#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Systems-Level Interrogation Identifies Regulators of Blood Cell Number and Survival


Signaling networks that drive cell survival and proliferation regulate cell number in development and disease. We use a simple Drosophila model of cell number control, which centers on PDGF/VEGF receptor signaling. Performing a genome-wide RNAi screen under Pvr-sensitized conditions, we identify regulators of cell number that have not been found in conventional screens. Validation by in vivo genetics reveals previously unrecognized roles for EcR and InR in the balance of cell survival in the Drosophila embryo. Phosphoproteomic analysis demonstrates distinct mechanisms of cell survival regulation by EcR and receptor tyrosine kinase signaling. It further identifies common phosphorylation targets of Pvr and InR including regulators of cell survival, and receptor-specific phosphorylation targets mediating unique functions of Pvr and InR. Importantly, the study provides precedence that the selection of phosphorylation targets by signaling receptors can change with the signaling status of the cell, which may have wide-reaching implications for other cell regulatory systems.


Vyšlo v časopise: A Systems-Level Interrogation Identifies Regulators of Blood Cell Number and Survival. PLoS Genet 11(3): e32767. doi:10.1371/journal.pgen.1005056
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005056

Souhrn

Signaling networks that drive cell survival and proliferation regulate cell number in development and disease. We use a simple Drosophila model of cell number control, which centers on PDGF/VEGF receptor signaling. Performing a genome-wide RNAi screen under Pvr-sensitized conditions, we identify regulators of cell number that have not been found in conventional screens. Validation by in vivo genetics reveals previously unrecognized roles for EcR and InR in the balance of cell survival in the Drosophila embryo. Phosphoproteomic analysis demonstrates distinct mechanisms of cell survival regulation by EcR and receptor tyrosine kinase signaling. It further identifies common phosphorylation targets of Pvr and InR including regulators of cell survival, and receptor-specific phosphorylation targets mediating unique functions of Pvr and InR. Importantly, the study provides precedence that the selection of phosphorylation targets by signaling receptors can change with the signaling status of the cell, which may have wide-reaching implications for other cell regulatory systems.


Zdroje

1. Opferman JT (2007) Life and death during hematopoietic differentiation. Curr Opin Immunol 19: 497–502. 17662585

2. Wright KM, Deshmukh M (2006) Restricting apoptosis for postmitotic cell survival and its relevance to cancer. Cell Cycle 5: 1616–1620. 16880745

3. Parihar A, Eubank TD, Doseff AI Monocytes and macrophages regulate immunity through dynamic networks of survival and cell death. J Innate Immun 2: 204–215. doi: 10.1159/000296507 20375558

4. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144: 646–674. doi: 10.1016/j.cell.2011.02.013 21376230

5. Green DR, Evan GI (2002) A matter of life and death. Cancer Cell 1: 19–30. 12086884

6. Hoffmann JA, Reichhart JM (2002) Drosophila innate immunity: an evolutionary perspective. Nat Immunol 3: 121–126. 11812988

7. Martinez-Agosto JA, Mikkola HK, Hartenstein V, Banerjee U (2007) The hematopoietic stem cell and its niche: a comparative view. Genes Dev 21: 3044–3060. 18056420

8. Hartenstein V (2006) Blood cells and blood cell development in the animal kingdom. Annu Rev Cell Dev Biol 22: 677–712. 16824014

9. Lemaitre B, Hoffmann J (2007) The host defense of Drosophila melanogaster. Annu Rev Immunol 25: 697–743. 17201680

10. Evans CJ, Hartenstein V, Banerjee U (2003) Thicker than blood: conserved mechanisms in Drosophila and vertebrate hematopoiesis. Dev Cell 5: 673–690. 14602069

11. Gold KS, Brückner K (2014) Drosophila as a model for the two myeloid blood cell systems in vertebrates. Experimental hematology 42: 717–727. doi: 10.1016/j.exphem.2014.06.002 24946019

12. Brückner K, Kockel L, Duchek P, Luque CM, Rørth P, et al. (2004) The PDGF/VEGF Receptor controls blood cell survival in Drosophila. Dev Cell 7.

