#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Stimulation of mTORC1 with L-leucine Rescues Defects Associated with Roberts Syndrome


Roberts syndrome (RBS) is a human disease characterized by defects in limb and craniofacial development and growth and mental retardation. RBS is caused by mutations in ESCO2, a gene which encodes an acetyltransferase for the cohesin complex. While the essential role of the cohesin complex in chromosome segregation has been well characterized, it plays additional roles in DNA damage repair, chromosome condensation, and gene expression. The developmental phenotypes of Roberts syndrome and other cohesinopathies suggest that gene expression is impaired during embryogenesis. It was previously reported that ribosomal RNA production and protein translation were impaired in immortalized RBS cells. It was speculated that cohesin binding at the rDNA was important for nucleolar form and function. We have explored the hypothesis that reduced ribosome function contributes to RBS in zebrafish models and human cells. Two key pathways that sense cellular stress are the p53 and mTOR pathways. We report that mTOR signaling is inhibited in human RBS cells based on the reduced phosphorylation of the downstream effectors S6K1, S6 and 4EBP1, and this correlates with p53 activation. Nucleoli, the sites of ribosome production, are highly fragmented in RBS cells. We tested the effect of inhibiting p53 or stimulating mTOR in RBS cells. The rescue provided by mTOR activation was more significant, with activation rescuing both cell division and cell death. To study this cohesinopathy in a whole animal model we used ESCO2-mutant and morphant zebrafish embryos, which have developmental defects mimicking RBS. Consistent with RBS patient cells, the ESCO2 mutant embryos show p53 activation and inhibition of the TOR pathway. Stimulation of the TOR pathway with L-leucine rescued many developmental defects of ESCO2-mutant embryos. Our data support the idea that RBS can be attributed in part to defects in ribosome biogenesis, and stimulation of the TOR pathway has therapeutic potential.


Vyšlo v časopise: Stimulation of mTORC1 with L-leucine Rescues Defects Associated with Roberts Syndrome. PLoS Genet 9(10): e32767. doi:10.1371/journal.pgen.1003857
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003857

Souhrn

Roberts syndrome (RBS) is a human disease characterized by defects in limb and craniofacial development and growth and mental retardation. RBS is caused by mutations in ESCO2, a gene which encodes an acetyltransferase for the cohesin complex. While the essential role of the cohesin complex in chromosome segregation has been well characterized, it plays additional roles in DNA damage repair, chromosome condensation, and gene expression. The developmental phenotypes of Roberts syndrome and other cohesinopathies suggest that gene expression is impaired during embryogenesis. It was previously reported that ribosomal RNA production and protein translation were impaired in immortalized RBS cells. It was speculated that cohesin binding at the rDNA was important for nucleolar form and function. We have explored the hypothesis that reduced ribosome function contributes to RBS in zebrafish models and human cells. Two key pathways that sense cellular stress are the p53 and mTOR pathways. We report that mTOR signaling is inhibited in human RBS cells based on the reduced phosphorylation of the downstream effectors S6K1, S6 and 4EBP1, and this correlates with p53 activation. Nucleoli, the sites of ribosome production, are highly fragmented in RBS cells. We tested the effect of inhibiting p53 or stimulating mTOR in RBS cells. The rescue provided by mTOR activation was more significant, with activation rescuing both cell division and cell death. To study this cohesinopathy in a whole animal model we used ESCO2-mutant and morphant zebrafish embryos, which have developmental defects mimicking RBS. Consistent with RBS patient cells, the ESCO2 mutant embryos show p53 activation and inhibition of the TOR pathway. Stimulation of the TOR pathway with L-leucine rescued many developmental defects of ESCO2-mutant embryos. Our data support the idea that RBS can be attributed in part to defects in ribosome biogenesis, and stimulation of the TOR pathway has therapeutic potential.


Zdroje

1. MichaelisC, CioskR, NasmythK (1997) Cohesins: chromosomal proteins that prevent premature separation of sister chromatids. Cell 91: 35–45.

