#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

An Evolutionarily Conserved Synthetic Lethal Interaction Network Identifies FEN1 as a Broad-Spectrum Target for Anticancer Therapeutic Development


Harnessing genetic differences between cancerous and noncancerous cells offers a strategy for the development of new therapies. Extrapolating from yeast genetic interaction data, we used cultured human cells and siRNA to construct and evaluate a synthetic lethal interaction network comprised of chromosome instability (CIN) genes that are frequently mutated in colorectal cancer. A small number of genes in this network were found to have synthetic lethal interactions with a large number of cancer CIN genes; these genes are thus attractive targets for anticancer therapeutic development. The protein product of one highly connected gene, the flap endonuclease FEN1, was used as a target for small-molecule inhibitor screening using a newly developed fluorescence-based assay for enzyme activity. Thirteen initial hits identified through in vitro biochemical screening were tested in cells, and it was found that two compounds could selectively inhibit the proliferation of cultured cancer cells carrying inactivating mutations in CDC4, a gene frequently mutated in a variety of cancers. Inhibition of flap endonuclease activity was also found to recapitulate a genetic interaction between FEN1 and MRE11A, another gene frequently mutated in colorectal cancers, and to lead to increased endogenous DNA damage. These chemical-genetic interactions in mammalian cells validate evolutionarily conserved synthetic lethal interactions and demonstrate that a cross-species candidate gene approach is successful in identifying small-molecule inhibitors that prove effective in a cell-based cancer model.


Vyšlo v časopise: An Evolutionarily Conserved Synthetic Lethal Interaction Network Identifies FEN1 as a Broad-Spectrum Target for Anticancer Therapeutic Development. PLoS Genet 9(1): e32767. doi:10.1371/journal.pgen.1003254
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003254

Souhrn

Harnessing genetic differences between cancerous and noncancerous cells offers a strategy for the development of new therapies. Extrapolating from yeast genetic interaction data, we used cultured human cells and siRNA to construct and evaluate a synthetic lethal interaction network comprised of chromosome instability (CIN) genes that are frequently mutated in colorectal cancer. A small number of genes in this network were found to have synthetic lethal interactions with a large number of cancer CIN genes; these genes are thus attractive targets for anticancer therapeutic development. The protein product of one highly connected gene, the flap endonuclease FEN1, was used as a target for small-molecule inhibitor screening using a newly developed fluorescence-based assay for enzyme activity. Thirteen initial hits identified through in vitro biochemical screening were tested in cells, and it was found that two compounds could selectively inhibit the proliferation of cultured cancer cells carrying inactivating mutations in CDC4, a gene frequently mutated in a variety of cancers. Inhibition of flap endonuclease activity was also found to recapitulate a genetic interaction between FEN1 and MRE11A, another gene frequently mutated in colorectal cancers, and to lead to increased endogenous DNA damage. These chemical-genetic interactions in mammalian cells validate evolutionarily conserved synthetic lethal interactions and demonstrate that a cross-species candidate gene approach is successful in identifying small-molecule inhibitors that prove effective in a cell-based cancer model.


Zdroje

1. HartwellLH, SzankasiP, RobertsCJ, MurrayAW, FriendSH (1997) Integrating genetic approaches into the discovery of anticancer drugs. Science 278: 1064–1068 10.1126/science.278.5340.1064.

2. FarmerH, McCabeN, LordCJ, TuttAN, JohnsonDA, et al. (2005) Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 434: 917–921.

3. BryantHE, SchultzN, ThomasHD, ParkerKM, FlowerD, et al. (2005) Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 434: 913–917.

4. WeinsteinJN, MyersTG, O'ConnorPM, FriendSH, FornaceAJJr, et al. (1997) An information-intensive approach to the molecular pharmacology of cancer. Science 275: 343–349.

5. BarretinaJ, CaponigroG, StranskyN, VenkatesanK, MargolinAA, et al. (2012) The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 483: 603–607.

6. GarnettMJ, EdelmanEJ, HeidornSJ, GreenmanCD, DasturA, et al. (2012) Systematic identification of genomic markers of drug sensitivity in cancer cells. Nature 483: 570–575.

7. GrebienF, HantschelO, WojcikJ, KaupeI, KovacicB, et al. (2011) Targeting the SH2-kinase interface in Bcr-Abl inhibits leukemogenesis. Cell 147: 306–319.

8. ZhaoC, ChenA, JamiesonCH, FereshtehM, AbrahamssonA, et al. (2009) Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia. Nature 458: 776–779.

