#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Defective Repair of Oxidative Base Lesions by the DNA Glycosylase Nth1 Associates with Multiple Telomere Defects


Telomeres are chromosome end structures and are essential for maintenance of genome stability. Highly repetitive telomere sequences appear to be susceptible to oxidative stress-induced damage. Oxidation may therefore have a severe impact on telomere integrity and function. A wide spectrum of oxidative pyrimidine-derivatives has been reported, including thymine glycol (Tg), that are primarily removed by a DNA glycosylase, Endonuclease III-like protein 1 (Nth1). Here, we investigate the effect of Nth1 deficiency on telomere integrity in mice. Nth1 null (Nth1−/−) mouse tissues and primary MEFs harbor higher levels of Endonuclease III-sensitive DNA lesions at telomeric repeats, in comparison to a non-telomeric locus. Furthermore, oxidative DNA damage induced by acute exposure to an oxidant is repaired slowly at telomeres in Nth1−/− MEFs. Although telomere length is not affected in the hematopoietic tissues of Nth1−/− adult mice, telomeres suffer from attrition and increased recombination and DNA damage foci formation in Nth1−/− bone marrow cells that are stimulated ex vivo in the presence of 20% oxygen. Nth1 deficiency also enhances telomere fragility in mice. Lastly, in a telomerase null background, Nth1−/− bone marrow cells undergo severe telomere loss at some chromosome ends and cell apoptosis upon replicative stress. These results suggest that Nth1 plays an important role in telomere maintenance and base repair against oxidative stress-induced base modifications. The fact that telomerase deficiency can exacerbate telomere shortening in Nth1 deficient mouse cells supports that base excision repair cooperates with telomerase to maintain telomere integrity.


Vyšlo v časopise: Defective Repair of Oxidative Base Lesions by the DNA Glycosylase Nth1 Associates with Multiple Telomere Defects. PLoS Genet 9(7): e32767. doi:10.1371/journal.pgen.1003639
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003639

Souhrn

Telomeres are chromosome end structures and are essential for maintenance of genome stability. Highly repetitive telomere sequences appear to be susceptible to oxidative stress-induced damage. Oxidation may therefore have a severe impact on telomere integrity and function. A wide spectrum of oxidative pyrimidine-derivatives has been reported, including thymine glycol (Tg), that are primarily removed by a DNA glycosylase, Endonuclease III-like protein 1 (Nth1). Here, we investigate the effect of Nth1 deficiency on telomere integrity in mice. Nth1 null (Nth1−/−) mouse tissues and primary MEFs harbor higher levels of Endonuclease III-sensitive DNA lesions at telomeric repeats, in comparison to a non-telomeric locus. Furthermore, oxidative DNA damage induced by acute exposure to an oxidant is repaired slowly at telomeres in Nth1−/− MEFs. Although telomere length is not affected in the hematopoietic tissues of Nth1−/− adult mice, telomeres suffer from attrition and increased recombination and DNA damage foci formation in Nth1−/− bone marrow cells that are stimulated ex vivo in the presence of 20% oxygen. Nth1 deficiency also enhances telomere fragility in mice. Lastly, in a telomerase null background, Nth1−/− bone marrow cells undergo severe telomere loss at some chromosome ends and cell apoptosis upon replicative stress. These results suggest that Nth1 plays an important role in telomere maintenance and base repair against oxidative stress-induced base modifications. The fact that telomerase deficiency can exacerbate telomere shortening in Nth1 deficient mouse cells supports that base excision repair cooperates with telomerase to maintain telomere integrity.


Zdroje

1. de LangeT (2005) Shelterin: the protein complex that shapes and safeguards human telomeres. Genes Dev 19: 2100–2110.

2. d'Adda di FagagnaF, TeoSH, JacksonSP (2004) Functional links between telomeres and proteins of the DNA-damage response. Genes Dev 18: 1781–1799.

3. BlackburnEH (2001) Switching and signaling at the telomere. Cell 106: 661–673.

4. von ZglinickiT (2000) Role of oxidative stress in telomere length regulation and replicative senescence. Ann N Y Acad Sci 908: 99–110.

5. Liu Y, Harrington L (2012) Murine models of dysfunctional telomeres and telomerase; Lue N, Autexier C, editors. New Jersey: John Wiley & Sons, Inc. 213–242 p.

6. CasperAM, NghiemP, ArltMF, GloverTW (2002) ATR regulates fragile site stability. Cell 111: 779–789.

7. SfeirA, KosiyatrakulST, HockemeyerD, MacRaeSL, KarlsederJ, et al. (2009) Mammalian telomeres resemble fragile sites and require TRF1 for efficient replication. Cell 138: 90–103.

8. PetersenS, SaretzkiG, von ZglinickiT (1998) Preferential accumulation of single-stranded regions in telomeres of human fibroblasts. Exp Cell Res 239: 152–160.

9. RheeDB, GhoshA, LuJ, BohrVA, LiuY (2010) Factors that influence telomeric oxidative base damage and repair by DNA glycosylase OGG1. DNA Repair (Amst) 10: 34–44.