2. XiongB, GertonJL Regulators of the cohesin network. Annu Rev Biochem 79: 131–153.

3. NasmythK Cohesin: a catenase with separate entry and exit gates? Nat Cell Biol 13: 1170–1177.

4. BeckouetF, HuB, RoigMB, SutaniT, KomataM, et al. An Smc3 acetylation cycle is essential for establishment of sister chromatid cohesion. Mol Cell 39: 689–699.

5. ZhangJ, ShiX, LiY, KimBJ, JiaJ, et al. (2008) Acetylation of Smc3 by Eco1 is required for S phase sister chromatid cohesion in both human and yeast. Mol Cell 31: 143–151.

6. LiuJ, KrantzID (2008) Cohesin and human disease. Annu Rev Genomics Hum Genet 9: 303–320.

7. VegaH, WaisfiszQ, GordilloM, SakaiN, YanagiharaI, et al. (2005) Roberts syndrome is caused by mutations in ESCO2, a human homolog of yeast ECO1 that is essential for the establishment of sister chromatid cohesion. Nat Genet 37: 468–470.

8. FreemanMV, WilliamsDW, SchimkeRN, TemtamySA, VachierE, et al. (1974) The Roberts syndrome. Birth Defects Orig Artic Ser 10: 87–95.

9. FreemanMV, WilliamsDW, SchimkeRN, TemtamySA, VachierE, et al. (1974) The Roberts syndrome. Clin Genet 5: 1–16.

10. GermanJ (1979) Roberts' syndrome. I. Cytological evidence for a disturbance in chromatid pairing. Clin Genet 16: 441–447.

11. TomkinsD, HunterA, RobertsM (1979) Cytogenetic findings in Roberts-SC phocomelia syndrome(s). Am J Med Genet 4: 17–26.

12. Van Den BergDJ, FranckeU (1993) Roberts syndrome: a review of 100 cases and a new rating system for severity. Am J Med Genet 47: 1104–1123.

13. GordilloM, VegaH, TrainerAH, HouF, SakaiN, et al. (2008) The molecular mechanism underlying Roberts syndrome involves loss of ESCO2 acetyltransferase activity. Hum Mol Genet 17: 2172–2180.

14. BoseT, LeeKK, LuS, XuB, HarrisB, et al. Cohesin proteins promote ribosomal RNA production and protein translation in yeast and human cells. PLoS Genet 8: e1002749.

15. GardS, LightW, XiongB, BoseT, McNairnAJ, et al. (2009) Cohesinopathy mutations disrupt the subnuclear organization of chromatin. J Cell Biol 187: 455–462.

16. LindstromMS, DeisenrothC, ZhangY (2007) Putting a finger on growth surveillance: insight into MDM2 zinc finger-ribosomal protein interactions. Cell Cycle 6: 434–437.

17. FumagalliS, Di CaraA, Neb-GulatiA, NattF, SchwembergerS, et al. (2009) Absence of nucleolar disruption after impairment of 40S ribosome biogenesis reveals an rpL11-translation-dependent mechanism of p53 induction. Nat Cell Biol 11: 501–508.

18. ZhangY, LuH (2009) Signaling to p53: ribosomal proteins find their way. Cancer Cell 16: 369–377.

19. CoqueretO (2003) New roles for p21 and p27 cell-cycle inhibitors: a function for each cell compartment? Trends Cell Biol 13: 65–70.

20. FumagalliS, IvanenkovVV, TengT, ThomasG 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.

21. DeisenrothC, ZhangY Ribosome biogenesis surveillance: probing the ribosomal protein-Mdm2-p53 pathway. Oncogene 29: 4253–4260.

22. ChakrabortyA, UechiT, HigaS, ToriharaH, KenmochiN (2009) Loss of ribosomal protein L11 affects zebrafish embryonic development through a p53-dependent apoptotic response. PLoS One 4: e4152.