9. HanahanD, WeinbergRA (2011) Hallmarks of cancer: the next generation. Cell 144: 646–674.

10. WeaverBA, ClevelandDW (2006) Does aneuploidy cause cancer? Curr Opin Cell Biol 18: 658–667.

11. StirlingPC, BloomMS, Solanki-PatilT, SmithS, SipahimalaniP, et al. (2011) The complete spectrum of yeast chromosome instability genes identifies candidate CIN cancer genes and functional roles for ASTRA complex components. PLoS Genet 7: e1002057 doi:10.1371/journal.pgen.1002057.

12. CahillDP, LengauerC, YuJ, RigginsGJ, WillsonJKV, et al. (1998) Mutations of mitotic checkpoint genes in human cancers. Nature 392: 300–303 10.1038/32688.

13. RajagopalanH, JallepalliPV, RagoC, VelculescuVE, KinzlerKW, et al. (2004) Inactivation of hCDC4 can cause chromosomal instability. Nature 428: 77–81 10.1038/nature02313.

14. BarberTD, McManusK, YuenKW, ReisM, ParmigianiG, et al. (2008) Chromatid cohesion defects may underlie chromosome instability in human colorectal cancers. Proc Natl Acad Sci U S A 105: 3443–3448.

15. WangZ, CumminsJM, ShenD, CahillDP, JallepalliPV, et al. (2004) Three classes of genes mutated in colorectal cancers with chromosomal instability. Cancer Res 64: 2998–3001.

16. TongAH, EvangelistaM, ParsonsAB, XuH, BaderGD, et al. (2001) Systematic genetic analysis with ordered arrays of yeast deletion mutants. Science 294: 2364–2368.

17. YuenKW, WarrenCD, ChenO, KwokT, HieterP, et al. (2007) Systematic genome instability screens in yeast and their potential relevance to cancer. Proc Natl Acad Sci U S A 104: 3925–3930.

18. McLellanJ, O'NeilN, TarailoS, StoepelJ, BryanJ, et al. (2009) Synthetic lethal genetic interactions that decrease somatic cell proliferation in Caenorhabditis elegans identify the alternative RFC CTF18 as a candidate cancer drug target. Mol Biol Cell 20: 5306–5313.

19. McLellanJL, O'NeilNJ, BarrettI, FerreeE, van PelDM, et al. (2012) Synthetic lethality of cohesins with PARPs and replication fork mediators. PLoS Genet 8: e1002574 doi:10.1371/journal.pgen.1002574.

20. HiramotoT, NakanishiT, SumiyoshiT, FukudaT, MatsuuraS, et al. (1999) Mutations of a novel human RAD54 homologue, RAD54B, in primary cancer. Oncogene 18: 3422–3426.

21. KempZ, RowanA, ChambersW, WorthamN, HalfordS, et al. (2005) CDC4 mutations occur in a subset of colorectal cancers but are not predicted to cause loss of function and are not associated with chromosomal instability. Cancer Res 65: 11361–11366.

22. CahillDP, KinzlerKW, VogelsteinB, LengauerC (1999) Genetic instability and darwinian selection in tumours. Trends Cell Biol 9: M57–M60 10.1016/S0962-8924(99)01661-X.

23. CostanzoM, BaryshnikovaA, BellayJ, KimY, SpearED, et al. (2010) The genetic landscape of a cell. Science 327: 425–431.

24. PrietoI, SujaJA, PezziN, KremerL, MartinezA, et al. (2001) Mammalian STAG3 is a cohesin specific to sister chromatid arms in meiosis I. Nat Cell Biol 3: 761–766.

25. LiuY, BambaraRA (2003) Analysis of human flap endonuclease 1 mutants reveals a mechanism to prevent triplet repeat expansion. J Biol Chem 278: 13728–13739.

26. ZhengL, JiaJ, FingerLD, GuoZ, ZerC, et al. (2011) Functional regulation of FEN1 nuclease and its link to cancer. Nucleic Acids Res 39: 781–794.

27. TumeyLN, BomD, HuckB, GleasonE, WangJ, et al. (2005) The identification and optimization of a N-hydroxy urea series of flap endonuclease 1 inhibitors. Bioorg Med Chem Lett 15: 277–281.

28. LipinskiCA, LombardoF, DominyBW, FeeneyPJ (2001) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 46: 3–26.

29. AkhoondiS, SunD, von derLN, ApostolidouS, KlotzK, et al. (2007) FBXW7/hCDC4 is a general tumor suppressor in human cancer. Cancer Res 67: 9006–9012.

