#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

A Kinome-Wide RNAi Screen in Glia Reveals That the RIO Kinases Mediate Cell Proliferation and Survival through TORC2-Akt Signaling in Glioblastoma


Glioblastoma, the most common primary malignant brain tumor, is incurable with current therapies. Genetic and molecular analyses demonstrate that glioblastomas frequently display mutations that activate receptor tyrosine kinase (RTK) and Pi-3 kinase (PI3K) signaling pathways. In Drosophila melanogaster, activation of RTK and PI3K pathways in glial progenitor cells creates malignant neoplastic glial tumors that display many features of human glioblastoma. In both human and Drosophila, activation of the RTK and PI3K pathways stimulates Akt signaling along with other as-yet-unknown changes that drive oncogenesis. We used this Drosophila glioblastoma model to perform a kinome-wide genetic screen for new genes required for RTK- and PI3K-dependent neoplastic transformation. Human orthologs of novel kinases uncovered by these screens were functionally assessed in mammalian glioblastoma models and human tumors. Our results revealed that the atypical kinases RIOK1 and RIOK2 are overexpressed in glioblastoma cells in an Akt-dependent manner. Moreover, we found that overexpressed RIOK2 formed a complex with RIOK1, mTor, and mTor-complex-2 components, and that overexpressed RIOK2 upregulated Akt signaling and promoted tumorigenesis in murine astrocytes. Conversely, reduced expression of RIOK1 or RIOK2 disrupted Akt signaling and caused cell cycle exit, apoptosis, and chemosensitivity in glioblastoma cells by inducing p53 activity through the RpL11-dependent ribosomal stress checkpoint. These results imply that, in glioblastoma cells, constitutive Akt signaling drives RIO kinase overexpression, which creates a feedforward loop that promotes and maintains oncogenic Akt activity through stimulation of mTor signaling. Further study of the RIO kinases as well as other kinases identified in our Drosophila screen may reveal new insights into defects underlying glioblastoma and related cancers and may reveal new therapeutic opportunities for these cancers.


Vyšlo v časopise: A Kinome-Wide RNAi Screen in Glia Reveals That the RIO Kinases Mediate Cell Proliferation and Survival through TORC2-Akt Signaling in Glioblastoma. PLoS Genet 9(2): e32767. doi:10.1371/journal.pgen.1003253
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003253

Souhrn

Glioblastoma, the most common primary malignant brain tumor, is incurable with current therapies. Genetic and molecular analyses demonstrate that glioblastomas frequently display mutations that activate receptor tyrosine kinase (RTK) and Pi-3 kinase (PI3K) signaling pathways. In Drosophila melanogaster, activation of RTK and PI3K pathways in glial progenitor cells creates malignant neoplastic glial tumors that display many features of human glioblastoma. In both human and Drosophila, activation of the RTK and PI3K pathways stimulates Akt signaling along with other as-yet-unknown changes that drive oncogenesis. We used this Drosophila glioblastoma model to perform a kinome-wide genetic screen for new genes required for RTK- and PI3K-dependent neoplastic transformation. Human orthologs of novel kinases uncovered by these screens were functionally assessed in mammalian glioblastoma models and human tumors. Our results revealed that the atypical kinases RIOK1 and RIOK2 are overexpressed in glioblastoma cells in an Akt-dependent manner. Moreover, we found that overexpressed RIOK2 formed a complex with RIOK1, mTor, and mTor-complex-2 components, and that overexpressed RIOK2 upregulated Akt signaling and promoted tumorigenesis in murine astrocytes. Conversely, reduced expression of RIOK1 or RIOK2 disrupted Akt signaling and caused cell cycle exit, apoptosis, and chemosensitivity in glioblastoma cells by inducing p53 activity through the RpL11-dependent ribosomal stress checkpoint. These results imply that, in glioblastoma cells, constitutive Akt signaling drives RIO kinase overexpression, which creates a feedforward loop that promotes and maintains oncogenic Akt activity through stimulation of mTor signaling. Further study of the RIO kinases as well as other kinases identified in our Drosophila screen may reveal new insights into defects underlying glioblastoma and related cancers and may reveal new therapeutic opportunities for these cancers.


