#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Genetic Interactions Implicating Postreplicative Repair in Okazaki Fragment Processing


Genome duplication via the process of DNA replication is a prerequisite for cell division and underlies the propagation of all living organisms. This fundamentally important mechanism has been highly conserved throughout eukaryotic evolution, allowing us to use the relatively simple and genetically tractable Saccharomyces cerevisiae as a model to better understand DNA replication in human cells. Furthermore, there is strong evidence to suggest that defects in DNA replication are prominent contributors to mutation and genome instability, a hallmark of cancer. Not surprisingly, evolution has selected for mechanisms to mitigate the effects of defective replication and avoid the most harmful outcomes. Postreplicative repair (PRR) pathways are two such mechanisms with well described functions in promoting the completion of replication under adverse conditions. In this study, we utilized a non-biased genome wide genetic screen to systematically identify conditions under which PRR is required. Our findings indicate that in addition to previously described roles in rescuing DNA synthesis defects, PRR is also required in response to aberrant DNA processing. Specifically, we report a requirement for PRR in cells lacking RAD27, the yeast homolog of the tumor suppressor FEN1. These findings expand the known functions of PRR and reveal their importance in promoting the viability of cells lacking a known tumor suppressor.


Vyšlo v časopise: Genetic Interactions Implicating Postreplicative Repair in Okazaki Fragment Processing. PLoS Genet 11(11): e32767. doi:10.1371/journal.pgen.1005659
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1005659

Souhrn

Genome duplication via the process of DNA replication is a prerequisite for cell division and underlies the propagation of all living organisms. This fundamentally important mechanism has been highly conserved throughout eukaryotic evolution, allowing us to use the relatively simple and genetically tractable Saccharomyces cerevisiae as a model to better understand DNA replication in human cells. Furthermore, there is strong evidence to suggest that defects in DNA replication are prominent contributors to mutation and genome instability, a hallmark of cancer. Not surprisingly, evolution has selected for mechanisms to mitigate the effects of defective replication and avoid the most harmful outcomes. Postreplicative repair (PRR) pathways are two such mechanisms with well described functions in promoting the completion of replication under adverse conditions. In this study, we utilized a non-biased genome wide genetic screen to systematically identify conditions under which PRR is required. Our findings indicate that in addition to previously described roles in rescuing DNA synthesis defects, PRR is also required in response to aberrant DNA processing. Specifically, we report a requirement for PRR in cells lacking RAD27, the yeast homolog of the tumor suppressor FEN1. These findings expand the known functions of PRR and reveal their importance in promoting the viability of cells lacking a known tumor suppressor.


Zdroje

1. Myung K, Chen C, Kolodner RD (2001) Multiple pathways cooperate in the suppression of genome instability in Saccharomyces cerevisiae. Nature 411: 1073–1076. 11429610

2. Cremona CA, Sarangi P, Yang Y, Hang LE, Rahman S, et al. (2012) Extensive DNA damage-induced sumoylation contributes to replication and repair and acts in addition to the Mec1 checkpoint. Molecular Cell 45: 422–432. doi: 10.1016/j.molcel.2011.11.028 22285753

3. Hoege C, Pfander B, Moldovan G-L, Pyrowolakis G, Jentsch S (2002) RAD6-dependent DNA repair is linked to modification of PCNA by ubiquitin and SUMO. Nature 419: 135–141. 12226657

4. Pfander B, Moldovan G-L, Sacher M, Hoege C, Jentsch S (2005) SUMO-modified PCNA recruits Srs2 to prevent recombination during S phase. Nature 436: 428–433. 15931174

5. Papouli E, Chen S, Davies AA, Huttner D, Krejci L, et al. (2005) Crosstalk between SUMO and ubiquitin on PCNA is mediated by recruitment of the helicase Srs2p. Molecular Cell 19: 123–133. 15989970

6. Haracska L, Torres-Ramos CA, Johnson RE, Prakash S, Prakash L (2004) Opposing effects of ubiquitin conjugation and SUMO modification of PCNA on replicational bypass of DNA lesions in Saccharomyces cerevisiae. Molecular and Cellular Biology 24: 4267–4274. 15121847

7. Xu G, Paige JS, Jaffrey SR (2010) Global analysis of lysine ubiquitination by ubiquitin remnant immunoaffinity profiling. Nature Biotechnology 28: 868–873. doi: 10.1038/nbt.1654 20639865

8. Das-Bradoo S, Nguyen HD, Wood JL, Ricke RM, Haworth JC, et al. (2010) Defects in DNA ligase I trigger PCNA ubiquitylation at Lys 107. Nature Cell Biology 12: 74–79. doi: 10.1038/ncb2007 20010813

9. Povlsen LK, Beli P, Wagner SA, Poulsen SL, Sylvestersen KB, et al. (2012) Systems-wide analysis of ubiquitylation dynamics reveals a key role for PAF15 ubiquitylation in DNA-damage bypass. Nature Cell Biology 14: 1089–1098. doi: 10.1038/ncb2579 23000965