30. KohJL, DingH, CostanzoM, BaryshnikovaA, ToufighiK, et al. (2010) DRYGIN: a database of quantitative genetic interaction networks in yeast. Nucleic Acids Res 38: D502–D507.

31. MiyakiM, YamaguchiT, IijimaT, TakahashiK, MatsumotoH, et al. (2009) Somatic mutations of the CDC4 (FBXW7) gene in hereditary colorectal tumors. Oncology 76: 430–434.

32. MilneAN, LeguitR, CorverWE, MorsinkFH, PolakM, et al. (2010) Loss of CDC4/FBXW7 in gastric carcinoma. Cell Oncol 32: 347–359.

33. DupreA, Boyer-ChatenetL, SattlerRM, ModiAP, LeeJH, et al. (2008) A forward chemical genetic screen reveals an inhibitor of the Mre11-Rad50-Nbs1 complex. Nat Chem Biol 4: 119–125.

34. AndersonL, HendersonC, AdachiY (2001) Phosphorylation and rapid relocalization of 53BP1 to nuclear foci upon DNA damage. Mol Cell Biol 21: 1719–1729.

35. RappoldI, IwabuchiK, DateT, ChenJ (2001) Tumor suppressor p53 binding protein 1 (53BP1) is involved in DNA damage-signaling pathways. J Cell Biol 153: 613–620.

36. MarkovaE, SchultzN, BelyaevIY (2007) Kinetics and dose-response of residual 53BP1/gamma-H2AX foci: co-localization, relationship with DSB repair and clonogenic survival. Int J Radiat Biol 83: 319–329.

37. LazebnikYA, KaufmannSH, DesnoyersS, PoirierGG, EarnshawWC (1994) Cleavage of poly(ADP-ribose) polymerase by a proteinase with properties like ICE. Nature 371: 346–347.

38. Mendes-PereiraAM, MartinSA, BroughR, McCarthyA, TaylorJR, et al. (2009) Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO Mol Med 1: 315–322.

39. ReaperPM, GriffithsMR, LongJM, CharrierJD, MaccormickS, et al. (2011) Selective killing of ATM- or p53-deficient cancer cells through inhibition of ATR. Nat Chem Biol

40. LengauerC, KinzlerKW, VogelsteinB (1997) Genetic instability in colorectal cancers. Nature 386: 623–627 10.1038/386623a0.

41. FengZ, ScottSP, BussenW, SharmaGG, GuoG, et al. (2011) Rad52 inactivation is synthetically lethal with BRCA2 deficiency. Proc Natl Acad Sci U S A 108: 686–691.

42. MartinSA, McCabeN, MullarkeyM, CumminsR, BurgessDJ, et al. (2010) DNA polymerases as potential therapeutic targets for cancers deficient in the DNA mismatch repair proteins MSH2 or MLH1. Cancer Cell 17: 235–248.

43. PanX, YuanDS, XiangD, WangX, Sookhai-MahadeoS, et al. (2004) A robust toolkit for functional profiling of the yeast genome. Mol Cell 16: 487–496 10.1016/j.molcel.2004.09.035.

44. LePF, LillicoS, PassetB, YoungR, WhitelawB, et al. (2010) Zinc finger nuclease technology heralds a new era in mammalian transgenesis. Trends Biotechnol 28: 134–141.

45. CermakT, DoyleEL, ChristianM, WangL, ZhangY, et al. (2011) Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res 39: e82.

46. TarailoM, TarailoS, RoseAM (2007) Synthetic lethal interactions identify phenotypic “interologs” of the spindle assembly checkpoint components. Genetics 177: 2525–2530.

47. McManusKJ, BarrettIJ, NouhiY, HieterP (2009) Specific synthetic lethal killing of RAD54B-deficient human colorectal cancer cells by FEN1 silencing. Proc Natl Acad Sci U S A 106: 3276–3281.

48. MuranteRS, RumbaughJA, BarnesCJ, NortonJR, BambaraRA (1996) Calf RTH-1 nuclease can remove the initiator RNAs of Okazaki fragments by endonuclease activity. J Biol Chem 271: 25888–25897.

49. HarringtonJJ, LieberMR (1994) The characterization of a mammalian DNA structure-specific endonuclease. EMBO J 13: 1235–1246.

50. KimK, BiadeS, MatsumotoY (1998) Involvement of flap endonuclease 1 in base excision DNA repair. J Biol Chem 273: 8842–8848.

