#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Knock-In Reporter Mice Demonstrate that DNA Repair by Non-homologous End Joining Declines with Age


DNA damage disrupting both DNA strands, termed double strand breaks (DSBs), poses a threat to cell survival. If repaired inappropriately, such DNA breaks lead to genomic rearrangements, mutations, and ultimately cancer. Nonhomologous end joining (NHEJ) is the major pathway for repairing double-stranded breaks in mammals. Errors associated with NHEJ have been implicated in the aging process because mice with mutations in NHEJ genes exhibit premature aging. It remains unknown, however, whether NHEJ becomes impaired during normal aging. Studies of age-related changes in NHEJ have been hampered by the lack of a mouse model that would allow detection and quantification of NHEJ events. Here we report generation of NHEJ reporter mice containing a GFP-based NHEJ cassette knocked-into the ROSA26 locus. Using this mouse model, we were able to compare NHEJ across different tissues and demonstrate that NHEJ becomes less efficient and more error-prone with age. Our results provide a mechanism for age-related genomic instability and increased cancer incidence with age. The NHEJ reporter mice will be useful for a broad range of studies in the fields of aging and DNA repair.


Vyšlo v časopise: Knock-In Reporter Mice Demonstrate that DNA Repair by Non-homologous End Joining Declines with Age. PLoS Genet 10(7): e32767. doi:10.1371/journal.pgen.1004511
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004511

Souhrn

DNA damage disrupting both DNA strands, termed double strand breaks (DSBs), poses a threat to cell survival. If repaired inappropriately, such DNA breaks lead to genomic rearrangements, mutations, and ultimately cancer. Nonhomologous end joining (NHEJ) is the major pathway for repairing double-stranded breaks in mammals. Errors associated with NHEJ have been implicated in the aging process because mice with mutations in NHEJ genes exhibit premature aging. It remains unknown, however, whether NHEJ becomes impaired during normal aging. Studies of age-related changes in NHEJ have been hampered by the lack of a mouse model that would allow detection and quantification of NHEJ events. Here we report generation of NHEJ reporter mice containing a GFP-based NHEJ cassette knocked-into the ROSA26 locus. Using this mouse model, we were able to compare NHEJ across different tissues and demonstrate that NHEJ becomes less efficient and more error-prone with age. Our results provide a mechanism for age-related genomic instability and increased cancer incidence with age. The NHEJ reporter mice will be useful for a broad range of studies in the fields of aging and DNA repair.


Zdroje

1. SzilardL (1959) On the Nature of the Aging Process. Proc Natl Acad Sci U S A 45: 30–45.

2. CurtisH, CrowleyC (1963) Chromosome aberrations in liver cells in relation to the somatic mutation theory of aging. Radiat Res 19: 337–344.

3. CrowleyC, CurtisHJ (1963) The Development of Somatic Mutations in Mice with Age. Proc Natl Acad Sci U S A 49: 626–628.

4. TuckerJD, SpruillMD, RamseyMJ, DirectorAD, NathJ (1999) Frequency of spontaneous chromosome aberrations in mice: effects of age. Mutat Res 425: 135–141.

5. LiH, MitchellJR, HastyP (2008) DNA double-strand breaks: a potential causative factor for mammalian aging? Mech Ageing Dev 129: 416–424.

6. BoerrigterME, DolleME, MartusHJ, GossenJA, VijgJ (1995) Plasmid-based transgenic mouse model for studying in vivo mutations. Nature 377: 657–659.

7. DolleME, GieseH, HopkinsCL, MartusHJ, HausdorffJM, et al. (1997) Rapid accumulation of genome rearrangements in liver but not in brain of old mice. Nat Genet 17: 431–434.

8. MartinGM, SmithAC, KettererDJ, OgburnCE, DistecheCM (1985) Increased chromosomal aberrations in first metaphases of cells isolated from the kidneys of aged mice. Isr J Med Sci 21: 296–301.

9. MaoZ, BozzellaM, SeluanovA, GorbunovaV (2008) DNA repair by nonhomologous end joining and homologous recombination during cell cycle in human cells. Cell cycle 7: 2902–2906.

10. McVeyM, LeeSE (2008) MMEJ repair of double-strand breaks (director's cut): deleted sequences and alternative endings. Trends Genet 24: 529–538.

