJM, LinkBA (2008) The primary open-angle glaucoma gene WDR36 functions in ribosomal RNA processing and interacts with the p53 stress-response pathway. Hum Mol Genet 17: 2474–2485.

26. ProvostE, WehnerKA, ZhongX, AsharF, NguyenE, et al. (2012) Ribosomal biogenesis genes play an essential and p53-independent role in zebrafish pancreas development. Development 139: 3232–3241.

27. BugnerV, TeczaA, GessertS, KuhlM (2011) Peter Pan functions independently of its role in ribosome biogenesis during early eye and craniofacial cartilage development in Xenopus laevis. Development 138: 2369–2378.

28. WangY, LuoY, HongY, PengJ, LoL (2012) Ribosome biogenesis factor Bms1-like is essential for liver development in zebrafish. J Genet Genomics 39: 451–462.

29. AndreevaV, ConnollyMH, Stewart-SwiftC, FraherD, BurtJ, et al. (2011) Identification of adult mineralized tissue zebrafish mutants. Genesis 49: 360–366.

30. LocascioA, NietoMA (2001) Cell movements during vertebrate development: integrated tissue behaviour versus individual cell migration. Curr Opin Genet Dev 11: 464–469.

31. YelickPC, SchillingTF (2002) Molecular dissection of craniofacial development using zebrafish. Crit Rev Oral Biol Med 13: 308–322.

32. Le DouarinNM, CalloniGW, DupinE (2008) The stem cells of the neural crest. Cell Cycle 7: 1013–1019.

33. YanYL, MillerCT, NissenRM, SingerA, LiuD, et al. (2002) A zebrafish sox9 gene required for cartilage morphogenesis. Development 129: 5065–5079.

34. 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.

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

36. ThomasT, KuriharaH, YamagishiH, KuriharaY, YazakiY, et al. (1998) A signaling cascade involving endothelin-1, dHAND and msx1 regulates development of neural-crest-derived branchial arch mesenchyme. Development 125: 3005–3014.

37. SperberSM, SaxenaV, HatchG, EkkerM (2008) Zebrafish dlx2a contributes to hindbrain neural crest survival, is necessary for differentiation of sensory ganglia and functions with dlx1a in maturation of the arch cartilage elements. Dev Biol 314: 59–70.

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

39. LiYP, BuschRK, ValdezBC, BuschH (1996) C23 interacts with B23, a putative nucleolar-localization-signal-binding protein. Eur J Biochem 237: 153–158.

40. GallagherJE, BasergaSJ (2004) Two-hybrid Mpp10p interaction-defective Imp4 proteins are not interaction defective in vivo but do confer specific pre-rRNA processing defects in Saccharomyces cerevisiae. Nucleic Acids Res 32: 1404–1413.

41. BevenAF, LeeR, RazazM, LeaderDJ, BrownJW, et al. (1996) The organization of ribosomal RNA processing correlates with the distribution of nucleolar snRNAs. J Cell Sci 109(Pt 6): 1241–1251.

42. MaN, MatsunagaS, TakataH, Ono-ManiwaR, UchiyamaS, et al. (2007) Nucleolin functions in nucleolus formation and chromosome congression. J Cell Sci 120: 2091–2105.

43. FumagalliS, ThomasG (2011) The role of p53 in ribosomopathies. Semin Hematol 48: 97–105.

44. FumagalliS, IvanenkovVV, TengT, ThomasG (2012) Suprainduction of p53 by disruption of 40S and 60S ribosome biogenesis leads to the activation of a novel G2/M checkpoint. Genes Dev 26: 1028–1040.

45. BursacS, BrdovcakMC, PfannkuchenM, OrsolicI, GolombL, et al. (2012) Mutual protection of ribosomal proteins L5 and L11 from degradation is essential for p53 activation upon ribosomal biogenesis stress. Proc Natl Acad Sci U S A 109: 20467–20472.

46. SuzukiA, KogoR, KawaharaK, SasakiM, NishioM, et al. (2012) A new PICTure of nucleolar stress. Cancer Sci 103: 632–637.

47. LeeKC, GohWL, XuM, KuaN, LunnyD, et al. (2008) Detection of the p53 response in zebrafish embryos using new monoclonal antibodies. Oncogene 27: 629–640.