13. Munier AI, Doucet D, Perrodou E, Zachary D, Meister M, et al. (2002) PVF2, a PDGF/VEGF-like growth factor, induces hemocyte proliferation in Drosophila larvae. EMBO Rep 3: 1195–1200. 12446570

14. Tran TA, Kinch L, Pena-Llopis S, Kockel L, Grishin N, et al. (2013) Platelet-derived growth factor/vascular endothelial growth factor receptor inactivation by sunitinib results in Tsc1/Tsc2-dependent inhibition of TORC1. Molecular and cellular biology 33: 3762–3779. doi: 10.1128/MCB.01570-12 23878397

15. Mondal BC, Mukherjee T, Mandal L, Evans CJ, Sinenko SA, et al. (2011) Interaction between differentiating cell- and niche-derived signals in hematopoietic progenitor maintenance. Cell 147: 1589–1600. doi: 10.1016/j.cell.2011.11.041 22196733

16. Mondal BC, Shim J, Evans CJ, Banerjee U (2014) Pvr expression regulators in equilibrium signal control and maintenance of Drosophila blood progenitors. eLife 3: e03626. doi: 10.7554/eLife.03626 25201876

17. Bjorklund M, Taipale M, Varjosalo M, Saharinen J, Lahdenpera J, et al. (2006) Identification of pathways regulating cell size and cell-cycle progression by RNAi. Nature 439: 1009–1013. 16496002

18. Sims D, Duchek P, Baum B (2009) PDGF/VEGF signaling controls cell size in Drosophila. Genome Biol 10: R20. doi: 10.1186/gb-2009-10-2-r20 19216764

19. Rosin D, Schejter E, Volk T, Shilo B (2004) Apical accumulation of the Drosophila PDGF/VEGF receptor ligands provides a mechanism for triggering localized actin polymerization. Development: in press.

20. Wu Y, Brock AR, Wang Y, Fujitani K, Ueda R, et al. (2009) A blood-borne PDGF/VEGF-like ligand initiates wound-induced epidermal cell migration in Drosophila larvae. Curr Biol 19: 1473–1477. doi: 10.1016/j.cub.2009.07.019 19646875

21. Duchek P, Somogyi K, Jekely G, Beccari S, Rørth P (2001) Guidance of cell migration by the Drosophila PDGF/VEGF receptor. Cell 107: 17–26. 11595182

22. McDonald JA, Pinheiro EM, Montell DJ (2003) PVF1, a PDGF/VEGF homolog, is sufficient to guide border cells and interacts genetically with Taiman. Development 130: 3469–3478. 12810594

23. Cho NK, Keyes L, Johnson E, Heller J, Ryner L, et al. (2002) Developmental control of blood cell migration by the Drosophila VEGF pathway. Cell 108: 865–876. 11955438

24. Ashman LK (1999) The biology of stem cell factor and its receptor C-kit. Int J Biochem Cell Biol 31: 1037–1051. 10582338

25. Ferrara N, Gerber HP, LeCouter J (2003) The biology of VEGF and its receptors. Nat Med 9: 669–676. 12778165

26. Kelly LM, Gilliland DG (2002) Genetics of myeloid leukemias. Annu Rev Genomics Hum Genet 3: 179–198. 12194988

27. Koelle MR, Talbot WS, Segraves WA, Bender MT, Cherbas P, et al. (1991) The Drosophila EcR gene encodes an ecdysone receptor, a new member of the steroid receptor superfamily. Cell 67: 59–77. 1913820

28. Thomas HE, Stunnenberg HG, Stewart AF (1993) Heterodimerization of the Drosophila ecdysone receptor with retinoid X receptor and ultraspiracle. Nature 362: 471–475. 8385270

29. Cherbas L, Willingham A, Zhang D, Yang L, Zou Y, et al. (2011) The transcriptional diversity of 25 Drosophila cell lines. Genome research 21: 301–314. doi: 10.1101/gr.112961.110 21177962

30. Hay BA, Wolff T, Rubin GM (1994) Expression of baculovirus P35 prevents cell death in Drosophila. Development 120: 2121–2129. 7925015

31. Adams MD, Celniker SE, Holt RA, Evans CA, Gocayne JD, et al. (2000) The genome sequence of Drosophila melanogaster. Science 287: 2185–2195. 10731132

32. Perrimon N, Mathey-Prevot B (2007) Matter arising: off-targets and genome-scale RNAi screens in Drosophila. Fly (Austin) 1: 1–5.

