#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Rosa26-GFP Direct Repeat (RaDR-GFP) Mice Reveal Tissue- and Age-Dependence of Homologous Recombination in Mammals


Cancer is a disease of the genome, caused by accumulated genetic changes, such as point mutations and large-scale sequence rearrangements. Homologous recombination (HR) is a critical DNA repair pathway. While generally accurate, HR between misaligned sequences or between homologous chromosomes can lead to insertions, deletions, and loss of heterozygosity, all of which are known to promote cancer. Indeed, most cancers harbor sequence changes caused by HR, and genetic and environmental conditions that induce or suppress HR are often carcinogenic. To enable studies of HR in vivo, we created the Rosa26 Direct Repeat-Green Fluorescent Protein (RaDR-GFP) mice that carry an integrated transgenic recombination reporter targeted to the ubiquitously expressed Rosa26 locus. Being able to detect recombinant cells by fluorescence reveals that the frequency of recombination is highly variable among tissues. Furthermore, new recombination events accumulate over time, which contributes to our understanding of why our risk for cancer increases with age. This mouse model provides new understanding of this important DNA repair pathway in vivo, and also enables future studies of genetic, environmental and physiological factors that impact the risk of HR-induced sequence rearrangements in vivo.


Vyšlo v časopise: Rosa26-GFP Direct Repeat (RaDR-GFP) Mice Reveal Tissue- and Age-Dependence of Homologous Recombination in Mammals. PLoS Genet 10(6): e32767. doi:10.1371/journal.pgen.1004299
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004299

Souhrn

Cancer is a disease of the genome, caused by accumulated genetic changes, such as point mutations and large-scale sequence rearrangements. Homologous recombination (HR) is a critical DNA repair pathway. While generally accurate, HR between misaligned sequences or between homologous chromosomes can lead to insertions, deletions, and loss of heterozygosity, all of which are known to promote cancer. Indeed, most cancers harbor sequence changes caused by HR, and genetic and environmental conditions that induce or suppress HR are often carcinogenic. To enable studies of HR in vivo, we created the Rosa26 Direct Repeat-Green Fluorescent Protein (RaDR-GFP) mice that carry an integrated transgenic recombination reporter targeted to the ubiquitously expressed Rosa26 locus. Being able to detect recombinant cells by fluorescence reveals that the frequency of recombination is highly variable among tissues. Furthermore, new recombination events accumulate over time, which contributes to our understanding of why our risk for cancer increases with age. This mouse model provides new understanding of this important DNA repair pathway in vivo, and also enables future studies of genetic, environmental and physiological factors that impact the risk of HR-induced sequence rearrangements in vivo.


Zdroje

1. Friedberg EC, Walker GC, Siede W, Wood RD, Schultz RA, et al.. (2006) DNA Repair and Mutagenesis. Washington, DC: ASM Press.

2. HoeijmakersJH (2009) DNA damage, aging, and cancer. N Engl J Med 361: 1475–1485.

3. LieberMR (2008) The mechanism of human nonhomologous DNA end joining. J Biol Chem 283: 1–5.

4. MoynahanME, JasinM (2010) Mitotic homologous recombination maintains genomic stability and suppresses tumorigenesis. Nat Rev Mol Cell Biol 11: 196–207.

5. HelledayT, LoJ, van GentDC, EngelwardBP (2007) DNA double-strand break repair: from mechanistic understanding to cancer treatment. DNA Repair (Amst) 6: 923–935.

6. San FilippoJ, SungP, KleinH (2008) Mechanism of eukaryotic homologous recombination. Annu Rev Biochem 77: 229–257.

7. HolthausenJT, WymanC, KanaarR (2010) Regulation of DNA strand exchange in homologous recombination. DNA Repair (Amst) 9: 1264–1272.

8. AllenC, AshleyAK, HromasR, NickoloffJA (2011) More forks on the road to replication stress recovery. J Mol Cell Biol 3: 4–12.

9. KassEM, JasinM (2010) Collaboration and competition between DNA double-strand break repair pathways. FEBS Lett 584: 3703–3708.

10. YuVP, KoehlerM, SteinleinC, SchmidM, HanakahiLA, et al. (2000) Gross chromosomal rearrangements and genetic exchange between nonhomologous chromosomes following BRCA2 inactivation. Genes Dev 14: 1400–1406.

11. van GentDC, HoeijmakersJH, KanaarR (2001) Chromosomal stability and the DNA double-stranded break connection. Nat Rev Genet 2: 196–206.