48. GazdaHT, SheenMR, VlachosA, ChoesmelV, O'DonohueMF, et al. (2008) Ribosomal protein L5 and L11 mutations are associated with cleft palate and abnormal thumbs in Diamond-Blackfan anemia patients. Am J Hum Genet 83: 769–780.

49. UechiT, NakajimaY, ChakrabortyA, ToriharaH, HigaS, et al. (2008) Deficiency of ribosomal protein S19 during early embryogenesis leads to reduction of erythrocytes in a zebrafish model of Diamond-Blackfan anemia. Hum Mol Genet 17: 3204–3211.

50. DraptchinskaiaN, GustavssonP, AnderssonB, PetterssonM, WilligTN, et al. (1999) The gene encoding ribosomal protein S19 is mutated in Diamond-Blackfan anaemia. Nat Genet 21: 169–175.

51. DanilovaN, SakamotoKM, LinS (2008) Ribosomal protein S19 deficiency in zebrafish leads to developmental abnormalities and defective erythropoiesis through activation of p53 protein family. Blood 112: 5228–5237.

52. IwanamiN, HiguchiT, SasanoY, FujiwaraT, HoaVQ, et al. (2008) WDR55 is a nucleolar modulator of ribosomal RNA synthesis, cell cycle progression, and teleost organ development. PLoS Genet 4: e1000171.

53. AmsterdamA, SadlerKC, LaiK, FarringtonS, BronsonRT, et al. (2004) Many ribosomal protein genes are cancer genes in zebrafish. PLoS Biol 2: E139.

54. SakaiD, TrainorPA (2009) Treacher Collins syndrome: unmasking the role of Tcof1/treacle. Int J Biochem Cell Biol 41: 1229–1232.

55. DauwerseJG, DixonJ, SelandS, RuivenkampCA, van HaeringenA, et al. (2011) Mutations in genes encoding subunits of RNA polymerases I and III cause Treacher Collins syndrome. Nat Genet 43: 20–22.

56. YungBY, BuschRK, BuschH, MaugerAB, ChanPK (1985) Effects of actinomycin D analogs on nucleolar phosphoprotein B23 (37,000 daltons/pI 5.1). Biochem Pharmacol 34: 4059–4063.

57. LinCI, YehNH (2009) Treacle recruits RNA polymerase I complex to the nucleolus that is independent of UBF. Biochem Biophys Res Commun 386: 396–401.

58. KongR, ZhangL, HuL, PengQ, HanW, et al. (2011) hALP, a novel transcriptional U three protein (t-UTP), activates RNA polymerase I transcription by binding and acetylating the upstream binding factor (UBF). J Biol Chem 286: 7139–7148.

59. Hernandez-VerdunD, RousselP, ThiryM, SirriV, LafontaineDL (2010) The nucleolus: structure/function relationship in RNA metabolism. Wiley Interdiscip Rev RNA 1: 415–431.

60. Anastassova-KristevaM (1977) The nucleolar cycle in man. J Cell Sci 25: 103–110.

61. SullivanGJ, BridgerJM, CuthbertAP, NewboldRF, BickmoreWA, et al. (2001) Human acrocentric chromosomes with transcriptionally silent nucleolar organizer regions associate with nucleoli. EMBO J 20: 2867–2874.

62. BrangwynneCP, MitchisonTJ, HymanAA (2011) Active liquid-like behavior of nucleoli determines their size and shape in Xenopus laevis oocytes. Proc Natl Acad Sci U S A 108: 4334–4339.

63. ChenJ, RuanH, NgSM, GaoC, SooHM, et al. (2005) Loss of function of def selectively up-regulates Delta113p53 expression to arrest expansion growth of digestive organs in zebrafish. Genes Dev 19: 2900–2911.

64. KeeganBR, FeldmanJL, LeeDH, KoosDS, HoRK, et al. (2002) The elongation factors Pandora/Spt6 and Foggy/Spt5 promote transcription in the zebrafish embryo. Development 129: 1623–1632.

65. VillefrancJA, AmigoJ, LawsonND (2007) Gateway compatible vectors for analysis of gene function in the zebrafish. Dev Dyn 236: 3077–3087.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 1
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#