10. Nguyen HD, Becker J, Thu YM, Costanzo M, Koch EN, et al. (2013) Unligated Okazaki fragments induce PCNA ubiquitination and a requirement for Rad59-dependent replication fork progression. PloS One 8: e66379. 23824283

11. Elia AE, Boardman AP, Wang DC, Huttlin EL, Everley RA, et al. (2015) Quantitative Proteomic Atlas of Ubiquitination and Acetylation in the DNA Damage Response. Molecular Cell 59: 867–881. doi: 10.1016/j.molcel.2015.05.006 26051181

12. Lopes M, Foiani M, Sogo JM (2006) Multiple mechanisms control chromosome integrity after replication fork uncoupling and restart at irreparable UV lesions. Molecular Cell 21: 15–27. 16387650

13. Davies AA, Huttner D, Daigaku Y, Chen S, Ulrich HD (2008) Activation of ubiquitin-dependent DNA damage bypass is mediated by replication protein a. Molecular Cell 29: 625–636. doi: 10.1016/j.molcel.2007.12.016 18342608

14. Stelter P, Ulrich HD (2003) Control of spontaneous and damage-induced mutagenesis by SUMO and ubiquitin conjugation. Nature 425: 188–191. 12968183

15. Garg P, Burgers PM (2005) Ubiquitinated proliferating cell nuclear antigen activates translesion DNA polymerases η and Rev1. Proceedings of the National Academy of Sciences of the United States of America 102: 18361–18366. 16344468

16. Freudenthal BD, Gakhar L, Ramaswamy S, Washington MT (2010) Structure of monoubiquitinated PCNA and implications for translesion synthesis and DNA polymerase exchange. Nature Structural & Molecular Biology 17: 479–484.

17. Kochenova OV, Daee DL, Mertz TM, Shcherbakova PV, Jinks-Robertson S (2015) DNA Polymerase ζ-dependent lesion bypass in Saccharomyces cerevisiae is accompanied by error-prone copying of long stretches of adjacent DNA. PLoS Genetics 11: e1005110 doi: 10.1371/journal.pgen.1005110 25826305

18. Branzei D (2011) Ubiquitin family modifications and template switching. FEBS Letters 585: 2810–2817. doi: 10.1016/j.febslet.2011.04.053 21539841

19. Vanoli F, Fumasoni M, Szakal B, Maloisel L, Branzei D (2010) Replication and recombination factors contributing to recombination-dependent bypass of DNA lesions by template switch. PLoS Genetics 6: e1001205. doi: 10.1371/journal.pgen.1001205 21085632

20. Northam MR, Garg P, Baitin DM, Burgers PM, Shcherbakova PV (2006) A novel function of DNA polymerase ζ regulated by PCNA. The EMBO Journal 25: 4316–4325. 16957771

21. Northam MR, Robinson HA, Kochenova OV, Shcherbakova PV (2010) Participation of DNA polymerase ζ in replication of undamaged DNA in Saccharomyces cerevisiae. Genetics 184: 27–42. doi: 10.1534/genetics.109.107482 19841096

22. Karras GI, Jentsch S (2010) The RAD6 DNA damage tolerance pathway operates uncoupled from the replication fork and is functional beyond S phase. Cell 141: 255–267. doi: 10.1016/j.cell.2010.02.028 20403322

23. Becker JR, Nguyen HD, Wang X, Bielinsky A- K (2014) Mcm10 deficiency causes defective-replisome-induced mutagenesis and a dependency on error-free postreplicative repair. Cell Cycle 13: 1737–1748. doi: 10.4161/cc.28652 24674891

24. Bellaoui M, Chang M, Ou J, Xu H, Boone C, et al. (2003) Elg1 forms an alternative RFC complex important for DNA replication and genome integrity. The EMBO Journal 22: 4304–4313. 12912927

25. Ayyagari R, Gomes XV, Gordenin DA, Burgers PM (2003) Okazaki fragment maturation in yeast I. Distribution of functions between FEN1 and DNA2. Journal of Biological Chemistry 278: 1618–1625. 12424238

26. Jin YH, Ayyagari R, Resnick MA, Gordenin DA, Burgers PM (2003) Okazaki Fragment Maturation in Yeast II. Cooperation between the polymerase and 3'-5'-exonuclease activities of Pol δ in the creation of a ligatable nick. Journal of Biological Chemistry 278: 1626–1633. 12424237

27. Bae S-H, Bae K-H, Kim J-A, Seo Y-S (2001) RPA governs endonuclease switching during processing of Okazaki fragments in eukaryotes. Nature 412: 456–461. 11473323

28. Levikova M, Cejka P (2015) The Saccharomyces cerevisiae Dna2 can function as a sole nuclease in the processing of Okazaki fragments in DNA replication. Nucleic Acids Research: gkv710.