12. DolleME, KuiperRV, RoodbergenM, RobinsonJ, de VlugtS, et al. (2011) Broad segmental progeroid changes in short-lived Ercc1(-/Delta7) mice. Pathobiol Aging Age Relat Dis doi: 10.3402/pba.v1i0.7219

13. RelieneR, BishopAJ, SchiestlRH (2007) Involvement of homologous recombination in carcinogenesis. Adv Genet 58: 67–87.

14. GuptaPK, SahotaA, BoyadjievSA, ByeS, ShaoC, et al. (1997) High frequency in vivo loss of heterozygosity is primarily a consequence of mitotic recombination. Cancer Res 57: 1188–1193.

15. MorleyAA, GristSA, TurnerDR, KutlacaA, BennettG (1990) Molecular nature of in vivo mutations in human cells at the autosomal HLA-A locus. Cancer Res 50: 4584–4587.

16. HaigisKM, CayaJG, ReichelderferM, DoveWF (2002) Intestinal adenomas can develop with a stable karyotype and stable microsatellites. Proc Natl Acad Sci U S A 99: 8927–8931.

17. ShammasMA, Shmookler ReisRJ, KoleyH, BatchuRB, LiC, et al. (2009) Dysfunctional homologous recombination mediates genomic instability and progression in myeloma. Blood 113: 2290–2297.

18. JamesCD, CarlbomE, NordenskjoldM, CollinsVP, CaveneeWK (1989) Mitotic recombination of chromosome 17 in astrocytomas. Proc Natl Acad Sci U S A 86: 2858–2862.

19. PalJ, BertheauR, BuonL, QaziA, BatchuRB, et al. (2011) Genomic evolution in Barrett's adenocarcinoma cells: critical roles of elevated hsRAD51, homologous recombination and Alu sequences in the genome. Oncogene 30: 3585–3598.

20. HagstromSA, DryjaTP (1999) Mitotic recombination map of 13cen-13q14 derived from an investigation of loss of heterozygosity in retinoblastomas. Proc Natl Acad Sci U S A 96: 2952–2957.

21. NeuwirthEA, HonmaM, GrosovskyAJ (2007) Interchromosomal crossover in human cells is associated with long gene conversion tracts. Mol Cell Biol 27: 5261–5274.

22. BishopAJ, SchiestlRH (2001) Homologous recombination as a mechanism of carcinogenesis. Biochim Biophys Acta 1471: M109–121.

23. De Weerd-KasteleinEA, KeijzerW, RainaldiG, BootsmaD (1977) Induction of sister chromatid exchanges in xeroderma pigmentosum cells after exposure to ultraviolet light. Mutat Res 45: 253–261.

24. BillCA, NickoloffJA (2001) Spontaneous and ultraviolet light-induced direct repeat recombination in mammalian cells frequently results in repeat deletion. Mutat Res 487: 41–50.

25. ConnellJR (1979) The relationship between sister chromatid exchange, chromosome aberration and gene mutation induction by several reactive polycyclic hydrocarbon metabolites in cultured mammalian cells. Int J Cancer 24: 485–489.

26. BishopAJ, KosarasB, SidmanRL, SchiestlRH (2000) Benzo(a)pyrene and X-rays induce reversions of the pink-eyed unstable mutation in the retinal pigment epithelium of mice. Mutat Res 457: 31–40.

27. EllisNA, LennonDJ, ProytchevaM, AlhadeffB, HendersonEE, et al. (1995) Somatic intragenic recombination within the mutated locus BLM can correct the high sister-chromatid exchange phenotype of Bloom syndrome cells. Am J Hum Genet 57: 1019–1027.

28. AllenC, KurimasaA, BrennemanMA, ChenDJ, NickoloffJA (2002) DNA-dependent protein kinase suppresses double-strand break-induced and spontaneous homologous recombination. Proc Natl Acad Sci U S A 99: 3758–3763.

29. PierceAJ, HuP, HanM, EllisN, JasinM (2001) Ku DNA end-binding protein modulates homologous repair of double-strand breaks in mammalian cells. Genes Dev 15: 3237–3242.

30. ChapmanJR, TaylorMR, BoultonSJ (2012) Playing the end game: DNA double-strand break repair pathway choice. Mol Cell 47: 497–510.

31. MimitouEP, SymingtonLS (2008) Sae2, Exo1 and Sgs1 collaborate in DNA double-strand break processing. Nature 455: 770–774.

32. ZhuZ, ChungWH, ShimEY, LeeSE, IraG (2008) Sgs1 helicase and two nucleases Dna2 and Exo1 resect DNA double-strand break ends. Cell 134: 981–994.