51. KlunglandA, LindahlT (1997) Second pathway for completion of human DNA base excision-repair: reconstitution with purified proteins and requirement for DNase IV (FEN1). EMBO J 16: 3341–3348.

52. SinghP, ZhengL, ChavezV, QiuJ, ShenB (2007) Concerted action of exonuclease and Gap-dependent endonuclease activities of FEN-1 contributes to the resolution of triplet repeat sequences (CTG)n- and (GAA)n-derived secondary structures formed during maturation of Okazaki fragments. J Biol Chem 282: 3465–3477.

53. ZhengL, ZhouM, ChaiQ, ParrishJ, XueD, et al. (2005) Novel function of the flap endonuclease 1 complex in processing stalled DNA replication forks. EMBO Rep 6: 83–89.

54. JaiswalAS, BanerjeeS, AnejaR, SarkarFH, OstrovDA, et al. (2011) DNA Polymerase beta as a Novel Target for Chemotherapeutic Intervention of Colorectal Cancer. PLoS ONE 6: e16691 doi:10.1371/journal.pone.0016691.

55. TongAH, LesageG, BaderGD, DingH, XuH, et al. (2004) Global mapping of the yeast genetic interaction network. Science 303: 808–813 10.1126/science.1091317.

56. StromCE, JohanssonF, UhlenM, SzigyartoCA, ErixonK, et al. (2011) Poly (ADP-ribose) polymerase (PARP) is not involved in base excision repair but PARP inhibition traps a single-strand intermediate. Nucleic Acids Res 39: 3166–3175.

57. YingS, HamdyFC, HelledayT (2012) Mre11-dependent degradation of stalled DNA replication forks is prevented by BRCA2 and PARP1. Cancer Res 72: 2814–2821.

58. BirkbakNJ, EklundAC, LiQ, McClellandSE, EndesfelderD, et al. (2011) Paradoxical relationship between chromosomal instability and survival outcome in cancer. Cancer Res 71: 3447–3452.

59. LeeAJ, EndesfelderD, RowanAJ, WaltherA, BirkbakNJ, et al. (2011) Chromosomal instability confers intrinsic multidrug resistance. Cancer Res 71: 1858–1870.

60. ZhengL, DaiH, HegdeML, ZhouM, GuoZ, et al. (2011) Fen1 mutations that specifically disrupt its interaction with PCNA cause aneuploidy-associated cancer. Cell Res

61. DuxinJP, MooreHR, SidorovaJ, KaranjaK, HonakerY, et al. (2012) An Okazaki fragment processing-independent role for human Dna2 during DNA replication. J Biol Chem

62. StrackerTH, PetriniJH (2011) The MRE11 complex: starting from the ends. Nat Rev Mol Cell Biol 12: 90–103.

63. DavisH, TomlinsonI (2012) CDC4/FBXW7 and the ‘just enough’ model of tumourigenesis. J Pathol 227: 131–135.

64. LeGM, O'HaraAJ, RuddML, UrickME, HansenNF, et al. (2012) Exome sequencing of serous endometrial tumors identifies recurrent somatic mutations in chromatin-remodeling and ubiquitin ligase complex genes. Nat Genet 10.

65. HoonS, SmithAM, WallaceIM, SureshS, MirandaM, et al. (2008) An integrated platform of genomic assays reveals small-molecule bioactivities. Nat Chem Biol 4: 498–506.

66. CoganN, BairdDM, PhillipsR, CromptonLA, CaldwellMA, et al. (2010) DNA damaging bystander signalling from stem cells, cancer cells and fibroblasts after Cr(VI) exposure and its dependence on telomerase. Mutat Res 683: 1–8.

67. BaryshnikovaA, CostanzoM, DixonS, VizeacoumarFJ, MyersCL, et al. (2010) Synthetic genetic array (SGA) analysis in Saccharomyces cerevisiae and Schizosaccharomyces pombe. Methods Enzymol 470: 145–79 Epub;%2010 Mar 1.: 145–179.

68. BreslowDK, CameronDM, CollinsSR, SchuldinerM, Stewart-OrnsteinJ, et al. (2008) A comprehensive strategy enabling high-resolution functional analysis of the yeast genome. Nat Methods 5: 711–718.

69. LiZ, VizeacoumarFJ, BahrS, LiJ, WarringerJ, et al. (2011) Systematic exploration of essential yeast gene function with temperature-sensitive mutants. Nat Biotechnol 29: 361–367.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 1
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#