#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The RNA-Binding Protein QKI Suppresses Cancer-Associated Aberrant Splicing


Alternative pre-mRNA splicing is a key mechanism for increasing proteomic diversity and modulating gene expression. Emerging evidence indicates that splicing program is frequently deregulated during tumorigenesis, and cancer cells favor to produce protein isoforms that can promote growth and survival. Lung cancer is one of the most common cancers and the leading cause of cancer-related death worldwide. Although a number of lung cancer-related splicing events have been detected in several genome-wide analyses, much less is known about how aberrant splicing takes place in lung cancer and how it contributes to tumor development. In this study, we characterized the RNA-binding protein QKI as a new critical regulator of alternative splicing in lung cancer and as a potential marker for prognosis. Genome-wide analysis of QKI-dependent splicing by RNA-Seq identified some cancer-associated splicing changes as its targets. Our results demonstrate that QKI-5 inhibits cancer cell proliferation and prevents inappropriate activation of the Notch signaling pathway by regulating its key target, NUMB. We further showed that QKI-5 represses the inclusion of NUMB alternative exon through competing with a core splicing factor SF1. In summary, our data indicate that down-regulation of QKI causes aberrant splicing in lung cancer and suggest a novel tumor suppression mechanism involving QKI-mediated repression of Notch signaling.


Vyšlo v časopise: The RNA-Binding Protein QKI Suppresses Cancer-Associated Aberrant Splicing. PLoS Genet 10(4): e32767. doi:10.1371/journal.pgen.1004289
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004289

Souhrn

Alternative pre-mRNA splicing is a key mechanism for increasing proteomic diversity and modulating gene expression. Emerging evidence indicates that splicing program is frequently deregulated during tumorigenesis, and cancer cells favor to produce protein isoforms that can promote growth and survival. Lung cancer is one of the most common cancers and the leading cause of cancer-related death worldwide. Although a number of lung cancer-related splicing events have been detected in several genome-wide analyses, much less is known about how aberrant splicing takes place in lung cancer and how it contributes to tumor development. In this study, we characterized the RNA-binding protein QKI as a new critical regulator of alternative splicing in lung cancer and as a potential marker for prognosis. Genome-wide analysis of QKI-dependent splicing by RNA-Seq identified some cancer-associated splicing changes as its targets. Our results demonstrate that QKI-5 inhibits cancer cell proliferation and prevents inappropriate activation of the Notch signaling pathway by regulating its key target, NUMB. We further showed that QKI-5 represses the inclusion of NUMB alternative exon through competing with a core splicing factor SF1. In summary, our data indicate that down-regulation of QKI causes aberrant splicing in lung cancer and suggest a novel tumor suppression mechanism involving QKI-mediated repression of Notch signaling.


Zdroje

1. SiegelR, NaishadhamD, JemalA (2012) Cancer statistics, 2012. CA Cancer J Clin 62: 10–29.

2. YouldenDR, CrambSM, BaadePD (2008) The international epidemiology of lung cancer: geographical distribution and secular trends. J Thorac Oncol 3: 819–831.

3. NilsenTW, GraveleyBR (2010) Expansion of the eukaryotic proteome by alternative splicing. Nature 463: 457–463.

4. CooperTA, WanL, DreyfussG (2009) RNA and disease. Cell 136: 777–793.

5. VenablesJP (2006) Unbalanced alternative splicing and its significance in cancer. Bioessays 28: 378–386.

6. DavidCJ, ManleyJL (2010) Alternative pre-mRNA splicing regulation in cancer: pathways and programs unhinged. Genes Dev 24: 2343–2364.

7. BlackDL (2003) Mechanisms of alternative pre-messenger RNA splicing. Annu Rev Biochem 72: 291–336.

8. MatlinAJ, ClarkF, SmithCW (2005) Understanding alternative splicing: towards a cellular code. Nat Rev Mol Cell Biol 6: 386–398.

9. ChenM, ManleyJL (2009) Mechanisms of alternative splicing regulation: insights from molecular and genomics approaches. Nat Rev Mol Cell Biol 10: 741–754.

10. KarniR, de StanchinaE, LoweSW, SinhaR, MuD, et al. (2007) The gene encoding the splicing factor SF2/ASF is a proto-oncogene. Nat Struct Mol Biol 14: 185–193.