Zdroje

1. FurnariFB, FentonT, BachooRM, MukasaA, StommelJM, et al. (2007) Malignant astrocytic glioma: genetics, biology, and paths to treatment. Genes Dev 21: 2683–2710.

2. NishikawaR, JiXD, HarmonRC, LazarCS, GillGN, et al. (1994) A mutant epidermal growth factor receptor common in human glioma confers enhanced tumorigenicity. Proc Natl Acad Sci U S A 91: 7727–7731.

3. MukasaA, WykoskyJ, LigonKL, ChinL, CaveneeWK, et al. (2010) Mutant EGFR is required for maintenance of glioma growth in vivo, and its ablation leads to escape from receptor dependence. Proc Natl Acad Sci U S A 107: 2616–2621.

4. HollandEC, CelestinoJ, DaiC, SchaeferL, SawayaRE, et al. (2000) Combined activation of Ras and Akt in neural progenitors induces glioblastoma formation in mice. Nat Genet 25: 55–57.

5. MarumotoT, TashiroA, Friedmann-MorvinskiD, ScadengM, SodaY, et al. (2009) Development of a novel mouse glioma model using lentiviral vectors. Nat Med 15: 110–116.

6. BachooRM, MaherEA, LigonKL, SharplessNE, ChanSS, et al. (2002) Epidermal growth factor receptor and Ink4a/Arf: convergent mechanisms governing terminal differentiation and transformation along the neural stem cell to astrocyte axis. Cancer Cell 1: 269–277.

7. HollandEC, HivelyWP, DePinhoRA, VarmusHE (1998) A constitutively active epidermal growth factor receptor cooperates with disruption of G1 cell-cycle arrest pathways to induce glioma-like lesions in mice. Genes Dev 12: 3675–3685.

8. CloughesyTF, YoshimotoK, NghiemphuP, BrownK, DangJ, et al. (2008) Antitumor activity of rapamycin in a Phase I trial for patients with recurrent PTEN-deficient glioblastoma. PLoS Med 5: e8 doi:10.1371/journal.pmed.0050008.

9. ReadRD, CaveneeWK, FurnariFB, ThomasJB (2009) A Drosophila model for EGFR-Ras and PI3K-dependent human glioma. PLoS Genet 5: e1000374 doi:10.1371/journal.pgen.1000374.

10. ReiterLT, BierE (2002) Using Drosophila melanogaster to uncover human disease gene function and potential drug target proteins. Expert Opin Ther Targets 6: 387–399.

11. St JohnstonD (2002) The art and design of genetic screens: Drosophila melanogaster. Nat Rev Genet 3: 176–188.

12. DietzlG, ChenD, SchnorrerF, SuKC, BarinovaY, et al. (2007) A genome-wide transgenic RNAi library for conditional gene inactivation in Drosophila. Nature 448: 151–156.

13. BellenHJ, TongC, TsudaH (2011) 100 years of Drosophila research and its impact on vertebrate neuroscience: a history lesson for the future. Nat Rev Neurosci 11: 514–522.

14. FreemanMR, DohertyJ (2006) Glial cell biology in Drosophila and vertebrates. Trends Neurosci 29: 82–90.

15. LaRonde-LeBlancN, WlodawerA (2005) A family portrait of the RIO kinases. J Biol Chem 280: 37297–37300.

16. VanrobaysE, GelugneJP, GleizesPE, Caizergues-FerrerM (2003) Late cytoplasmic maturation of the small ribosomal subunit requires RIO proteins in Saccharomyces cerevisiae. Mol Cell Biol 23: 2083–2095.

17. WidmannB, WandreyF, BadertscherL, WylerE, PfannstielJ, et al. (2011) The kinase activity of human Rio1 is required for final steps of cytoplasmic maturation of 40S subunits. Mol Biol Cell

18. ZempI, WildT, O'DonohueMF, WandreyF, WidmannB, et al. (2009) Distinct cytoplasmic maturation steps of 40S ribosomal subunit precursors require hRio2. J Cell Biol 185: 1167–1180.

19. BaumasK, SoudetJ, Caizergues-FerrerM, FaubladierM, HenryY, et al. (2012) Human RioK3 is a novel component of cytoplasmic pre-40S pre-ribosomal particles. RNA Biol 9: 162–174.

