#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Ccdc94 Protects Cells from Ionizing Radiation by Inhibiting the Expression of


DNA double-strand breaks (DSBs) represent one of the most deleterious forms of DNA damage to a cell. In cancer therapy, induction of cell death by DNA DSBs by ionizing radiation (IR) and certain chemotherapies is thought to mediate the successful elimination of cancer cells. However, cancer cells often evolve to evade the cytotoxicity induced by DNA DSBs, thereby forming the basis for treatment resistance. As such, a better understanding of the DSB DNA damage response (DSB–DDR) pathway will facilitate the design of more effective strategies to overcome chemo- and radioresistance. To identify novel mechanisms that protect cells from the cytotoxic effects of DNA DSBs, we performed a forward genetic screen in zebrafish for recessive mutations that enhance the IR–induced apoptotic response. Here, we describe radiosensitizing mutation 7 (rs7), which causes a severe sensitivity of zebrafish embryonic neurons to IR–induced apoptosis and is required for the proper development of the central nervous system. The rs7 mutation disrupts the coding sequence of ccdc94, a highly conserved gene that has no previous links to the DSB–DDR pathway. We demonstrate that Ccdc94 is a functional member of the Prp19 complex and that genetic knockdown of core members of this complex causes increased sensitivity to IR–induced apoptosis. We further show that Ccdc94 and the Prp19 complex protect cells from IR–induced apoptosis by repressing the expression of p53 mRNA. In summary, we have identified a new gene regulating a dosage-sensitive response to DNA DSBs during embryonic development. Future studies in human cancer cells will determine whether pharmacological inactivation of CCDC94 reduces the threshold of the cancer cell apoptotic response.


Vyšlo v časopise: Ccdc94 Protects Cells from Ionizing Radiation by Inhibiting the Expression of. PLoS Genet 8(8): e32767. doi:10.1371/journal.pgen.1002922
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1002922

Souhrn

DNA double-strand breaks (DSBs) represent one of the most deleterious forms of DNA damage to a cell. In cancer therapy, induction of cell death by DNA DSBs by ionizing radiation (IR) and certain chemotherapies is thought to mediate the successful elimination of cancer cells. However, cancer cells often evolve to evade the cytotoxicity induced by DNA DSBs, thereby forming the basis for treatment resistance. As such, a better understanding of the DSB DNA damage response (DSB–DDR) pathway will facilitate the design of more effective strategies to overcome chemo- and radioresistance. To identify novel mechanisms that protect cells from the cytotoxic effects of DNA DSBs, we performed a forward genetic screen in zebrafish for recessive mutations that enhance the IR–induced apoptotic response. Here, we describe radiosensitizing mutation 7 (rs7), which causes a severe sensitivity of zebrafish embryonic neurons to IR–induced apoptosis and is required for the proper development of the central nervous system. The rs7 mutation disrupts the coding sequence of ccdc94, a highly conserved gene that has no previous links to the DSB–DDR pathway. We demonstrate that Ccdc94 is a functional member of the Prp19 complex and that genetic knockdown of core members of this complex causes increased sensitivity to IR–induced apoptosis. We further show that Ccdc94 and the Prp19 complex protect cells from IR–induced apoptosis by repressing the expression of p53 mRNA. In summary, we have identified a new gene regulating a dosage-sensitive response to DNA DSBs during embryonic development. Future studies in human cancer cells will determine whether pharmacological inactivation of CCDC94 reduces the threshold of the cancer cell apoptotic response.


Zdroje

1. LordCJ, AshworthA (2012) The DNA damage response and cancer therapy. Nature 481: 287–294.

2. GudkovAV, KomarovaEA (2003) The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer 3: 117–129.

3. KratzE, EimonPM, MukhyalaK, SternH, ZhaJ, et al. (2006) Functional characterization of the Bcl-2 gene family in the zebrafish. Cell Death Differ 13: 1631–1640.

4. SidiS, SandaT, KennedyRD, HagenAT, JetteCA, et al. (2008) Chk1 suppresses a caspase-2 apoptotic response to DNA damage that bypasses p53, Bcl-2, and caspase-3. Cell 133: 864–877.