33. Kulkarni MM, Booker M, Silver SJ, Friedman A, Hong P, et al. (2006) Evidence of off-target effects associated with long dsRNAs in Drosophila melanogaster cell-based assays. Nat Methods 3: 833–838. 16964256

34. Goberdhan DC, Wilson C (2003) The functions of insulin signaling: size isn't everything, even in Drosophila. Differentiation; research in biological diversity 71: 375–397. 12969331

35. King-Jones K, Korge G, Lehmann M (1999) The helix-loop-helix proteins dAP-4 and daughterless bind both in vitro and in vivo to SEBP3 sites required for transcriptional activation of the Drosophila gene Sgs-4. J Mol Biol 291: 71–82. 10438607

36. Rodriguez-Magadan H, Merino E, Schnabel D, Ramirez L, Lomeli H (2008) Spatial and temporal expression of Zimp7 and Zimp10 PIAS-like proteins in the developing mouse embryo. Gene Expr Patterns 8: 206–213. 18053775

37. Gutierrez L, Zurita M, Kennison JA, Vazquez M (2003) The Drosophila trithorax group gene tonalli (tna) interacts genetically with the Brahma remodeling complex and encodes an SP-RING finger protein. Development 130: 343–354. 12466201

38. Wang YH, Huang ML (2009) Reduction of Lobe leads to TORC1 hypoactivation that induces ectopic Jak/STAT signaling to impair Drosophila eye development. Mechanisms of development 126: 781–790. doi: 10.1016/j.mod.2009.08.005 19733656

39. Zoncu R, Efeyan A, Sabatini DM (2011) mTOR: from growth signal integration to cancer, diabetes and ageing. Nat Rev Mol Cell Biol 12: 21–35. doi: 10.1038/nrm3025 21157483

40. Sarbassov DD, Ali SM, Sabatini DM (2005) Growing roles for the mTOR pathway. Curr Opin Cell Biol 17: 596–603. 16226444

41. Manning BD, Cantley LC (2007) AKT/PKB signaling: navigating downstream. Cell 129: 1261–1274. 17604717

42. Janssens V, Goris J (2001) Protein phosphatase 2A: a highly regulated family of serine/threonine phosphatases implicated in cell growth and signalling. Biochem J 353: 417–439. 11171037

43. Van Hoof C, Goris J (2003) Phosphatases in apoptosis: to be or not to be, PP2A is in the heart of the question. Biochim Biophys Acta 1640: 97–104. 12729918

44. Kim M, Cha GH, Kim S, Lee JH, Park J, et al. (2004) MKP-3 has essential roles as a negative regulator of the Ras/mitogen-activated protein kinase pathway during Drosophila development. Mol Cell Biol 24: 573–583. 14701731

45. Yao TP, Segraves WA, Oro AE, McKeown M, Evans RM (1992) Drosophila ultraspiracle modulates ecdysone receptor function via heterodimer formation. Cell 71: 63–72. 1327536

46. Papoulas O, Beek SJ, Moseley SL, McCallum CM, Sarte M, et al. (1998) The Drosophila trithorax group proteins BRM, ASH1 and ASH2 are subunits of distinct protein complexes. Development 125: 3955–3966. 9735357

47. Tamkun JW (1995) The role of brahma and related proteins in transcription and development. Curr Opin Genet Dev 5: 473–477. 7580139

48. Colombani J, Bianchini L, Layalle S, Pondeville E, Dauphin-Villemant C, et al. (2005) Antagonistic actions of ecdysone and insulins determine final size in Drosophila. Science 310: 667–670. 16179433

49. Oro AE, McKeown M, Evans RM (1990) Relationship between the product of the Drosophila ultraspiracle locus and the vertebrate retinoid X receptor. Nature 347: 298–301. 2169594

50. Kozlova T, Thummel CS (2000) Steroid regulation of postembryonic development and reproduction in Drosophila. Trends Endocrinol Metab 11: 276–280. 10920384

51. Chavez VM, Marques G, Delbecque JP, Kobayashi K, Hollingsworth M, et al. (2000) The Drosophila disembodied gene controls late embryonic morphogenesis and codes for a cytochrome P450 enzyme that regulates embryonic ecdysone levels. Development 127: 4115–4126. 10976044

52. Cherbas L, Koehler MM, Cherbas P (1989) Effects of juvenile hormone on the ecdysone response of Drosophila Kc cells. Dev Genet 10: 177–188. 2500282

53. Andres AJ, Fletcher JC, Karim FD, Thummel CS (1993) Molecular analysis of the initiation of insect metamorphosis: a comparative study of Drosophila ecdysteroid-regulated transcription. Dev Biol 160: 388–404. 8253272

54. Stevens B, Alvarez CM, Bohman R, O'Connor JD (1980) An ecdysteroid-induced alteration in the cell cycle of cultured Drosophila cells. Cell 22: 675–682. 6780197