33. SungP, RobbersonDL (1995) DNA strand exchange mediated by a RAD51-ssDNA nucleoprotein filament with polarity opposite to that of RecA. Cell 82: 453–461.

34. BensonFE, StasiakA, WestSC (1994) Purification and characterization of the human Rad51 protein, an analogue of E. coli RecA. EMBO J 13: 5764–5771.

35. WongJM, IonescuD, InglesCJ (2003) Interaction between BRCA2 and replication protein A is compromised by a cancer-predisposing mutation in BRCA2. Oncogene 22: 28–33.

36. DaviesAA, MassonJY, McIlwraithMJ, StasiakAZ, StasiakA, et al. (2001) Role of BRCA2 in control of the RAD51 recombination and DNA repair protein. Mol Cell 7: 273–282.

37. ThorslundT, WestSC (2007) BRCA2: a universal recombinase regulator. Oncogene 26: 7720–7730.

38. HastingsPJ (1992) Mechanism and control of recombination in fungi. Mutat Res 284: 97–110.

39. BzymekM, ThayerNH, OhSD, KlecknerN, HunterN (2010) Double Holliday junctions are intermediates of DNA break repair. Nature 464: 937–941.

40. McIlwraithMJ, WestSC (2008) DNA repair synthesis facilitates RAD52-mediated second-end capture during DSB repair. Mol Cell 29: 510–516.

41. IraG, MalkovaA, LiberiG, FoianiM, HaberJE (2003) Srs2 and Sgs1-Top3 suppress crossovers during double-strand break repair in yeast. Cell 115: 401–411.

42. CoxMM, GoodmanMF, KreuzerKN, SherrattDJ, SandlerSJ, et al. (2000) The importance of repairing stalled replication forks. Nature 404: 37–41.

43. RelieneR, SchiestlRH (2003) Mouse models for induced genetic instability at endogenous loci. Oncogene 22: 7000–7010.

44. RelieneR, BishopAJ, AubrechtJ, SchiestlRH (2004) In vivo DNA deletion assay to detect environmental and genetic predisposition to cancer. Methods Mol Biol 262: 125–140.

45. LiangL, DengL, ShaoC, StambrookPJ, TischfieldJA (2000) In vivo loss of heterozygosity in T-cells of B6C3F1 Aprt(+/−) mice. Environ Mol Mutagen 35: 150–157.

46. ShaoC, DengL, HenegariuO, LiangL, RaikwarN, et al. (1999) Mitotic recombination produces the majority of recessive fibroblast variants in heterozygous mice. Proc Natl Acad Sci USA 96: 9230–9235.

47. RoederGS, FinkGR (1982) Movement of yeast transposable elements by gene conversion. Proc Natl Acad Sci U S A 79: 5621–5625.

48. SchiestlRH, PrakashS (1988) RAD1, an excision repair gene of Saccharomyces cerevisiae, is also involved in recombination. Mol Cell Biol 8: 3619–3626.

49. BrennanRJ, SchiestlRH (2004) Detecting carcinogens with the yeast DEL assay. Methods Mol Biol 262: 111–124.

50. LiskayRM, StachelekJL (1983) Evidence for intrachromosomal gene conversion in cultured mouse cells. Cell 35: 157–165.

51. BollagRJ, LiskayRM (1991) Direct-repeat analysis of chromatid interactions during intrachromosomal recombination in mouse cells. Mol Cell Biol 11: 4839–4845.

52. BollagRJ, LiskayRM (1988) Conservative intrachromosomal recombination between inverted repeats in mouse cells: association between reciprocal exchange and gene conversion. Genetics 119: 161–169.

53. TsujimuraT, MaherVM, GodwinAR, LiskayRM, McCormickJJ (1990) Frequency of intrachromosomal homologous recombination induced by UV radiation in normally repairing and excision repair-deficient human cells. Proc Natl Acad Sci USA 87: 1566–1570.

54. TaghianDG, NickoloffJA (1997) Chromosomal double-strand breaks induce gene conversion at high frequency in mammalian cells. Mol Cell Biol 17: 6386–6393.

55. AllenC, MillerCA, NickoloffJA (2003) The mutagenic potential of a single DNA double-strand break in a mammalian chromosome is not influenced by transcription. DNA Repair (Amst) 2: 1147–1156.

56. PierceAJ, HuP, HanM, EllisN, JasinM (2001) Ku DNA end-binding protein modulates homologous repair of double-strand breaks in mammalian cells. Genes Dev 15: 3237–42.