10. RochettePJ, BrashDE (2010) Human telomeres are hypersensitive to UV-induced DNA Damage and refractory to repair. PLoS Genet 6: e1000926.

11. FumagalliM, RossielloF, ClericiM, BarozziS, CittaroD, et al. (2012) Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat Cell Biol 14: 355–365.

12. HewittG, JurkD, MarquesFD, Correia-MeloC, HardyT, et al. (2012) Telomeres are favoured targets of a persistent DNA damage response in ageing and stress-induced senescence. Nat Commun 3: 708–717.

13. HenleES, HanZ, TangN, RaiP, LuoY, et al. (1999) Sequence-specific DNA cleavage by Fe2+-mediated fenton reactions has possible biological implications. J Biol Chem 274: 962–971.

14. OikawaS, KawanishiS (1999) Site-specific DNA damage at GGG sequence by oxidative stress may accelerate telomere shortening. FEBS Lett 453: 365–368.

15. OikawaS, Tada-OikawaS, KawanishiS (2001) Site-specific DNA damage at the GGG sequence by UVA involves acceleration of telomere shortening. Biochemistry 40: 4763–4768.

16. DavidSS, O'SheaVL, KunduS (2007) Base-excision repair of oxidative DNA damage. Nature 447: 941–950.

17. SvilarD, GoellnerEM, AlmeidaKH, SobolRW (2011) Base excision repair and lesion-dependent subpathways for repair of oxidative DNA damage. Antioxid Redox Signal 14: 2491–2507.

18. ClarkJM, BeardsleyGP (1986) Thymlne glycol lesions terminate chain elongation by DNA polymerase I in vitro. Nucleic Acids Research 14: 737–749.

19. EvansJ, MaccabeeM, HatahetZ, CourcelleJ, BockrathR, et al. (1993) Thymine ring saturation and fragmentation products: lesion bypass, misinsertion and implications for mutagenesis. Mutation Research/Genetic Toxicology 299: 147–156.

20. AllerP, RouldMA, HoggM, WallaceSS, DoublieS (2007) A structural rationale for stalling of a replicative DNA polymerase at the most common oxidative thymine lesion, thymine glycol. Proc Natl Acad Sci U S A 104: 814–818.

21. OpreskoPL, FanJ, DanzyS, WilsonDM3rd, BohrVA (2005) Oxidative damage in telomeric DNA disrupts recognition by TRF1 and TRF2. Nucleic Acids Res 33: 1230–1239.

22. LuJ, LiuY (2010) Deletion of Ogg1 DNA glycosylase results in telomere base damage and length alteration in yeast. Embo J 29: 398–409.

23. HegdeML, HazraTK, MitraS (2008) Early steps in the DNA base excision/single-strand interruption repair pathway in mammalian cells. Cell Res 18: 27–47.

24. HuJ, de Souza-PintoNC, HaraguchiK, HogueBA, JarugaP, et al. (2005) Repair of Formamidopyrimidines in DNA Involves Different Glycosylases. Journal of Biological Chemistry 280: 40544–40551.

25. LunaL, BjøråsM, HoffE, RognesT, SeebergE (2000) Cell-cycle regulation, intracellular sorting and induced overexpression of the human NTH1 DNA glycosylase involved in removal of formamidopyrimidine residues from DNA. Mutation Research/DNA Repair 460: 95–104.

26. WangZ, RheeDB, LuJ, BohrCT, ZhouF, et al. (2010) Characterization of oxidative guanine damage and repair in mammalian telomeres. PLoS Genet 6: e1000951.

27. O'CallaghanN, BaackN, SharifR, FenechM (2012) A qPCR-based assay to quantify oxidized guanine and other FPG-sensitive base lesions within telomeric DNA. Biotechniques 51: 403–411.

28. LeadonSA, StampferMR, BartleyJ (1988) Production of oxidative DNA damage during the metabolic activation of benzo[a]pyrene in human mammary epithelial cells correlates with cell killing. Proc Natl Acad Sci U S A 85: 4365–4368.

29. MartinezP, ThanasoulaM, MunozP, LiaoC, TejeraA, et al. (2009) Increased telomere fragility and fusions resulting from TRF1 deficiency lead to degenerative pathologies and increased cancer in mice. Genes Dev 23: 2060–2075.

30. BadieS, EscandellJM, BouwmanP, CarlosAR, ThanasoulaM, et al. (2010) BRCA2 acts as a RAD51 loader to facilitate telomere replication and capping. Nat Struct Mol Biol 17: 1461–1469.

31. McNeesCJ, TejeraAM, MartinezP, MurgaM, MuleroF, et al. (2010) ATR suppresses telomere fragility and recombination but is dispensable for elongation of short telomeres by telomerase. J Cell Biol 188: 639–652.

32. GuP, MinJN, WangY, HuangC, PengT, et al. (2012) CTC1 deletion results in defective telomere replication, leading to catastrophic telomere loss and stem cell exhaustion. Embo J 31: 2309–2321.

33. RemeseiroS, CuadradoA, CarreteroM, MartinezP, DrosopoulosWC, et al. (2012) Cohesin-SA1 deficiency drives aneuploidy and tumourigenesis in mice due to impaired replication of telomeres. EMBO J 31: 2076–2089.