55. Rosset R (1978) Effects of ecdysone on a Drosophila cell line. Exp Cell Res 111: 31–36. 620695

56. Jiang C, Baehrecke EH, Thummel CS (1997) Steroid regulated programmed cell death during Drosophila metamorphosis. Development 124: 4673–4683. 9409683

57. Lee CY, Wendel DP, Reid P, Lam G, Thummel CS, et al. (2000) E93 directs steroid-triggered programmed cell death in Drosophila. Mol Cell 6: 433–443. 10983989

58. Lee CY, Simon CR, Woodard CT, Baehrecke EH (2002) Genetic mechanism for the stage- and tissue-specific regulation of steroid triggered programmed cell death in Drosophila. Developmental biology 252: 138–148. 12453466

59. Cherbas L, Hu X, Zhimulev I, Belyaeva E, Cherbas P (2003) EcR isoforms in Drosophila: testing tissue-specific requirements by targeted blockade and rescue. Development 130: 271–284. 12466195

60. Brown HL, Cherbas L, Cherbas P, Truman JW (2006) Use of time-lapse imaging and dominant negative receptors to dissect the steroid receptor control of neuronal remodeling in Drosophila. Development 133: 275–285. 16354717

61. Thummel CS (1995) From embryogenesis to metamorphosis: the regulation and function of Drosophila nuclear receptor superfamily members. Cell 83: 871–877. 8521511

62. Dayon L, Hainard A, Licker V, Turck N, Kuhn K, et al. (2008) Relative quantification of proteins in human cerebrospinal fluids by MS/MS using 6-plex isobaric tags. Analytical chemistry 80: 2921–2931. doi: 10.1021/ac702422x 18312001

63. Thompson A, Schafer J, Kuhn K, Kienle S, Schwarz J, et al. (2003) Tandem mass tags: a novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS. Analytical chemistry 75: 1895–1904. 12713048

64. Huang J, Wu S, Barrera J, Matthews K, Pan D (2005) The Hippo signaling pathway coordinately regulates cell proliferation and apoptosis by inactivating Yorkie, the Drosophila Homolog of YAP. Cell 122: 421–434. 16096061

65. Remillieux-Leschelle N, Santamaria P, Randsholt NB (2002) Regulation of larval hematopoiesis in Drosophila melanogaster: a role for the multi sex combs gene. Genetics 162: 1259–1274. 12454071

66. Luo H, Rose PE, Roberts TM, Dearolf CR (2002) The Hopscotch Jak kinase requires the Raf pathway to promote blood cell activation and differentiation in Drosophila. Mol Genet Genomics 267: 57–63. 11919715

67. Vrailas AD, Marenda DR, Cook SE, Powers MA, Lorenzen JA, et al. (2006) smoothened and thickveins regulate Moleskin/Importin 7-mediated MAP kinase signaling in the developing Drosophila eye. Development 133: 1485–1494. 16540506

68. Wilson TR, Fridlyand J, Yan Y, Penuel E, Burton L, et al. (2012) Widespread potential for growth-factor-driven resistance to anticancer kinase inhibitors. Nature 487: 505–509. doi: 10.1038/nature11249 22763448

69. Learte AR, Forero MG, Hidalgo A (2008) Gliatrophic and gliatropic roles of PVF/PVR signaling during axon guidance. Glia 56: 164–176. 18000865

70. Hoch RV, Soriano P (2003) Roles of PDGF in animal development. Development 130: 4769–4784. 12952899

71. Breen EC (2007) VEGF in biological control. J Cell Biochem 102: 1358–1367. 17979153

72. Scheijen B, Griffin JD (2002) Tyrosine kinase oncogenes in normal hematopoiesis and hematological disease. Oncogene 21: 3314–3333. 12032772

73. Gerber HP, Ferrara N (2003) The role of VEGF in normal and neoplastic hematopoiesis. J Mol Med 81: 20–31. 12545246

74. Ishimaru S, Ueda R, Hinohara Y, Ohtani M, Hanafusa H (2004) PVR plays a critical role via JNK activation in thorax closure during Drosophila metamorphosis. Embo J 23: 3984–3994. 15457211

75. Boutros M, Kiger AA, Armknecht S, Kerr K, Hild M, et al. (2004) Genome-wide RNAi analysis of growth and viability in Drosophila cells. Science 303: 832–835. 14764878