20. StrunkBS, NovakMN, YoungCL, KarbsteinK (2012) A translation-like cycle is a quality control checkpoint for maturing 40S ribosome subunits. Cell 150: 111–121.

21. OlsenJV, BlagoevB, GnadF, MacekB, KumarC, et al. (2006) Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell 127: 635–648.

22. BreitkreutzA, ChoiH, SharomJR, BoucherL, NeduvaV, et al. (2010) A global protein kinase and phosphatase interaction network in yeast. Science 328: 1043–1046.

23. LuoJ, EmanueleMJ, LiD, CreightonCJ, SchlabachMR, et al. (2009) A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell 137: 835–848.

24. ManningG, PlowmanGD, HunterT, SudarsanamS (2002) Evolution of protein kinase signaling from yeast to man. Trends Biochem Sci 27: 514–520.

25. MorrisonDK, MurakamiMS, CleghonV (2000) Protein kinases and phosphatases in the Drosophila genome. J Cell Biol 150: F57–62.

26. SzklarczykD, FranceschiniA, KuhnM, SimonovicM, RothA, et al. (2011) The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res 39: D561–568.

27. BjorklundM, TaipaleM, VarjosaloM, SaharinenJ, LahdenperaJ, et al. (2006) Identification of pathways regulating cell size and cell-cycle progression by RNAi. Nature 439: 1009–1013.

28. Bettencourt-DiasM, GietR, SinkaR, MazumdarA, LockWG, et al. (2004) Genome-wide survey of protein kinases required for cell cycle progression. Nature 432: 980–987.

29. FriedmanA, PerrimonN (2006) A functional RNAi screen for regulators of receptor tyrosine kinase and ERK signalling. Nature 444: 230–234.

30. BoutrosM, KigerAA, ArmknechtS, KerrK, HildM, et al. (2004) Genome-wide RNAi analysis of growth and viability in Drosophila cells. Science 303: 832–835.

31. NeumullerRA, RichterC, FischerA, NovatchkovaM, NeumullerKG, et al. (2011) Genome-Wide Analysis of Self-Renewal in Drosophila Neural Stem Cells by Transgenic RNAi. Cell Stem Cell 8: 580–593.

32. Sousa-NunesR, YeeLL, GouldAP (2011) Fat cells reactivate quiescent neuroblasts via TOR and glial insulin relays in Drosophila. Nature 471: 508–512.

33. ReddyBV, IrvineKD (2011) Regulation of Drosophila glial cell proliferation by Merlin-Hippo signaling. Development 138: 5201–5212.

34. HuangHS, NaganeM, KlingbeilCK, LinH, NishikawaR, et al. (1997) The enhanced tumorigenic activity of a mutant epidermal growth factor receptor common in human cancers is mediated by threshold levels of constitutive tyrosine phosphorylation and unattenuated signaling. J Biol Chem 272: 2927–2935.

35. LaksDR, Masterman-SmithM, VisnyeiK, AngenieuxB, OrozcoNM, et al. (2009) Neurosphere formation is an independent predictor of clinical outcome in malignant glioma. Stem Cells 27: 980–987.

36. PollardSM, YoshikawaK, ClarkeID, DanoviD, StrickerS, et al. (2009) Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell 4: 568–580.

37. LeeJ, KotliarovaS, KotliarovY, LiA, SuQ, et al. (2006) Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 9: 391–403.

38. IshiiN, MaierD, MerloA, TadaM, SawamuraY, et al. (1999) Frequent co-alterations of TP53, p16/CDKN2A, p14ARF, PTEN tumor suppressor genes in human glioma cell lines. Brain Pathol 9: 469–479.

39. CiesielskiMJ, FenstermakerRA (2000) Oncogenic epidermal growth factor receptor mutants with tandem duplication: gene structure and effects on receptor function. Oncogene 19: 810–820.

40. GalliaGL, TylerBM, HannCL, SiuIM, GirandaVL, et al. (2009) Inhibition of Akt inhibits growth of glioblastoma and glioblastoma stem-like cells. Mol Cancer Ther 8: 386–393.