11. AnczukówO, RosenbergAZ, AkermanM, DasS, ZhanL, et al. (2012) The splicing factor SRSF1 regulates apoptosis and proliferation to promote mammary epithelial cell transformation. Nat Struct Mol Biol 19: 220–228.

12. VenablesJP, KlinckR, KohC, Gervais-BirdJ, BramardA, et al. (2009) Cancer-associated regulation of alternative splicing. Nat Struct Mol Biol 16: 670–676.

13. LefaveCV, SquatritoM, VorlovaS, RoccoGL, BrennanCW, et al. (2011) Splicing factor hnRNPH drives an oncogenic splicing switch in gliomas. EMBO J 30: 4084–4097.

14. DavidCJ, ChenM, AssanahM, CanollP, ManleyJL (2010) HnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature 463: 364–368.

15. GrossoAR, MartinsS, Carmo-FonsecaM (2008) The emerging role of splicing factors in cancer. EMBO Rep 9: 1087–1093.

16. PioR, MontuengaLM (2009) Alternative splicing in lung cancer. J Thorac Oncol 4: 674–678.

17. XiL, FeberA, GuptaV, WuM, BergemannAD, et al. (2008) Whole genome exon arrays identify differential expression of alternatively spliced, cancer-related genes in lung cancer. Nucleic Acids Res 36: 6535–6547.

18. PioR, BlancoD, PajaresMJ, AibarE, DuranyO, et al. (2010) Development of a novel splice array platform and its application in the identification of alternative splice variants in lung cancer. BMC Genomics 11: 352.

19. LangerW, SohlerF, LederG, BeckmannG, SeidelH, et al. (2010) Exon array analysis using re-defined probe sets results in reliable identification of alternatively spliced genes in non-small cell lung cancer. BMC Genomics 11: 676.

20. Misquitta-AliCM, ChengE, O'HanlonD, LiuN, McGladeCJ, et al. (2011) Global profiling and molecular characterization of alternative splicing events misregulated in lung cancer. Mol Cell Biol 31: 138–150.

21. SeoJS, JuYS, LeeWC, ShinJY, LeeJK, et al. (2012) The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Res 22: 2109–2119.

22. SuLJ, ChangCW, WuYC, ChenKC, LinCJ, et al. (2007) Selection of DDX5 as a novel internal control for Q-RT-PCR from microarray data using a block bootstrap re-sampling scheme. BMC Genomics 8: 140.

23. LandiMT, DrachevaT, RotunnoM, FigueroaJD, LiuH, et al. (2008) Gene expression signature of cigarette smoking and its role in lung adenocarcinoma development and survival. PLoS One 3: e1651.

24. WrageM, RuosaariS, EijkPP, KaifiJT, HollménJ, et al. (2009) Genomic profiles associated with early micrometastasis in lung cancer: relevance of 4q deletion. Clin Cancer Res 15: 1566–1574.

25. FengY, BankstonA (2010) The star family member QKI and cell signaling. Adv Exp Med Biol 693: 25–36.

26. WuJ, ZhouL, TonissenK, TeeR, ArtztK (1999) The quaking I-5 protein (QKI-5) has a novel nuclear localization signal and shuttles between the nucleus and the cytoplasm. J Biol Chem 274: 29202–29210.

27. HardyRJ, LoushinCL, FriedrichVLJr, ChenQ, EbersoleTA, et al. (1996) Neural cell type-specific expression of QKI proteins is altered in quakingviable mutant mice. J Neurosci 16: 7941–7949.

28. EbersoleTA, ChenQ, JusticeMJ, ArtztK (1996) The quaking gene product necessary in embryogenesis and myelination combines features of RNA binding and signal transduction proteins. Nat Genet 12: 260–265.

29. KondoT, FurutaT, MitsunagaK, EbersoleTA, ShichiriM, et al. (1999) Genomic organization and expression analysis of the mouse qkI locus. Mamm Genome 10: 662–669.

30. WuJI, ReedRB, GrabowskiPJ, ArtztK (2002) Function of quaking in myelination: regulation of alternative splicing. Proc Natl Acad Sci USA 99: 4233–4238.

31. NovikovL, ParkJW, ChenH, KlermanH, JallohAS, et al. (2011) QKI-mediated alternative splicing of the histone variant MacroH2A1 regulates cancer cell proliferation. Mol Cell Biol 31: 4244–4255.