57. KiziltepeT, YanA, DongM, JonnalagaddaVS, DedonPC, et al. (2005) Delineation of the chemical pathways underlying nitric oxide-induced homologous recombination in mammalian cells. Chem Biol 12: 357–369.

58. HendricksCA, AlmeidaKH, StittMS, JonnalagaddaVS, RugoRE, et al. (2003) Spontaneous mitotic homologous recombination at an enhanced yellow fluorescent protein (EYFP) cDNA direct repeat in transgenic mice. Proc Natl Acad Sci U S A 100: 6325–6330.

59. HendricksCA, EngelwardBP (2004) “Recombomice”: the past, present, and future of recombinationdetection in mice. DNA Repair (Amst) 3: 1255–1261.

60. Wiktor-BrownDM, HendricksCA, OlipitzW, EngelwardBP (2006) Age-dependent accumulation of recombinant cells in the mouse pancreas revealed by in situ fluorescence imaging. Proc Natl Acad Sci U S A 103: 11862–11867.

61. Wiktor-BrownDM, KwonHS, NamYS, SoPT, EngelwardBP (2008) Integrated one- and two-photon imaging platform reveals clonal expansion as a major driver of mutation load. Proc Natl Acad Sci U S A 105: 10314–10319.

62. Wiktor-BrownDM, OlipitzW, HendricksCA, RugoRE, EngelwardBP (2008) Tissue-specific differences in the accumulation of sequence rearrangements with age. DNA Repair (Amst) 7: 694–703.

63. Wiktor-BrownDM, Sukup-JacksonMR, FakhraldeenSA, HendricksCA, EngelwardBP (2011) p53 null fluorescent yellow direct repeat (FYDR) mice have normal levels of homologous recombination. DNA Repair (Amst) 10: 1294–1299.

64. KimKH, RaganT, PreviteMJ, BahlmannK, HarleyBA, et al. (2007) Three-dimensional tissue cytometer based on high-speed multiphoton microscopy. Cytometry A 71: 991–1002.

65. Wiktor-BrownDM, HendricksCA, OlipitzW, RogersAB, EngelwardBP (2006) Applications of fluorescence for detecting rare sequence rearrangements in vivo. Cell Cycle 5: 2715–2719.

66. JonnalagaddaVS, MatsuguchiT, EngelwardBP (2005) Interstrand crosslink-induced homologous recombination carries an increased risk of deletions and insertions. DNA Repair (Amst) 4: 594–605.

67. OkabeM, IkawaM, KominamiK, NakanishiT, NishimuneY (1997) ‘Green mice’ as a source of ubiquitous green cells. FEBS Lett 407: 313–319.

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

69. OriiKE, LeeY, KondoN, McKinnonPJ (2006) Selective utilization of nonhomologous end-joining and homologous recombination DNA repair pathways during nervous system development. Proc Natl Acad Sci USA 103: 10017–10022.

70. FrappartPO, McKinnonPJ (2007) BRCA2 function and the central nervous system. Cell Cycle 6: 2453–2457.

71. CosentinoL, Shaver-WalkerP, HeddleJA (1996) The relationships among stem cells, crypts, and villi in the small intestine of mice as determined by mutation tagging. Dev Dyn 207: 420–428.

72. FreemanHJ (2008) Crypt region localization of intestinal stem cells in adults. World J Gastroenterol 14: 7160–7162.

73. BarkerN, van de WeteringM, CleversH (2008) The intestinal stem cell. Genes Dev 22: 1856–1864.

74. FuD, CalvoJA, SamsonLD (2012) Balancing repair and tolerance of DNA damage caused by alkylating agents. Nat Rev Cancer 12: 104–120.

75. SchiestlRH, AubrechtJ, KhogaliF, CarlsN (1997) Carcinogens induce reversion of the mouse pink-eyed unstable mutation. Proc Natl Acad Sci U S A 94: 4576–4581.

76. SharanSK, MorimatsuM, AlbrechtU, LimDS, RegelE, et al. (1997) Embryonic lethality and radiation hypersensitivity mediated by Rad51 in mice lacking Brca2. Nature 386: 804–810.

77. SonodaE, SasakiMS, BuersteddeJM, BezzubovaO, ShinoharaA, et al. (1998) Rad51-deficient vertebrate cells accumulate chromosomal breaks prior to cell death. EMBO J 17: 598–608.

78. BartschDK, GressTM, LangerP (2012) Familial pancreatic cancer–current knowledge. Nat Rev Gastroenterol Hepatol 9: 445–453.