5. VillungerA, MichalakEM, CoultasL, MullauerF, BockG, et al. (2003) p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science 302: 1036–1038.

6. GallenneT, GautierF, OliverL, HervouetE, NoelB, et al. (2009) Bax activation by the BH3-only protein Puma promotes cell dependence on antiapoptotic Bcl-2 family members. J Cell Biol 185: 279–290.

7. KirkinV, JoosS, ZornigM (2004) The role of Bcl-2 family members in tumorigenesis. Biochim Biophys Acta 1644: 229–249.

8. McKinnonPJ (2009) DNA repair deficiency and neurological disease. Nat Rev Neurosci 10: 100–112.

9. TaylorAM, HarndenDG, ArlettCF, HarcourtSA, LehmannAR, et al. (1975) Ataxia telangiectasia: a human mutation with abnormal radiation sensitivity. Nature 258: 427–429.

10. SavitskyK, Bar-ShiraA, GiladS, RotmanG, ZivY, et al. (1995) A single ataxia telangiectasia gene with a product similar to PI-3 kinase. Science 268: 1749–1753.

11. ShilohY (2003) ATM and related protein kinases: safeguarding genome integrity. Nat Rev Cancer 3: 155–168.

12. JetteCA, FlanaganAM, RyanJ, PyatiUJ, CarbonneauS, et al. (2008) BIM and other BCL-2 family proteins exhibit cross-species conservation of function between zebrafish and mammals. Cell Death Differ 15: 1063–1072.

13. JacksonSP, BartekJ (2009) The DNA-damage response in human biology and disease. Nature 461: 1071–1078.

14. LiuYC, ChenHC, WuNY, ChengSC (2007) A novel splicing factor, Yju2, is associated with NTC and acts after Prp2 in promoting the first catalytic reaction of pre-mRNA splicing. Mol Cell Biol 27: 5403–5413.

15. ChanSP, KaoDI, TsaiWY, ChengSC (2003) The Prp19p-associated complex in spliceosome activation. Science 302: 279–282.

16. ChengSC, TarnWY, TsaoTY, AbelsonJ (1993) PRP19: a novel spliceosomal component. Mol Cell Biol 13: 1876–1882.

17. LegerskiRJ (2009) The Pso4 complex splices into the DNA damage response. Cell Cycle 8: 3448–3449.

18. AbdelilahS, Mountcastle-ShahE, HarveyM, Solnica-KrezelL, SchierAF, et al. (1996) Mutations affecting neural survival in the zebrafish Danio rerio. Development 123: 217–227.

19. Furutani-SeikiM, JiangYJ, BrandM, HeisenbergCP, HouartC, et al. (1996) Neural degeneration mutants in the zebrafish, Danio rerio. Development 123: 229–239.

20. LiuTX, HowlettNG, DengM, LangenauDM, HsuK, et al. (2003) Knockdown of zebrafish Fancd2 causes developmental abnormalities via p53-dependent apoptosis. Dev Cell 5: 903–914.

21. BladenCL, NavarreS, DynanWS, KozlowskiDJ (2007) Expression of the Ku70 subunit (XRCC6) and protection from low dose ionizing radiation during zebrafish embryogenesis. Neurosci Lett 422: 97–102.

22. MiyashitaT, ReedJC (1992) bcl-2 gene transfer increases relative resistance of S49.1 and WEHI7.2 lymphoid cells to cell death and DNA fragmentation induced by glucocorticoids and multiple chemotherapeutic drugs. Cancer Res 52: 5407–5411.

23. MiyashitaT, ReedJC (1993) Bcl-2 oncoprotein blocks chemotherapy-induced apoptosis in a human leukemia cell line. Blood 81: 151–157.

24. SentmanCL, ShutterJR, HockenberyD, KanagawaO, KorsmeyerSJ (1991) bcl-2 inhibits multiple forms of apoptosis but not negative selection in thymocytes. Cell 67: 879–888.