23. OliverER, SaundersTL, TarleSA, GlaserT (2004) Ribosomal protein L24 defect in belly spot and tail (Bst), a mouse Minute. Development 131: 3907–3920.

24. Ferreira-CercaS, HurtE (2009) Cell biology: Arrest by ribosome. Nature 459: 46–47.

25. PanicL, TamarutS, Sticker-JantscheffM, BarkicM, SolterD, et al. (2006) Ribosomal protein S6 gene haploinsufficiency is associated with activation of a p53-dependent checkpoint during gastrulation. Mol Cell Biol 26: 8880–8891.

26. SchmelzleT, HallMN (2000) TOR, a central controller of cell growth. Cell 103: 253–262.

27. HolzMK, BallifBA, GygiSP, BlenisJ (2005) mTOR and S6K1 mediate assembly of the translation preinitiation complex through dynamic protein interchange and ordered phosphorylation events. Cell 123: 569–580.

28. ZoncuR, EfeyanA, SabatiniDM mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol 12: 21–35.

29. FingarDC, RichardsonCJ, TeeAR, CheathamL, TsouC, et al. (2004) mTOR controls cell cycle progression through its cell growth effectors S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. Mol Cell Biol 24: 200–216.

30. FingarDC, SalamaS, TsouC, HarlowE, BlenisJ (2002) Mammalian cell size is controlled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E. Genes Dev 16: 1472–1487.

31. HayN, SonenbergN (2004) Upstream and downstream of mTOR. Genes Dev 18: 1926–1945.

32. ChenH, FanM, PfefferLM, LaribeeRN The histone H3 lysine 56 acetylation pathway is regulated by target of rapamycin (TOR) signaling and functions directly in ribosomal RNA biogenesis. Nucleic Acids Res 40: 6534–6546.

33. Vazquez-MartinA, CufiS, Oliveras-FerrarosC, MenendezJA Raptor, a positive regulatory subunit of mTOR complex 1, is a novel phosphoprotein of the rDNA transcription machinery in nucleoli and chromosomal nucleolus organizer regions (NORs). Cell Cycle 10: 3140–3152.

34. ReiterA, SteinbauerR, PhilippiA, GerberJ, TschochnerH, et al. Reduction in ribosomal protein synthesis is sufficient to explain major effects on ribosome production after short-term TOR inactivation in Saccharomyces cerevisiae. Mol Cell Biol 31: 803–817.

35. DickinsonJM, RasmussenBB Essential amino acid sensing, signaling, and transport in the regulation of human muscle protein metabolism. Curr Opin Clin Nutr Metab Care 14: 83–88.

36. NicklinP, BergmanP, ZhangB, TriantafellowE, WangH, et al. (2009) Bidirectional transport of amino acids regulates mTOR and autophagy. Cell 136: 521–534.

37. NobukuniT, JoaquinM, RoccioM, DannSG, KimSY, et al. (2005) Amino acids mediate mTOR/raptor signaling through activation of class 3 phosphatidylinositol 3OH-kinase. Proc Natl Acad Sci U S A 102: 14238–14243.

38. BonfilsG, JaquenoudM, BontronS, OstrowiczC, UngermannC, et al. (2012) Leucyl-tRNA synthetase controls TORC1 via the EGO complex. Mol Cell 46: 105–110.

39. HanJM, JeongSJ, ParkMC, KimG, KwonNH, et al. (2012) Leucyl-tRNA synthetase is an intracellular leucine sensor for the mTORC1-signaling pathway. Cell 149: 410–424.

40. GertonJL (2012) Translational mechanisms at work in the cohesinopathies. Nucleus 3: 520–525.

41. van der LelijP, GodthelpBC, van ZonW, van GosligaD, OostraAB, et al. (2009) The cellular phenotype of Roberts syndrome fibroblasts as revealed by ectopic expression of ESCO2. PLoS One 4: e6936.

42. MonnichM, KurigerZ, PrintCG, HorsfieldJA A zebrafish model of Roberts syndrome reveals that Esco2 depletion interferes with development by disrupting the cell cycle. PLoS One 6: e20051.