41. PanditaA, AldapeKD, ZadehG, GuhaA, JamesCD (2004) Contrasting in vivo and in vitro fates of glioblastoma cell subpopulations with amplified EGFR. Gene Chromosome Canc 39: 29–36.

42. IndaMD, BonaviaR, MukasaA, NaritaY, SahDW, et al. (2010) Tumor heterogeneity is an active process maintained by a mutant EGFR-induced cytokine circuit in glioblastoma. Genes Dev 24: 1731–1745.

43. SzerlipNJ, PedrazaA, ChakravartyD, AzimM, McGuireJ, et al. (2012) Intratumoral heterogeneity of receptor tyrosine kinases EGFR and PDGFRA amplification in glioblastoma defines subpopulations with distinct growth factor response. Proc Natl Acad Sci U S A 109: 3041–3046.

44. ChenAJ, PaikJH, ZhangH, ShuklaSA, MortensenR, et al. (2012) STAR RNA-binding protein Quaking suppresses cancer via stabilization of specific miRNA. Genes Dev 26: 1459–1472.

45. YingH, ZhengH, ScottK, WiedemeyerR, YanH, et al. (2010) Mig-6 controls EGFR trafficking and suppresses gliomagenesis. Proc Natl Acad Sci U S A 107: 6912–6917.

46. GuertinDA, StevensDM, ThoreenCC, BurdsAA, KalaanyNY, et al. (2006) Ablation in mice of the mTORC components raptor, rictor, or mLST8 reveals that mTORC2 is required for signaling to Akt-FOXO and PKCalpha, but not S6K1. Dev Cell 11: 859–871.

47. SunayamaJ, SatoA, MatsudaK, TachibanaK, WatanabeE, et al. (2011) FoxO3a functions as a key integrator of cellular signals that control glioblastoma stem-like cell differentiation and tumorigenicity. Stem Cells 29: 1327–1337.

48. TanakaK, BabicI, NathansonD, AkhavanD, GuoD, et al. (2011) Oncogenic EGFR signaling activates an mTORC2-NF-kappaB pathway that promotes chemotherapy resistance. Cancer Discov 1: 524–538.

49. BashirT, CloningerC, ArtinianN, AndersonL, BernathA, et al. (2012) Conditional astroglial rictor overexpression induces malignant glioma in mice. PLoS ONE 7: e47741 doi:10.1371/journal.pone.0047741.

50. FurnariFB, LinH, HuangHS, CaveneeWK (1997) Growth suppression of glioma cells by PTEN requires a functional phosphatase catalytic domain. Proc Natl Acad Sci U S A 94: 12479–12484.

51. VanrobaysE, GleizesPE, Bousquet-AntonelliC, Noaillac-DepeyreJ, Caizergues-FerrerM, et al. (2001) Processing of 20S pre-rRNA to 18S ribosomal RNA in yeast requires Rrp10p, an essential non-ribosomal cytoplasmic protein. EMBO J 20: 4204–4213.

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

53. NarlaA, EbertBL (2010) Ribosomopathies: human disorders of ribosome dysfunction. Blood 115: 3196–3205.

54. SimmonsML, LambornKR, TakahashiM, ChenP, IsraelMA, et al. (2001) Analysis of complex relationships between age, p53, epidermal growth factor receptor, and survival in glioblastoma patients. Cancer Res 61: 1122–1128.

55. McLendonR, FriedmanA, BignerD, Van MeirEG, BratDJ, et al. (2008) Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455: 1061–1068.

56. OhgakiH, DessenP, JourdeB, HorstmannS, NishikawaT, et al. (2004) Genetic pathways to glioblastoma: a population-based study. Cancer Res 64: 6892–6899.

57. LoweSW, RuleyHE, JacksT, HousmanDE (1993) p53-dependent apoptosis modulates the cytotoxicity of anticancer agents. Cell 74: 957–967.

58. StuppR, MasonWP, van den BentMJ, WellerM, FisherB, et al. (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352: 987–996.

59. Villalonga-PlanellsR, Coll-MuletL, Martinez-SolerF, CastanoE, AcebesJJ, et al. (2011) Activation of p53 by nutlin-3a induces apoptosis and cellular senescence in human glioblastoma multiforme. PLoS ONE 6: e18588 doi:10.1371/journal.pone.0018588.