25. RobuME, LarsonJD, NaseviciusA, BeiraghiS, BrennerC, et al. (2007) p53 activation by knockdown technologies. PLoS Genet 3: e78 doi:10.1371/journal.pgen.0030078.

26. BerghmansS, MurpheyRD, WienholdsE, NeubergD, KutokJL, et al. (2005) tp53 mutant zebrafish develop malignant peripheral nerve sheath tumors. Proc Natl Acad Sci U S A 102: 407–412.

27. BiswasAK, JohnsonDG (2012) Transcriptional and nontranscriptional functions of E2F1 in response to DNA damage. Cancer Res 72: 13–17.

28. LeeKC, GohWL, XuM, KuaN, LunnyD, et al. (2008) Detection of the p53 response in zebrafish embryos using new monoclonal antibodies. Oncogene 27: 629–640.

29. JeffersJR, ParganasE, LeeY, YangC, WangJ, et al. (2003) Puma is an essential mediator of p53-dependent and -independent apoptotic pathways. Cancer Cell 4: 321–328.

30. GavinAC, BoscheM, KrauseR, GrandiP, MarziochM, et al. (2002) Functional organization of the yeast proteome by systematic analysis of protein complexes. Nature 415: 141–147.

31. GroteM, WolfE, WillCL, LemmI, AgafonovDE, et al. (2010) Molecular architecture of the human Prp19/CDC5L complex. Mol Cell Biol 30: 2105–2119.

32. RenL, McLeanJR, HazbunTR, FieldsS, Vander KooiC, et al. (2011) Systematic two-hybrid and comparative proteomic analyses reveal novel yeast pre-mRNA splicing factors connected to Prp19. PLoS ONE 6: e16719 doi:10.1371/journal.pone.0016719.

33. BenathenIA, BeamCA (1977) The genetic control of X-ray resistance in budding yeast cells. Radiat Res 69: 99–116.

34. HenriquesJA, VicenteEJ, Leandro da SilvaKV, SchenbergAC (1989) PSO4: a novel gene involved in error-prone repair in Saccharomyces cerevisiae. Mutat Res 218: 111–124.

35. MahajanKN, MitchellBS (2003) Role of human Pso4 in mammalian DNA repair and association with terminal deoxynucleotidyl transferase. Proc Natl Acad Sci U S A 100: 10746–10751.

36. ReversLF, CardoneJM, BonattoD, SaffiJ, GreyM, et al. (2002) Thermoconditional modulation of the pleiotropic sensitivity phenotype by the Saccharomyces cerevisiae PRP19 mutant allele pso4-1. Nucleic Acids Res 30: 4993–5003.

37. ZhangN, KaurR, LuX, ShenX, LiL, et al. (2005) The Pso4 mRNA splicing and DNA repair complex interacts with WRN for processing of DNA interstrand cross-links. J Biol Chem 280: 40559–40567.

38. KleinriddersA, PogodaHM, IrlenbuschS, SmythN, KonczC, et al. (2009) PLRG1 is an essential regulator of cell proliferation and apoptosis during vertebrate development and tissue homeostasis. Mol Cell Biol 29: 3173–3185.

39. 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.

40. KruseJP, GuW (2009) Modes of p53 regulation. Cell 137: 609–622.

41. RaffMC, BarresBA, BurneJF, ColesHS, IshizakiY, et al. (1993) Programmed cell death and the control of cell survival: lessons from the nervous system. Science 262: 695–700.

42. Ni ChonghaileT, SarosiekKA, VoTT, RyanJA, TammareddiA, et al. (2011) Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science 334: 1129–1133.

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

44. Westerfield M (1993) The Zebrafish Book.: University of Oregon Press, Eugene, OR.

45. NixDA, Di SeraTL, DalleyBK, MilashBA, CundickRM, et al. (2010) Next generation tools for genomic data generation, distribution, and visualization. BMC Bioinformatics 11: 455.

46. FujitaPA, RheadB, ZweigAS, HinrichsAS, KarolchikD, et al. (2011) The UCSC Genome Browser database: update 2011. Nucleic Acids Res 39: D876–882.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 8
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#