43. KomarovPG, KomarovaEA, KondratovRV, Christov-TselkovK, CoonJS, et al. (1999) A chemical inhibitor of p53 that protects mice from the side effects of cancer therapy. Science 285: 1733–1737.

44. GaryRK, JensenDA (2005) The p53 inhibitor pifithrin-alpha forms a sparingly soluble derivative via intramolecular cyclization under physiological conditions. Mol Pharm 2: 462–474.

45. BarlowJL, DrynanLF, HewettDR, HolmesLR, Lorenzo-AbaldeS, et al. A p53-dependent mechanism underlies macrocytic anemia in a mouse model of human 5q- syndrome. Nat Med 16: 59–66.

46. RiekerC, EngblomD, KreinerG, DomanskyiA, SchoberA, et al. Nucleolar disruption in dopaminergic neurons leads to oxidative damage and parkinsonism through repression of mammalian target of rapamycin signaling. J Neurosci 31: 453–460.

47. ParlatoR, KreinerG, ErdmannG, RiekerC, StotzS, et al. (2008) Activation of an endogenous suicide response after perturbation of rRNA synthesis leads to neurodegeneration in mice. J Neurosci 28: 12759–12764.

48. PietrzakM, RempalaG, NelsonPT, ZhengJJ, HetmanM Epigenetic silencing of nucleolar rRNA genes in Alzheimer's disease. PLoS One 6: e22585.

49. LuS, GoeringM, GardS, XiongB, McNairnAJ, et al. (2010) Eco1 is important for DNA damage repair in S. cerevisiae. Cell Cycle 9: 3315–3327.

50. DennisPB, PullenN, KozmaSC, ThomasG (1996) The principal rapamycin-sensitive p70(s6k) phosphorylation sites, T-229 and T-389, are differentially regulated by rapamycin-insensitive kinase kinases. Mol Cell Biol 16: 6242–6251.

51. von ManteuffelSR, DennisPB, PullenN, GingrasAC, SonenbergN, et al. (1997) The insulin-induced signalling pathway leading to S6 and initiation factor 4E binding protein 1 phosphorylation bifurcates at a rapamycin-sensitive point immediately upstream of p70s6k. Mol Cell Biol 17: 5426–5436.

52. ChiangGG, AbrahamRT (2005) Phosphorylation of mammalian target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6 kinase. J Biol Chem 280: 25485–25490.

53. NaveBT, OuwensM, WithersDJ, AlessiDR, ShepherdPR (1999) Mammalian target of rapamycin is a direct target for protein kinase B: identification of a convergence point for opposing effects of insulin and amino-acid deficiency on protein translation. Biochem J 344 Pt 2: 427–431.

54. SekulicA, HudsonCC, HommeJL, YinP, OtternessDM, et al. (2000) A direct linkage between the phosphoinositide 3-kinase-AKT signaling pathway and the mammalian target of rapamycin in mitogen-stimulated and transformed cells. Cancer Res 60: 3504–3513.

55. AlexanderA, CaiSL, KimJ, NanezA, SahinM, et al. ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS. Proc Natl Acad Sci U S A 107: 4153–4158.

56. NortonLE, LaymanDK (2006) Leucine regulates translation initiation of protein synthesis in skeletal muscle after exercise. J Nutr 136: 533S–537S.

57. KimballSR, ShantzLM, HoretskyRL, JeffersonLS (1999) Leucine regulates translation of specific mRNAs in L6 myoblasts through mTOR-mediated changes in availability of eIF4E and phosphorylation of ribosomal protein S6. J Biol Chem 274: 11647–11652.

58. AnthonyJC, YoshizawaF, AnthonyTG, VaryTC, JeffersonLS, et al. (2000) Leucine stimulates translation initiation in skeletal muscle of postabsorptive rats via a rapamycin-sensitive pathway. J Nutr 130: 2413–2419.