11. DecottigniesA (2013) Alternative end-joining mechanisms: a historical perspective. Front Genet 4: 48.

12. LieberMR (2010) The mechanism of double-strand DNA break repair by the nonhomologous DNA end-joining pathway. Annu Rev Biochem 79: 181–211.

13. MaoZ, HineC, TianX, Van MeterM, AuM, et al. (2011) SIRT6 promotes DNA repair under stress by activating PARP1. Science 332: 1443–1446.

14. OshimaJ, HuangS, PaeC, CampisiJ, SchiestlRH (2002) Lack of WRN results in extensive deletion at nonhomologous joining ends. Cancer Res 62: 547–551.

15. Della-MariaJ, ZhouY, TsaiMS, KuhnleinJ, CarneyJP, et al. (2011) Human Mre11/human Rad50/Nbs1 and DNA ligase IIIalpha/XRCC1 protein complexes act together in an alternative nonhomologous end joining pathway. J Biol Chem 286: 33845–33853.

16. TruongLN, LiY, ShiLZ, HwangPY, HeJ, et al. (2013) Microhomology-mediated End Joining and Homologous Recombination share the initial end resection step to repair DNA double-strand breaks in mammalian cells. Proc Natl Acad Sci U S A 110: 7720–7725.

17. AudebertM, SallesB, CalsouP (2004) Involvement of poly(ADP-ribose) polymerase-1 and XRCC1/DNA ligase III in an alternative route for DNA double-strand breaks rejoining. J Biol Chem 279: 55117–55126.

18. WangM, WuW, WuW, RosidiB, ZhangL, et al. (2006) PARP-1 and Ku compete for repair of DNA double strand breaks by distinct NHEJ pathways. Nucleic Acids Res 34: 6170–6182.

19. WangH, RosidiB, PerraultR, WangM, ZhangL, et al. (2005) DNA ligase III as a candidate component of backup pathways of nonhomologous end joining. Cancer Res 65: 4020–4030.

20. YuAM, McVeyM (2010) Synthesis-dependent microhomology-mediated end joining accounts for multiple types of repair junctions. Nucleic Acids Res 38: 5706–5717.

21. GuY, SeidlKJ, RathbunGA, ZhuC, ManisJP, et al. (1997) Growth retardation and leaky SCID phenotype of Ku70-deficient mice. Immunity 7: 653–665.

22. VogelH, LimDS, KarsentyG, FinegoldM, HastyP (1999) Deletion of Ku86 causes early onset of senescence in mice. Proc Natl Acad Sci USA 96: 10770–10775.

23. BusuttilRA, MunozDP, GarciaAM, RodierF, KimWH, et al. (2008) Effect of Ku80 deficiency on mutation frequencies and spectra at a LacZ reporter locus in mouse tissues and cells. PLoS One 3: e3458.

24. LiH, VogelH, HolcombVB, GuY, HastyP (2007) Deletion of Ku70, Ku80, or both causes early aging without substantially increased cancer. Mol Cell Biol 27: 8205–8214.

25. MostoslavskyR, ChuaKF, LombardDB, PangWW, FischerMR, et al. (2006) Genomic instability and aging-like phenotype in the absence of mammalian SIRT6. Cell 124: 315–329.

26. OzgencA, LoebLA (2005) Current advances in unraveling the function of the Werner syndrome protein. Mutat Res 577: 237–251.

27. RenK, de OrtizSP (2002) Non-homologous DNA end joining in the mature rat brain. J Neurochem 80: 949–959.

28. VyjayantiVN, RaoKS (2006) DNA double strand break repair in brain: reduced NHEJ activity in aging rat neurons. Neurosci Lett 393: 18–22.

29. FrascaD, BarattiniP, TirindelliD, GuidiL, BartoloniC, et al. (1999) Effect of age on DNA binding of the ku protein in irradiated human peripheral blood mononuclear cells (PBMC). Exp Gerontol 34: 645–658.

30. JuYJ, LeeKH, ParkJE, YiYS, YunMY, et al. (2006) Decreased expression of DNA repair proteins Ku70 and Mre11 is associated with aging and may contribute to the cellular senescence. Exp Mol Med 38: 686–693.

31. ScarpaciS, FrascaD, BarattiniP, GuidiL, DoriaG (2003) DNA damage recognition and repair capacities in human naive and memory T cells from peripheral blood of young and elderly subjects. Mech Ageing Dev 124: 517–524.