76. Bettencourt-Dias M, Giet R, Sinka R, Mazumdar A, Lock WG, et al. (2004) Genome-wide survey of protein kinases required for cell cycle progression. Nature 432: 980–987. 15616552

77. Chew SK, Chen P, Link N, Galindo KA, Pogue K, et al. (2009) Genome-wide silencing in Drosophila captures conserved apoptotic effectors. Nature 460: 123–127. doi: 10.1038/nature08087 19483676

78. Yi CH, Sogah DK, Boyce M, Degterev A, Christofferson DE, et al. (2007) A genome-wide RNAi screen reveals multiple regulators of caspase activation. J Cell Biol 179: 619–626. 17998402

79. Friedman AA, Tucker G, Singh R, Yan D, Vinayagam A, et al. (2011) Proteomic and functional genomic landscape of receptor tyrosine kinase and ras to extracellular signal-regulated kinase signaling. Sci Signal 4: rs10. doi: 10.1126/scisignal.2002029 22028469

80. Silvera D, Formenti SC, Schneider RJ (2010) Translational control in cancer. Nat Rev Cancer 10: 254–266. doi: 10.1038/nrc2824 20332778

81. Stumpf CR, Ruggero D (2011) The cancerous translation apparatus. Curr Opin Genet Dev.

82. Xue S, Barna M (2012) Specialized ribosomes: a new frontier in gene regulation and organismal biology. Nature reviews Molecular cell biology 13: 355–369. doi: 10.1038/nrm3359 22617470

83. Friedman A, Perrimon N (2006) A functional RNAi screen for regulators of receptor tyrosine kinase and ERK signalling. Nature 444: 230–234. 17086199

84. Marin-Hincapie M, Garofalo RS (1999) The carboxyl terminal extension of the Drosophila insulin receptor homologue binds IRS-1 and influences cell survival. The Journal of biological chemistry 274: 24987–24994. 10455177

85. van der Geer P, Hunter T, Lindberg RA (1994) Receptor protein-tyrosine kinases and their signal transduction pathways. Annu Rev Cell Biol 10: 251–337. 7888178

86. McCubrey JA, Steelman LS, Chappell WH, Sun L, Davis NM, et al. (2012) Advances in targeting signal transduction pathways. Oncotarget 3: 1505–1521. 23455493

87. Kockel L, Kerr KS, Melnick M, Bruckner K, Hebrok M, et al. (2010) Dynamic Switch of Negative Feedback Regulation in Drosophila Akt-TOR Signaling. PLoS Genet 6: e1000990. doi: 10.1371/journal.pgen.1000990 20585550

88. Nakashima A, Yoshino K, Miyamoto T, Eguchi S, Oshiro N, et al. (2007) Identification of TBC7 having TBC domain as a novel binding protein to TSC1-TSC2 complex. Biochem Biophys Res Commun 361: 218–223. 17658474

89. Matsuki T, Matthews RT, Cooper JA, van der Brug MP, Cookson MR, et al. (2011) Reelin and stk25 have opposing roles in neuronal polarization and dendritic Golgi deployment. Cell 143: 826–836.

90. Nogueira E, Fidalgo M, Molnar A, Kyriakis J, Force T, et al. (2008) SOK1 translocates from the Golgi to the nucleus upon chemical anoxia and induces apoptotic cell death. J Biol Chem 283: 16248–16258. doi: 10.1074/jbc.M709724200 18364353

91. Sorrentino RP, Carton Y, Govind S (2002) Cellular immune response to parasite infection in the Drosophila lymph gland is developmentally regulated. Dev Biol 243: 65–80. 11846478

92. Kozlova T, Thummel CS (2003) Essential roles for ecdysone signaling during Drosophila mid-embryonic development. Science 301: 1911–1914. 12958367

93. Gauhar Z, Sun LV, Hua S, Mason CE, Fuchs F, et al. (2009) Genomic mapping of binding regions for the Ecdysone receptor protein complex. Genome Res 19: 1006–1013. doi: 10.1101/gr.081349.108 19237466

94. Baehrecke EH, Thummel CS (1995) The Drosophila E93 gene from the 93F early puff displays stage- and tissue-specific regulation by 20-hydroxyecdysone. Dev Biol 171: 85–97. 7556910

95. Goyal L, McCall K, Agapite J, Hartwieg E, Steller H (2000) Induction of apoptosis by Drosophila reaper, hid and grim through inhibition of IAP function. Embo J 19: 589–597. 10675328

96. Meier P, Silke J, Leevers SJ, Evan GI (2000) The Drosophila caspase DRONC is regulated by DIAP1. EMBO J 19: 598–611. 10675329