59. PeyrollierK, HajduchE, BlairAS, HydeR, HundalHS (2000) L-leucine availability regulates phosphatidylinositol 3-kinase, p70 S6 kinase and glycogen synthase kinase-3 activity in L6 muscle cells: evidence for the involvement of the mammalian target of rapamycin (mTOR) pathway in the L-leucine-induced up-regulation of system A amino acid transport. Biochem J 350 Pt 2: 361–368.

60. ChenL, XuB, LiuL, LuoY, YinJ, et al. Hydrogen peroxide inhibits mTOR signaling by activation of AMPKalpha leading to apoptosis of neuronal cells. Lab Invest 90: 762–773.

61. StockerH, RadimerskiT, SchindelholzB, WittwerF, BelawatP, et al. (2003) Rheb is an essential regulator of S6K in controlling cell growth in Drosophila. Nat Cell Biol 5: 559–565.

62. ZhangY, GaoX, SaucedoLJ, RuB, EdgarBA, et al. (2003) Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins. Nat Cell Biol 5: 578–581.

63. GaramiA, ZwartkruisFJ, NobukuniT, JoaquinM, RoccioM, et al. (2003) Insulin activation of Rheb, a mediator of mTOR/S6K/4E-BP signaling, is inhibited by TSC1 and 2. Mol Cell 11: 1457–1466.

64. InokiK, LiY, XuT, GuanKL (2003) Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev 17: 1829–1834.

65. TeeAR, ManningBD, RouxPP, CantleyLC, BlenisJ (2003) Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr Biol 13: 1259–1268.

66. ManningBD, CantleyLC (2003) Rheb fills a GAP between TSC and TOR. Trends Biochem Sci 28: 573–576.

67. WullschlegerS, LoewithR, HallMN (2006) TOR signaling in growth and metabolism. Cell 124: 471–484.

68. FengZ, ZhangH, LevineAJ, JinS (2005) The coordinate regulation of the p53 and mTOR pathways in cells. Proc Natl Acad Sci U S A 102: 8204–8209.

69. BudanovAV, KarinM (2008) p53 target genes sestrin1 and sestrin2 connect genotoxic stress and mTOR signaling. Cell 134: 451–460.

70. HosoiH, DillingMB, ShikataT, LiuLN, ShuL, et al. (1999) Rapamycin causes poorly reversible inhibition of mTOR and induces p53-independent apoptosis in human rhabdomyosarcoma cells. Cancer Res 59: 886–894.

71. TsangCK, LiuH, ZhengXF mTOR binds to the promoters of RNA polymerase I- and III-transcribed genes. Cell Cycle 9: 953–957.

72. WinderWW (2001) Energy-sensing and signaling by AMP-activated protein kinase in skeletal muscle. J Appl Physiol 91: 1017–1028.

73. TerretME, SherwoodR, RahmanS, QinJ, JallepalliPV (2009) Cohesin acetylation speeds the replication fork. Nature 462: 231–234.

74. KangMA, SoEY, SimonsAL, SpitzDR, OuchiT DNA damage induces reactive oxygen species generation through the H2AX-Nox1/Rac1 pathway. Cell Death Dis 3: e249.

75. VafaO, WadeM, KernS, BeecheM, PanditaTK, et al. (2002) c-Myc can induce DNA damage, increase reactive oxygen species, and mitigate p53 function: a mechanism for oncogene-induced genetic instability. Mol Cell 9: 1031–1044.

76. MarchettiMA, WeinbergerM, MurakamiY, BurhansWC, HubermanJA (2006) Production of reactive oxygen species in response to replication stress and inappropriate mitosis in fission yeast. J Cell Sci 119: 124–131.

77. AmsterdamA, NissenRM, SunZ, SwindellEC, FarringtonS, et al. (2004) Identification of 315 genes essential for early zebrafish development. Proc Natl Acad Sci U S A 101: 12792–12797.