60. ZinzallaV, StrackaD, OppligerW, HallMN (2011) Activation of mTORC2 by association with the ribosome. Cell 144: 757–768.

61. LiuT, DengM, LiJ, TongX, WeiQ, et al. (2011) Phosphorylation of right open reading frame 2 (Rio2) protein kinase by polo-like kinase 1 regulates mitotic progression. J Biol Chem 286: 36352–36360.

62. MarygoldSJ, RooteJ, ReuterG, LambertssonA, AshburnerM, et al. (2007) The ribosomal protein genes and Minute loci of Drosophila melanogaster. Genome Biol 8: R216.

63. MaciasE, JinA, DeisenrothC, BhatK, MaoH, et al. (2010) An ARF-independent c-MYC-activated tumor suppression pathway mediated by ribosomal protein-Mdm2 Interaction. Cancer Cell 18: 231–243.

64. BurgerK, MuhlB, HarasimT, RohrmoserM, MalamoussiA, et al. (2010) Chemotherapeutic drugs inhibit ribosome biogenesis at various levels. J Biol Chem 285: 12416–12425.

65. SunXX, DaiMS, LuH (2007) 5-fluorouracil activation of p53 involves an MDM2-ribosomal protein interaction. J Biol Chem 282: 8052–8059.

66. GoidtsV, BageritzJ, PuccioL, NakataS, ZapatkaM, et al. (2012) RNAi screening in glioma stem-like cells identifies PFKFB4 as a key molecule important for cancer cell survival. Oncogene 31: 3235–3243.

67. WurdakH, ZhuS, RomeroA, LorgerM, WatsonJ, et al. (2010) An RNAi screen identifies TRRAP as a regulator of brain tumor-initiating cell differentiation. Cell Stem Cell 6: 37–47.

68. WiedemeyerWR, DunnIF, QuayleSN, ZhangJ, ChhedaMG, et al. (2010) Pattern of retinoblastoma pathway inactivation dictates response to CDK4/6 inhibition in GBM. Proc Natl Acad Sci U S A 107: 11501–11506.

69. KimYW, LiuTJ, KoulD, TiaoN, FerozeAH, et al. (2011) Identification of novel synergistic targets for rational drug combinations with PI3 kinase inhibitors using siRNA synthetic lethality screening against GBM. Neuro Oncol 13: 367–375.

70. MellinghoffIK, WangMY, VivancoI, Haas-KoganDA, ZhuS, et al. (2005) Molecular determinants of the response of glioblastomas to EGFR kinase inhibitors. N Engl J Med 353: 2012–2024.

71. RaynaudFI, EcclesSA, PatelS, AlixS, BoxG, et al. (2009) Biological properties of potent inhibitors of class I phosphatidylinositide 3-kinases: from PI-103 through PI-540, PI-620 to the oral agent GDC-0941. Mol Cancer Ther 8: 1725–1738.

72. HiraiH, SootomeH, NakatsuruY, MiyamaK, TaguchiS, et al. (2010) MK-2206, an allosteric Akt inhibitor, enhances antitumor efficacy by standard chemotherapeutic agents or molecular targeted drugs in vitro and in vivo. Mol Cancer Ther 9: 1956–1967.

73. UhlenM, OksvoldP, FagerbergL, LundbergE, JonassonK, et al. (2010) Towards a knowledge-based Human Protein Atlas. Nat Biotechnol 28: 1248–1250.

74. ChooAY, YoonSO, KimSG, RouxPP, BlenisJ (2008) Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci U S A 105: 17414–17419.

75. ObenauerJC, CantleyLC, YaffeMB (2003) Scansite 2.0: Proteome-wide prediction of cell signaling interactions using short sequence motifs. Nucleic Acids Res 31: 3635–3641.

76. HornbeckPV, KornhauserJM, TkachevS, ZhangB, SkrzypekE, et al. (2011) PhosphoSitePlus: a comprehensive resource for investigating the structure and function of experimentally determined post-translational modifications in man and mouse. Nucleic Acids Res 40: D261–270.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 2
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#