34. SedelnikovaOA, RedonCE, DickeyJS, NakamuraAJ, GeorgakilasAG, et al. (2010) Role of oxidatively induced DNA lesions in human pathogenesis. Mutation Research/Reviews in Mutation Research 704: 152–159.

35. CicciaA, ElledgeSJ (2010) The DNA damage response: making it safe to play with knives. Mol Cell 40: 179–204.

36. GloverTW, BergerC, CoyleJ, EchoB (1984) DNA polymerase alpha inhibition by aphidicolin induces gaps and breaks at common fragile sites in human chromosomes. Hum Genet 67: 136–142.

37. BrownEJ, BaltimoreD (2003) Essential and dispensable roles of ATR in cell cycle arrest and genome maintenance. Genes Dev 17: 615–628.

38. HarleyCB, FutcherAB, GreiderCW (1990) Telomeres shorten during ageing of human fibroblasts. Nature 345: 458–460.

39. HastieND, DempsterM, DunlopMG, ThompsonAM, GreenDK, et al. (1990) Telomere reduction in human colorectal carcinoma and with ageing. Nature 346: 866–868.

40. WrightWE, PiatyszekMA, RaineyWE, ByrdW, ShayJW (1996) Telomerase activity in human germline and embryonic tissues and cells. Dev Genet 18: 173–179.

41. ProwseKR, GreiderCW (1995) Developmental and tissue-specific regulation of mouse telomerase and telomere length. Proc Natl Acad Sci U S A 92: 4818–4822.

42. HemannMT, StrongMA, HaoLY, GreiderCW (2001) The shortest telomere, not average telomere length, is critical for cell viability and chromosome stability. Cell 107: 67–77.

43. LeeHW, BlascoMA, GottliebGJ, HornerJW2nd, GreiderCW, et al. (1998) Essential role of mouse telomerase in highly proliferative organs. Nature 392: 569–574.

44. BaileySM, GoodwinEH, CornforthMN (2004) Strand-specific fluorescence in situ hybridization: the CO-FISH family. Cytogenet Genome Res 107: 14–17.

45. HagelstromRT, BlagoevKB, NiedernhoferLJ, GoodwinEH, BaileySM (2010) Hyper telomere recombination accelerates replicative senescence and may promote premature aging. Proc Natl Acad Sci U S A 107: 15768–15773.

46. ChanMK, Ocampo-HafallaMT, VartanianV, JarugaP, KirkaliG, et al. (2009) Targeted deletion of the genes encoding NTH1 and NEIL1 DNA N-glycosylases reveals the existence of novel carcinogenic oxidative damage to DNA. DNA Repair 8: 786–794.

47. KarahalilB, de Souza-PintoNC, ParsonsJL, ElderRH, BohrVA (2003) Compromised incision of oxidized pyrimidines in liver mitochondria of mice deficient in NTH1 and OGG1 glycosylases. J Biol Chem 278: 33701–33707.

48. OtterleiM, WarbrickE, NagelhusTA, HaugT, SlupphaugG, et al. (1999) Post-replicative base excision repair in replication foci. Embo J 18: 3834–3844.

49. OyamaM, WakasugiM, HamaT, HashidumeH, IwakamiY, et al. (2004) Human NTH1 physically interacts with p53 and proliferating cell nuclear antigen. Biochem Biophys Res Commun 321: 183–191.

50. de Souza-PintoNC, MaynardS, HashiguchiK, HuJ, MuftuogluM, et al. (2009) The recombination protein RAD52 cooperates with the excision repair protein OGG1 for the repair of oxidative lesions in mammalian cells. Mol Cell Biol 29: 4441–4454.

51. MaynardS, SchurmanSH, HarboeC, de Souza-PintoNC, BohrVA (2009) Base excision repair of oxidative DNA damage and association with cancer and aging. Carcinogenesis 30: 2–10.

52. LiuY, SnowBE, HandeMP, YeungD, ErdmannNJ, et al. (2000) The telomerase reverse transcriptase is limiting and necessary for telomerase function in vivo. Curr Biol 10: 1459–1462.

53. ElderRH, DianovGL (2002) Repair of dihydrouracil supported by base excision repair in mNTH1 knock-out cell extracts. J Biol Chem 277: 50487–50490.

54. O'CallaghanN, DhillonV, ThomasP, FenechM (2008) A quantitative real-time PCR method for absolute telomere length. Biotechniques 44: 807–809.

55. RuferN, DragowskaW, ThornburyG, RoosnekE, LansdorpPM (1998) Telomere length dynamics in human lymphocyte subpopulations measured by flow cytometry. Nat Biotechnol 16: 743–747.

56. ZijlmansJM, MartensUM, PoonSS, RaapAK, TankeHJ, et al. (1997) Telomeres in the mouse have large inter-chromosomal variations in the number of T2AG3 repeats. Proc Natl Acad Sci U S A 94: 7423–7428.

57. BenjaminiY, HochbergY (1995) Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing. J R Statist Soc B 57: 289–300.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2013 Číslo 7
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#