32. HallMP, NagelRJ, FaggWS, ShiueL, ClineMS, et al. (2013) Quaking and PTB control overlapping splicing regulatory networks during muscle cell differentiation. RNA 19: 627–638.

33. van der VeerEP, de BruinRG, KraaijeveldAO, de VriesMR, BotI, et al. (2013) Quaking, an RNA-binding protein, is a critical regulator of vascular smooth muscle cell phenotype. Circ Res 113: 1065–1075.

34. LarocqueD, PilotteJ, ChenT, CloutierF, MassieB, et al. (2002) Nuclear retention of MBP mRNAs in the quaking viable mice. Neuron 36: 815–829.

35. LiZ, ZhangY, LiD, FengY (2000) Destabilization and mislocalization of myelin basic protein mRNAs in quaking dysmyelination lacking the QKI RNA-binding proteins. J Neurosci 20: 4944–4953.

36. LarocqueD, GalarneauA, LiuHN, ScottM, AlmazanG, et al. (2005) Protection of p27(Kip1) mRNA by quaking RNA binding proteins promotes oligodendrocyte differentiation. Nat Neurosci 8: 27–33.

37. ZhaoL, KuL, ChenY, XiaM, LoPrestiP, et al. (2006) QKI binds MAP1B mRNA and enhances MAP1B expression during oligodendrocyte development. Mol Biol Cell 17: 4179–4186.

38. DoukhanineE, GavinoC, HainesJD, AlmazanG, RichardS (2010) The QKI-6 RNA binding protein regulates actin-interacting protein-1 mRNA stability during oligodendrocyte differentiation. Mol Biol Cell 21: 3029–3040.

39. ZearfossNR, ClingmanCC, FarleyBM, McCoigLM, RyderSP (2011) Quaking regulates Hnrnpa1 expression through its 3′ UTR in oligodendrocyte precursor cells. PLoS Genet 7: e1001269.

40. 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.

41. SaccomannoL, LoushinC, JanE, PunkayE, ArtztK, et al. (1999) The STAR protein QKI-6 is a translational repressor. Proc Natl Acad Sci U S A 96: 12605–12610.

42. ZhaoL, MandlerMD, YiH, FengY (2010) Quaking I controls a unique cytoplasmic pathway that regulates alternative splicing of myelin-associated glycoprotein. Proc Natl Acad Sci USA 107: 19061–19066.

43. WangY, VogelG, YuZ, RichardS (2013) The QKI-5 and QKI-6 RNA binding proteins regulate the expression of microRNA 7 in glial cells. Mol Cell Biol 33: 1233–1243.

44. RyderSP, WilliamsonJR (2004) Specificity of the STAR/GSG domain protein Qk1: implications for the regulation of myelination. RNA 10: 1449–1458.

45. GalarneauA, RichardS (2005) Target RNA motif and target mRNAs of the Quaking STAR protein. Nat Struct Mol Biol 12: 691–698.

46. HafnerM, LandthalerM, BurgerL, KhorshidM, HausserJ, et al. (2010) Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell 141: 129–141.

47. GyőrffyB, SurowiakP, BudcziesJ, LánczkyA (2013) Online survival analysis software to assess the prognostic value of biomarkers using transcriptomic data in non-small-cell lung cancer. PLoS One 8: e82241.

48. GuoM, JanLY, JanYN (2007) Control of daughter cell fates during asymmetric division: interaction of Numb and Notch. Neuron 17: 27–41.

49. McGillMA, McGladeCJ (2003) Mammalian numb proteins promote Notch1 receptor ubiquitination and degradation of the Notch1 intracellular domain. J Biol Chem 278: 23196–23203.

50. McGillMA, DhoSE, WeinmasterG, McGladeCJ (2009) Numb regulates post-endocytic trafficking and degradation of Notch1. J Biol Chem 284: 26427–26438.

51. DhoSE, FrenchMB, WoodsSA, McGladeCJ (1999) Characterization of four mammalian numb protein isoforms. Identification of cytoplasmic and membrane-associated variants of the phosphotyrosine binding domain. J Biol Chem 274: 33097–33104.

52. RandMD, GrimmLM, Artavanis-TsakonasS, PatriubV, BlacklowSC, et al. (2000) Calcium depletion dissociates and activates heterodimeric notch receptors. Mol Cell Biol 20: 1825–1835.