78. RooryckC, Diaz-FontA, OsbornDP, ChabchoubE, Hernandez-HernandezV, et al. Mutations in lectin complement pathway genes COLEC11 and MASP1 cause 3MC syndrome. Nat Genet 43: 197–203.

79. RokutandaS, FujitaT, KanataniN, YoshidaCA, KomoriH, et al. (2009) Akt regulates skeletal development through GSK3, mTOR, and FoxOs. Dev Biol 328: 78–93.

80. PengXD, XuPZ, ChenML, Hahn-WindgassenA, SkeenJ, et al. (2003) Dwarfism, impaired skin development, skeletal muscle atrophy, delayed bone development, and impeded adipogenesis in mice lacking Akt1 and Akt2. Genes Dev 17: 1352–1365.

81. WhelanG, KreidlE, WutzG, EgnerA, PetersJM, et al. Cohesin acetyltransferase Esco2 is a cell viability factor and is required for cohesion in pericentric heterochromatin. Embo J 31: 71–82.

82. RhodesJM, BentleyFK, PrintCG, DorsettD, MisulovinZ, et al. Positive regulation of c-Myc by cohesin is direct, and evolutionarily conserved. Dev Biol 344: 637–649.

83. MutoA, CalofAL, LanderAD, SchillingTF Multifactorial origins of heart and gut defects in nipbl-deficient zebrafish, a model of Cornelia de Lange Syndrome. PLoS Biol 9: e1001181.

84. GhiselliG (2006) SMC3 knockdown triggers genomic instability and p53-dependent apoptosis in human and zebrafish cells. Mol Cancer 5: 52.

85. DeardorffMA, KaurM, YaegerD, RampuriaA, KorolevS, et al. (2007) Mutations in cohesin complex members SMC3 and SMC1A cause a mild variant of cornelia de Lange syndrome with predominant mental retardation. Am J Hum Genet 80: 485–494.

86. DeardorffMA, WildeJJ, AlbrechtM, DickinsonE, TennstedtS, et al. RAD21 mutations cause a human cohesinopathy. Am J Hum Genet 90: 1014–1027.

87. BoseT, LeeKK, LuS, XuB, HarrisB, et al. (2012) Cohesin proteins promote ribosomal RNA production and protein translation in yeast and human cells. PLoS Genet 8: e1002749.

88. KawauchiS, CalofAL, SantosR, Lopez-BurksME, YoungCM, et al. (2009) Multiple organ system defects and transcriptional dysregulation in the Nipbl(+/−) mouse, a model of Cornelia de Lange Syndrome. PLoS Genet 5: e1000650.

89. MutoA, CalofAL, LanderAD, SchillingTF (2011) Multifactorial Origins of Heart and Gut Defects in nipbl-Deficient Zebrafish, a Model of Cornelia de Lange Syndrome. PLoS Biol 9: e1001181.

90. CarnevalliLS, MasudaK, FrigerioF, Le BacquerO, UmSH, et al. S6K1 plays a critical role in early adipocyte differentiation. Dev Cell 18: 763–774.

91. RubioED, ReissDJ, WelcshPL, DistecheCM, FilippovaGN, et al. (2008) CTCF physically links cohesin to chromatin. Proc Natl Acad Sci U S A 105: 8309–8314.

92. McEwanMV, EcclesMR, HorsfieldJA Cohesin is required for activation of MYC by estradiol. PLoS One 7: e49160.

93. StedmanW, KangH, LinS, KissilJL, BartolomeiMS, et al. (2008) Cohesins localize with CTCF at the KSHV latency control region and at cellular c-myc and H19/Igf2 insulators. Embo J 27: 654–666.

94. FilippovaGN, FagerlieS, KlenovaEM, MyersC, DehnerY, et al. (1996) An exceptionally conserved transcriptional repressor, CTCF, employs different combinations of zinc fingers to bind diverged promoter sequences of avian and mammalian c-myc oncogenes. Mol Cell Biol 16: 2802–2813.