97. Lee CY, Cooksey BA, Baehrecke EH (2002) Steroid regulation of midgut cell death during Drosophila development. Dev Biol 250: 101–111. 12297099

98. Choi YJ, Lee G, Park JH (2006) Programmed cell death mechanisms of identifiable peptidergic neurons in Drosophila melanogaster. Development 133: 2223–2232. 16672345

99. Kilpatrick ZE, Cakouros D, Kumar S (2005) Ecdysone-mediated up-regulation of the effector caspase DRICE is required for hormone-dependent apoptosis in Drosophila cells. J Biol Chem 280: 11981–11986. 15657059

100. Chittaranjan S, McConechy M, Hou YC, Freeman JD, Devorkin L, et al. (2009) Steroid hormone control of cell death and cell survival: molecular insights using RNAi. PLoS Genet 5: e1000379. doi: 10.1371/journal.pgen.1000379 19214204

101. Rewitz KF, Rybczynski R, Warren JT, Gilbert LI (2006) The Halloween genes code for cytochrome P450 enzymes mediating synthesis of the insect moulting hormone. Biochem Soc Trans 34: 1256–1260. 17073797

102. Cherbas L, Lee K, Cherbas P (1991) Identification of ecdysone response elements by analysis of the Drosophila Eip28/29 gene. Genes Dev 5: 120–131. 1899227

103. Schubiger M, Truman JW (2000) The RXR ortholog USP suppresses early metamorphic processes in Drosophila in the absence of ecdysteroids. Development 127: 1151–1159. 10683169

104. Owen GI, Zelent A (2000) Origins and evolutionary diversification of the nuclear receptor superfamily. Cell Mol Life Sci 57: 809–827. 10892345

105. King-Jones K, Thummel CS (2005) Nuclear receptors—a perspective from Drosophila. Nat Rev Genet 6: 311–323. 15803199

106. Ross SA, McCaffery PJ, Drager UC, De Luca LM (2000) Retinoids in embryonal development. Physiol Rev 80: 1021–1054. 10893430

107. Altucci L, Gronemeyer H (2001) Nuclear receptors in cell life and death. Trends Endocrinol Metab 12: 460–468. 11701345

108. Bastien J, Rochette-Egly C (2004) Nuclear retinoid receptors and the transcription of retinoid-target genes. Gene 328: 1–16. 15019979

109. Altucci L, Leibowitz MD, Ogilvie KM, de Lera AR, Gronemeyer H (2007) RAR and RXR modulation in cancer and metabolic disease. Nat Rev Drug Discov 6: 793–810. 17906642

110. Means AL, Gudas LJ (1995) The roles of retinoids in vertebrate development. Annu Rev Biochem 64: 201–233. 7574480

111. Bushue N, Wan YJ (2010) Retinoid pathway and cancer therapeutics. Adv Drug Deliv Rev 62: 1285–1298. doi: 10.1016/j.addr.2010.07.003 20654663

112. Lin RJ, Sternsdorf T, Tini M, Evans RM (2001) Transcriptional regulation in acute promyelocytic leukemia. Oncogene 20: 7204–7215. 11704848

113. Billottet C, Banerjee L, Vanhaesebroeck B, Khwaja A (2009) Inhibition of class I phosphoinositide 3-kinase activity impairs proliferation and triggers apoptosis in acute promyelocytic leukemia without affecting atra-induced differentiation. Cancer research 69: 1027–1036. doi: 10.1158/0008-5472.CAN-08-2608 19176369

114. Shim J, Gururaja-Rao S, Banerjee U (2013) Nutritional regulation of stem and progenitor cells in Drosophila. Development 140: 4647–4656. doi: 10.1242/dev.079087 24255094

115. Dragojlovic-Munther M, Martinez-Agosto JA (2012) Multifaceted roles of PTEN and TSC orchestrate growth and differentiation of Drosophila blood progenitors. Development 139: 3752–3763. 22951642

116. Benmimoun B, Polesello C, Waltzer L, Haenlin M (2012) Dual role for Insulin/TOR signaling in the control of hematopoietic progenitor maintenance in Drosophila. Development 139: 1713–1717. doi: 10.1242/dev.080259 22510984

117. Hsu HJ, Drummond-Barbosa D (2009) Insulin levels control female germline stem cell maintenance via the niche in Drosophila. Proceedings of the National Academy of Sciences of the United States of America 106: 1117–1121. doi: 10.1073/pnas.0809144106 19136634