32. GarmC, Moreno-VillanuevaM, BurkleA, PetersenI, BohrVA, et al. (2013) Age and gender effects on DNA strand break repair in peripheral blood mononuclear cells. Aging Cell 12: 58–66.

33. GorbunovaV, SeluanovA, MittelmanD, Pereira-SmithOM, WilsonJH (2004) DNA end joining becomes less efficient and more error-prone during cellular senescence. Proceedings of the National Academy of Sciences of the United States of America 101: 7624–7629.

34. SeluanovA, DanekJ, HauseN, GorbunovaV (2007) Changes in the level and distribution of Ku proteins during cellular senescence. DNA repair 6: 1740–1748.

35. SeluanovA, MittelmanD, Pereira-SmithOM, WilsonJH, GorbunovaV (2004) DNA end joining becomes less efficient and more error-prone during cellular senescence. Proc Natl Acad Sci U S A 101: 7624–7629.

36. MaoZ, SeluanovA, JiangY, GorbunovaV (2007) TRF2 is required for repair of nontelomeric DNA double-strand breaks by homologous recombination. Proc Natl Acad Sci U S A 104: 13068–13073.

37. Mouse Genome SequencingC, WaterstonRH, Lindblad-TohK, BirneyE, RogersJ, et al. (2002) Initial sequencing and comparative analysis of the mouse genome. Nature 420: 520–562.

38. ZambrowiczBP, ImamotoA, FieringS, HerzenbergLA, KerrWG, et al. (1997) Disruption of overlapping transcripts in the ROSA beta geo 26 gene trap strain leads to widespread expression of beta-galactosidase in mouse embryos and hematopoietic cells. Proc Natl Acad Sci U S A 94: 3789–3794.

39. SorianoP (1999) Generalized lacZ expression with the ROSA26 Cre reporter strain. Nat Genet 21: 70–71.

40. SrinivasS, WatanabeT, LinCS, WilliamCM, TanabeY, et al. (2001) Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev Biol 1: 4.

41. VijgJ, DolleMET (2002) Large genome rearrangements as a primary cause of aging. Mech Ageing Dev 123: 907–915.

42. GarinisGA, van der HorstGT, VijgJ, HoeijmakersJH (2008) DNA damage and ageing: new-age ideas for an age-old problem. Nat Cell Biol 10: 1241–1247.

43. GorbunovaV, SeluanovA (2005) Making ends meet in old age: DSB repair and aging. Mech Ageing Dev 126: 621–628.

44. GorbunovaV, SeluanovA, MaoZ, HineC (2007) Changes in DNA repair during aging. Nucleic Acids Res 35: 7466–7474.

45. ParkCW, ParkJ, KrenBT, SteerCJ (2006) Sleeping Beauty transposition in the mouse genome is associated with changes in DNA methylation at the site of insertion. Genomics 88: 204–213.

46. BaharR, HartmannCH, RodriguezKA, DennyAD, BusuttilRA, et al. (2006) Increased cell-to-cell variation in gene expression in ageing mouse heart. Nature 441: 1011–1014.

47. KimelbergHK, NedergaardM (2010) Functions of astrocytes and their potential as therapeutic targets. Neurotherapeutics 7: 338–353.

48. KalluriR, ZeisbergM (2006) Fibroblasts in cancer. Nat Rev Cancer 6: 392–401.

49. FisherGJ, VaraniJ, VoorheesJJ (2008) Looking older: fibroblast collapse and therapeutic implications. Arch Dermatol 144: 666–672.

50. BezziP, VolterraA (2011) Astrocytes: powering memory. Cell 144: 644–645.

51. BradyN, GaymesTJ, CheungM, MuftiGJ, RassoolFV (2003) Increased error-prone NHEJ activity in myeloid leukemias is associated with DNA damage at sites that recruit key nonhomologous end-joining proteins. Cancer Res 63: 1798–1805.

52. BentleyJ, DiggleCP, HarndenP, KnowlesMA, KiltieAE (2004) DNA double strand break repair in human bladder cancer is error prone and involves microhomology-associated end-joining. Nucleic Acids Res 32: 5249–5259.

53. SeluanovA, VaidyaA, GorbunovaV (2010) Establishing primary adult fibroblast cultures from rodents. J Vis Exp doi: 10.3791/2033

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 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#