95. GimiglianoA, ManniniL, BianchiL, PugliaM, DeardorffMA, et al. Proteomic profile identifies dysregulated pathways in Cornelia de Lange syndrome cells with distinct mutations in SMC1A and SMC3 genes. J Proteome Res 11: 6111–6123.

96. RemeseiroS, CuadradoA, Gomez-LopezG, PisanoDG, LosadaA A unique role of cohesin-SA1 in gene regulation and development. Embo J 31: 2090–2102.

97. van RiggelenJ, YetilA, FelsherDW MYC as a regulator of ribosome biogenesis and protein synthesis. Nat Rev Cancer 10: 301–309.

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

99. PellagattiA, Hellstrom-LindbergE, GiagounidisA, PerryJ, MalcovatiL, et al. (2008) Haploinsufficiency of RPS14 in 5q- syndrome is associated with deregulation of ribosomal- and translation-related genes. Br J Haematol 142: 57–64.

100. ChoesmelV, BacquevilleD, RouquetteJ, Noaillac-DepeyreJ, FribourgS, et al. (2007) Impaired ribosome biogenesis in Diamond-Blackfan anemia. Blood 109: 1275–1283.

101. BoocockGR, MorrisonJA, PopovicM, RichardsN, EllisL, et al. (2003) Mutations in SBDS are associated with Shwachman-Diamond syndrome. Nat Genet 33: 97–101.

102. PereboomTC, van WeeleLJ, BondtA, MacInnesAW A zebrafish model of dyskeratosis congenita reveals hematopoietic stem cell formation failure resulting from ribosomal protein-mediated p53 stabilization. Blood 118: 5458–5465.

103. FumagalliS, ThomasG The role of p53 in ribosomopathies. Semin Hematol 48: 97–105.

104. NarlaA, EbertBL Ribosomopathies: human disorders of ribosome dysfunction. Blood 115: 3196–3205.

105. YuanX, ZhouY, CasanovaE, ChaiM, KissE, et al. (2005) Genetic inactivation of the transcription factor TIF-IA leads to nucleolar disruption, cell cycle arrest, and p53-mediated apoptosis. Mol Cell 19: 77–87.

106. 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.

107. McGowanKA, PangWW, BhardwajR, PerezMG, PluvinageJV, et al. Reduced ribosomal protein gene dosage and p53 activation in low-risk myelodysplastic syndrome. Blood 118: 3622–3633.

108. McGowanKA, LiJZ, ParkCY, BeaudryV, TaborHK, et al. (2008) Ribosomal mutations cause p53-mediated dark skin and pleiotropic effects. Nat Genet 40: 963–970.

109. ToriharaH, UechiT, ChakrabortyA, ShinyaM, SakaiN, et al. (2011) Erythropoiesis failure due to RPS19 deficiency is independent of an activated Tp53 response in a zebrafish model of Diamond-Blackfan anaemia. Br J Haematol 152: 648–654.

110. 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.

111. PayneEM, VirgilioM, NarlaA, SunH, LevineM, et al. L-leucine improves the anemia and developmental defects associated with Diamond-Blackfan anemia and del(5q) MDS by activating the mTOR pathway. Blood 120: 2214–2224.

112. JaakoP, DebnathS, OlssonK, BryderD, FlygareJ, et al. Dietary L-leucine improves the anemia in a mouse model for Diamond-Blackfan anemia. Blood 120: 2225–2228.

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

114. ZhangY, SikesML, BeyerAL, SchneiderDA (2009) The Paf1 complex is required for efficient transcription elongation by RNA polymerase I. Proc Natl Acad Sci U S A 106: 2153–2158.

115. JonesRG, PlasDR, KubekS, BuzzaiM, MuJ, et al. (2005) AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol Cell 18: 283–293.

116. OkoshiR, OzakiT, YamamotoH, AndoK, KoidaN, et al. (2008) Activation of AMP-activated protein kinase induces p53-dependent apoptotic cell death in response to energetic stress. J Biol Chem 283: 3979–3987.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 10
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#