118. Pritchett TL, McCall K (2012) Role of the insulin/Tor signaling network in starvation-induced programmed cell death in Drosophila oogenesis. Cell Death Differ 19: 1069–1079. doi: 10.1038/cdd.2011.200 22240900

119. Brogiolo W, Stocker H, Ikeya T, Rintelen F, Fernandez R, et al. (2001) An evolutionarily conserved function of the Drosophila insulin receptor and insulin-like peptides in growth control. Current biology: CB 11: 213–221. 11250149

120. Kurmasheva RT, Houghton PJ (2006) IGF-I mediated survival pathways in normal and malignant cells. Biochimica et biophysica acta 1766: 1–22. 16844299

121. Hsu PP, Kang SA, Rameseder J, Zhang Y, Ottina KA, et al. (2011) The mTOR-regulated phosphoproteome reveals a mechanism of mTORC1-mediated inhibition of growth factor signaling. Science 332: 1317–1322. doi: 10.1126/science.1199498 21659604

122. Yu Y, Yoon SO, Poulogiannis G, Yang Q, Ma XM, et al. (2011) Phosphoproteomic analysis identifies Grb10 as an mTORC1 substrate that negatively regulates insulin signaling. Science 332: 1322–1326. doi: 10.1126/science.1199484 21659605

123. Zhang Y, Liu Y, Li X, Gao W, Zhang W, et al. (2013) Effects of insulin and IGF-I on growth hormone- induced STAT5 activation in 3T3-F442A adipocytes. Lipids in health and disease 12: 56. doi: 10.1186/1476-511X-12-56 23631823

124. Coffer PJ, van Puijenbroek A, Burgering BM, Klop-de Jonge M, Koenderman L, et al. (1997) Insulin activates Stat3 independently of p21ras-ERK and PI-3K signal transduction. Oncogene 15: 2529–2539. 9399641

125. Orphanides G, Lagrange T, Reinberg D (1996) The general transcription factors of RNA polymerase II. Genes & development 10: 2657–2683.

126. Bianco A, Poukkula M, Cliffe A, Mathieu J, Luque CM, et al. (2007) Two distinct modes of guidance signalling during collective migration of border cells. Nature 448: 362–365. 17637670

127. Lindstrom DL, Squazzo SL, Muster N, Burckin TA, Wachter KC, et al. (2003) Dual roles for Spt5 in pre-mRNA processing and transcription elongation revealed by identification of Spt5-associated proteins. Molecular and cellular biology 23: 1368–1378. 12556496

128. Glatter T, Schittenhelm RB, Rinner O, Roguska K, Wepf A, et al. (2011) Modularity and hormone sensitivity of the Drosophila melanogaster insulin receptor/target of rapamycin interaction proteome. Molecular systems biology 7: 547. doi: 10.1038/msb.2011.79 22068330

129. Chung H, Kim AK, Jung SA, Kim SW, Yu K, et al. (2010) The Drosophila homolog of methionine sulfoxide reductase A extends lifespan and increases nuclear localization of FOXO. FEBS letters 584: 3609–3614. doi: 10.1016/j.febslet.2010.07.033 20655917

130. Roesijadi G, Rezvankhah S, Binninger DM, Weissbach H (2007) Ecdysone induction of MsrA protects against oxidative stress in Drosophila. Biochem Biophys Res Commun 354: 511–516. 17239346

131. Buszczak M, Lu X, Segraves WA, Chang TY, Cooley L (2002) Mutations in the midway gene disrupt a Drosophila acyl coenzyme A: diacylglycerol acyltransferase. Genetics 160: 1511–1518. 11973306

132. Buszczak M, Freeman MR, Carlson JR, Bender M, Cooley L, et al. (1999) Ecdysone response genes govern egg chamber development during mid-oogenesis in Drosophila. Development 126: 4581–4589. 10498692

133. Vettaikkorumakankauv AK, Lauffart B, Gangisetty O, Cincotta MA, Hawthorne LA, et al. (2008) The TACC proteins are coregulators of the Retinoid X Receptor beta. Cancer Therapy 6. 19129927

134. Mendoza MC, Er EE, Blenis J (2011) The Ras-ERK and PI3K-mTOR pathways: cross-talk and compensation. Trends in biochemical sciences 36: 320–328. doi: 10.1016/j.tibs.2011.03.006 21531565