53. CorioniM, AntihN, TanackovicG, ZavolanM, KrämerA (2011) Analysis of in situ pre-mRNA targets of human splicing factor SF1 reveals a function in alternative splicing. Nucleic Acids Res 39: 1868–1879.

54. GaurRK, ValcárcelJ, GreenMR (1995) Sequential recognition of the pre-mRNA branch point by U2AF65 and a novel spliceosome-associated 28-kDa protein. RNA 1: 407–417.

55. RoyM, PearWS, AsterJC (2007) The multifaceted role of Notch in cancer. Curr Opin Genet Dev 17: 52–59.

56. RanganathanP, WeaverKL, CapobiancoAJ (2011) Notch signalling in solid tumours: a little bit of everything but not all the time. Nat Rev Cancer 11: 338–351.

57. PeceS, SerresiM, SantoliniE, CapraM, HullemanE, et al. (2004) Loss of negative regulation by Numb over Notch is relevant to human breast carcinogenesis. J Cell Biol 167: 215–221.

58. WesthoffB, ColalucaIN, D'ArioG, DonzelliM, TosoniD, et al. (2009) Alterations of the Notch pathway in lung cancer. Proc Natl Acad Sci USA 106: 22293–22298.

59. VerdiJM, BashirullahA, GoldhawkDE, KubuCJ, JamaliM, et al. (1999) Distinct human NUMB isoforms regulate differentiation vs. proliferation in the neuronal lineage. Proc Natl Acad Sci USA 96: 10472–10476.

60. ToriyaM, TokunagaA, SawamotoK, NakaoK, OkanoH (2006) Distinct functions of human numb isoforms revealed by misexpression in the neural stem cell lineage in the Drosophila larval brain. Dev Neurosci 28: 142–155.

61. Bani-YaghoubM, KubuCJ, CowlingR, RochiraJ, NikopoulosGN, et al. (2007) A switch in numb isoforms is a critical step in cortical development. Dev Dyn 236: 696–705.

62. KimKK, NamJ, MukouyamaYS, KawamotoS (2013) Rbfox3-regulated alternative splicing of Numb promotes neuronal differentiation during development. J Cell Biol 200: 443–458.

63. UleJ, StefaniG, MeleA, RuggiuM, WangX, et al. (2006) An RNA map predicting Nova-dependent splicing regulation. Nature 444: 580–586.

64. YeoGW, CoufalNG, LiangTY, PengGE, FuXD, et al. (2009) An RNA code for the FOX2 splicing regulator revealed by mapping RNA-protein interactions in stem cells. Nat Struct Mol Biol 16: 130–137.

65. ZhangC, ZhangZ, CastleJ, SunS, JohnsonJ, et al. (2008) Defining the regulatory network of the tissue-specific splicing factors Fox-1 and Fox-2. Genes Dev 22: 2550–2563.

66. DuH, ClineMS, OsborneRJ, TuttleDL, ClarkTA, et al. (2010) Aberrant alternative splicing and extracellular matrix gene expression in mouse models of myotonic dystrophy. Nat Struct Mol Biol 17: 187–193.

67. TaggartAJ, DeSimoneAM, ShihJS, FillouxME, FairbrotherWG (2012) Large-scale mapping of branchpoints in human pre-mRNA transcripts in vivo. Nat Struct Mol Biol 19: 719–721.

68. YangG, FuH, ZhangJ, LuX, YuF, et al. (2010) RNA-binding protein quaking, a critical regulator of colon epithelial differentiation and a suppressor of colon cancer. Gastroenterology 138: 231–240.

69. GeK, DuHadawayJ, DuW, HerlynM, RodeckU, et al. (1999) Mechanism for elimination of a tumor suppressor: aberrant splicing of a brain-specific exon causes loss of function of Bin1 in melanoma. Proc Natl Acad Sci USA 96: 9689–9694.

70. WeiWJ, MuSR, HeinerM, FuX, CaoLJ, et al. (2012) YB-1 binds to CAUC motifs and stimulates exon inclusion by enhancing the recruitment of U2AF to weak polypyrimidine tracts. Nucleic Acids Res 40: 8622–8636.

71. BrooksAN, YangL, DuffMO, HansenKD, ParkJW, et al. (2011) Conservation of an RNA regulatory map between Drosophila and mammals. Genome Res 21: 193–202.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 4
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#