79. RoyR, ChunJ, PowellSN (2012) BRCA1 and BRCA2: different roles in a common pathway of genome protection. Nat Rev Cancer 12: 68–78.

80. ReichertM, RustgiAK (2011) Pancreatic ductal cells in development, regeneration, and neoplasia. J Clin Invest 121: 4572–4578.

81. GrippoPJ, NowlinPS, DemeureMJ, LongneckerDS, SandgrenEP (2003) Preinvasive pancreatic neoplasia of ductal phenotype induced by acinar cell targeting of mutant Kras in transgenic mice. Cancer Res 63: 2016–2019.

82. MeansAL, MeszoelyIM, SuzukiK, MiyamotoY, RustgiAK, et al. (2005) Pancreatic epithelial plasticity mediated by acinar cell transdifferentiation and generation of nestin-positive intermediates. Development 132: 3767–3776.

83. Gomez-LechonMJ, LahozA, GombauL, CastellJV, DonatoMT (2010) In vitro evaluation of potential hepatotoxicity induced by drugs. Curr Pharm Des 16: 1963–1977.

84. RueffJ, ChiapellaC, ChipmanJK, DarroudiF, SilvaID, et al. (1996) Development and validation of alternative metabolic systems for mutagenicity testing in short-term assays. Mutat Res 353: 151–176.

85. JiaL, LiuX (2007) The conduct of drug metabolism studies considered good practice (II): in vitro experiments. Curr Drug Metab 8: 822–829.

86. WintonDJ, BlountMA, PonderBA (1988) A clonal marker induced by mutation in mouse intestinal epithelium. Nature 333: 463–466.

87. ZhangJ, StevensMF, BradshawTD (2012) Temozolomide: mechanisms of action, repair and resistance. Curr Mol Pharmacol 5: 102–14.

88. AllanJM, TravisLB (2005) Mechanisms of therapy-related carcinogenesis. Nat Rev Cancer 5: 943–55.

89. KassEM, HelgadottirHR, ChenCC, BarberaM, WangR, et al. (2013) Double-strand break repair by homologous recombination in primary mouse somatic cells requires BRCA1 but not the ATM kinase. Proc Natl Acad Sci U S A 110: 5564–5569.

90. KimotoT, SuzukiK, KobayashiXM, DobrovolskyVN, HeflichRH, et al. (2011) Manifestation of Pig-a mutant bone marrow erythroids and peripheral blood erythrocytes in mice treated with N-ethyl-Nnitrosourea: direct sequencing of Pig a cDNA from bone marrow cells negative for GPI-anchored protein expression. Mutat Res 723: 36–42.

91. GossenJA, de LeeuwWJ, TanCH, ZwarthoffEC, BerendsF, et al. (1989) Efficient rescue of integrated shuttle vectors from transgenic mice: a model for studying mutations in vivo. Proc Natl Acad Sci U S A 86: 7971–7975.

92. StieglerGL, StillwellLC (1993) Big Blue transgenic mouse lacI mutation analysis. Environ Mol Mutagen 22: 127–129.

93. DolleME, MartusHJ, GossenJA, BoerrigterME, VijgJ (1996) Evaluation of a plasmid-based transgenic mouse model for detecting in vivo mutations. Mutagenesis 11: 111–118.

94. SwigerRR, CosentinoL, ShimaN, BielasJH, Cruz-MunozW, et al. (1999) The cII locus in the MutaMouse system. Environ Mol Mutagen 34: 201–207.

95. NohmiT, KatohM, SuzukiH, MatsuiM, YamadaM, et al. (1996) A new transgenic mouse mutagenesis test system using Spi- and 6-thioguanine selections. Environ Mol Mutagen 28: 465–470.

96. FriedbergEC (2005) Suffering in silence: the tolerance of DNA damage. Nat Rev Mol Cell Biol 6: 943–953.

97. van GentDC, HoeijmakersJH, KanaarR (2001) Chromosomal stability and the DNA double-stranded break connection. Nat Rev Gen 2: 196–206.

98. SamuelS, WalshR, WebbJ, RobinsA, PottenC, et al. (2009) Characterization of putative stem cells in isolated human colonic crypt epithelial cells and their interactions with myofibroblasts. Am J Physiol Cell Physiol 296: C296–305.

99. Soille P (2003) Morphological image analysis: principles and applications: pringer-Verlag New York, IncS.

100. Ledda-ColumbanoGM, PerraA, PibiriM, MolotzuF, ColumbanoA (2005) Induction of pancreatic acinar cell proliferation by thyroid hormone. J Endocrinol 185: 393–399.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 6
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#