135. Britten CD (2013) PI3K and MEK inhibitor combinations: examining the evidence in selected tumor types. Cancer chemotherapy and pharmacology 71: 1395–1409. doi: 10.1007/s00280-013-2121-1 23443307

136. Stramer B, Wood W, Galko MJ, Redd MJ, Jacinto A, et al. (2005) Live imaging of wound inflammation in Drosophila embryos reveals key roles for small GTPases during in vivo cell migration. J Cell Biol 168: 567–573. 15699212

137. Lee T, Luo L (1999) Mosaic analysis with a repressible cell marker for studies of gene function in neuronal morphogenesis. Neuron 22: 451–461. 10197526

138. Barolo S, Castro B, Posakony JW (2004) New Drosophila transgenic reporters: insulated P-element vectors expressing fast-maturing RFP. Biotechniques 36: 436–440, 442. 15038159

139. Parks AL, Cook KR, Belvin M, Dompe NA, Fawcett R, et al. (2004) Systematic generation of high-resolution deletion coverage of the Drosophila melanogaster genome. Nat Genet 36: 288–292. 14981519

140. Echalier G, Ohanessian A (1969) [Isolation, in tissue culture, of Drosophila melangaster cell lines]. C R Acad Sci Hebd Seances Acad Sci D 268: 1771–1773. 4976834

141. Clemens JC, Worby CA, Simonson-Leff N, Muda M, Maehama T, et al. (2000) Use of double-stranded RNA interference in Drosophila cell lines to dissect signal transduction pathways. Proc Natl Acad Sci U S A 97: 6499–6503. 10823906

142. Hild M, Beckmann B, Haas SA, Koch B, Solovyev V, et al. (2003) An integrated gene annotation and transcriptional profiling approach towards the full gene content of the Drosophila genome. Genome Biol 5: R3. 14709175

143. Eisen MB, Spellman PT, Brown PO, Botstein D (1998) Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci U S A 95: 14863–14868. 9843981

144. Villen J, Gygi SP (2008) The SCX/IMAC enrichment approach for global phosphorylation analysis by mass spectrometry. Nature protocols 3: 1630–1638. doi: 10.1038/nprot.2008.150 18833199

145. Kettenbach AN, Gerber SA (2011) Rapid and reproducible single-stage phosphopeptide enrichment of complex peptide mixtures: application to general and phosphotyrosine-specific phosphoproteomics experiments. Anal Chem 83: 7635–7644. doi: 10.1021/ac201894j 21899308

146. Rappsilber J, Ishihama Y, Mann M (2003) Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics. Analytical chemistry 75: 663–670. 12585499

147. Olsen JV, Macek B, Lange O, Makarov A, Horning S, et al. (2007) Higher-energy C-trap dissociation for peptide modification analysis. Nature methods 4: 709–712. 17721543

148. Dephoure N, Zhou C, Villen J, Beausoleil SA, Bakalarski CE, et al. (2008) A quantitative atlas of mitotic phosphorylation. Proceedings of the National Academy of Sciences of the United States of America 105: 10762–10767. doi: 10.1073/pnas.0805139105 18669648

149. Eng JKM, A.L., Yates J.R. 3rd (1994) An Approach to Correlate Tandem Mass Spectral Data of Peptides with Amino Acid Sequences in a Protein Database. J Am Soc Mass Spectrom 5: 976–989. doi: 10.1016/1044-0305(94)80016-2 24226387

150. Elias JE, Gygi SP (2007) Target-decoy search strategy for increased confidence in large-scale protein identifications by mass spectrometry. Nature methods 4: 207–214. 17327847

151. Huttlin EL, Jedrychowski MP, Elias JE, Goswami T, Rad R, et al. (2010) A tissue-specific atlas of mouse protein phosphorylation and expression. Cell 143: 1174–1189. doi: 10.1016/j.cell.2010.12.001 21183079

152. Beausoleil SA, Villen J, Gerber SA, Rush J, Gygi SP (2006) A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nature biotechnology 24: 1285–1292. 16964243

153. McAlister GC, Huttlin EL, Haas W, Ting L, Jedrychowski MP, et al. (2012) Increasing the multiplexing capacity of TMTs using reporter ion isotopologues with isobaric masses. Analytical chemistry 84: 7469–7478. doi: 10.1021/ac301572t 22880955

154. Vinayagam A, Hu Y, Kulkarni M, Roesel C, Sopko R, et al. (2013) Protein complex-based analysis framework for high-throughput data sets. Sci Signal 6: rs5. doi: 10.1126/scisignal.2003629 23443684

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 3
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#