29. Bae S-H, Seo Y-S (2000) Characterization of the enzymatic properties of the yeast DNA2 helicase/endonuclease suggests a new model for Okazaki fragment processing. Journal of Biological Chemistry 275: 38022–38031. 10984490

30. Reagan MS, Pittenger C, Siede W, Friedberg EC (1995) Characterization of a mutant strain of Saccharomyces cerevisiae with a deletion of the RAD27 gene, a structural homolog of the RAD2 nucleotide excision repair gene. Journal of Bacteriology 177: 364–371. 7814325

31. Tishkoff DX, Filosi N, Gaida GM, Kolodner RD (1997) A novel mutation avoidance mechanism dependent on S. cerevisiae RAD27 is distinct from DNA mismatch repair. Cell 88: 253–263. 9008166

32. Symington LS (1998) Homologous recombination is required for the viability of rad27 mutants. Nucleic Acids Research 26: 5589–5595. 9837987

33. Jin YH, Obert R, Burgers PM, Kunkel TA, Resnick MA, et al. (2001) The 3′→ 5′ exonuclease of DNA polymerase δ can substitute for the 5′ flap endonuclease Rad27/Fen1 in processing Okazaki fragments and preventing genome instability. Proceedings of the National Academy of Sciences 98: 5122–5127.

34. Parnas O, Zipin‐Roitman A, Pfander B, Liefshitz B, Mazor Y, et al. (2010) Elg1, an alternative subunit of the RFC clamp loader, preferentially interacts with SUMOylated PCNA. The EMBO Journal 29: 2611–2622. doi: 10.1038/emboj.2010.128 20571511

35. Kubota T, Nishimura K, Kanemaki MT, Donaldson AD (2013) The Elg1 replication factor C-like complex functions in PCNA unloading during DNA replication. Molecular Cell 50: 273–280. doi: 10.1016/j.molcel.2013.02.012 23499004

36. Kubota T, Katou Y, Nakato R, Shirahige K, Donaldson AD (2015) Replication-coupled PCNA unloading by the Elg1 complex occurs genome-wide and requires Okazaki fragment ligation. Cell Reports 12: 774–787. doi: 10.1016/j.celrep.2015.06.066 26212319

37. Li Z, Vizeacoumar FJ, Bahr S, Li J, Warringer J, et al. (2011) Systematic exploration of essential yeast gene function with temperature-sensitive mutants. Nature Biotechnology 29: 361–367. doi: 10.1038/nbt.1832 21441928

38. Baryshnikova A, Costanzo M, Dixon S, Vizeacoumar FJ, Myers CL, et al. (2010) Synthetic genetic array (SGA) analysis in Saccharomyces cerevisiae and Schizosaccharomyces pombe. Methods in Enzymology 470: 145–179. doi: 10.1016/S0076-6879(10)70007-0 20946810

39. Schuldiner M, Collins SR, Thompson NJ, Denic V, Bhamidipati A, et al. (2005) Exploration of the function and organization of the yeast early secretory pathway through an epistatic miniarray profile. Cell 123: 507–519. 16269340

40. Beltrao P, Cagney G, Krogan NJ (2010) Quantitative genetic interactions reveal biological modularity. Cell 141: 739–745. doi: 10.1016/j.cell.2010.05.019 20510918

41. Huang M-E, Kolodner RD (2005) A biological network in Saccharomyces cerevisiae prevents the deleterious effects of endogenous oxidative DNA damage. Molecular Cell 17: 709–720. 15749020

42. Motegi A, Kuntz K, Majeed A, Smith S, Myung K (2006) Regulation of gross chromosomal rearrangements by ubiquitin and SUMO ligases in Saccharomyces cerevisiae. Molecular and Cellular Biology 26: 1424–1433. 16449653

43. Mann HB, Whitney DR (1947) On a test of whether one of two random variables is stochastically larger than the other. The Annals of Mathematical Statistics: 50–60.

44. Windecker H, Ulrich HD (2008) Architecture and assembly of poly-SUMO chains on PCNA in Saccharomyces cerevisiae. Journal of Molecular Biology 376: 221–231. 18155241

45. Chen XL, Silver HR, Xiong L, Belichenko I, Adegite C, et al. (2007) Topoisomerase I-dependent viability loss in Saccharomyces cerevisiae mutants defective in both SUMO conjugation and DNA repair. Genetics 177: 17–30. 17603101

46. Waters LS, Minesinger BK, Wiltrout ME, D'Souza S, Woodruff RV, et al. (2009) Eukaryotic translesion polymerases and their roles and regulation in DNA damage tolerance. Microbiology and Molecular Biology Reviews 73: 134–154. doi: 10.1128/MMBR.00034-08 19258535

47. Nelson JR, Lawrence CW, Hinkle DC (1996) Thymine-thymine dimer bypass by yeast DNA polymerase ζ. Science 272: 1646–1649. 8658138

48. Serero A, Jubin C, Loeillet S, Legoix-Né P, Nicolas AG (2014) Mutational landscape of yeast mutator strains. Proceedings of the National Academy of Sciences 111: 1897–1902.

49. Osborn AJ, Elledge SJ (2003) Mrc1 is a replication fork component whose phosphorylation in response to DNA replication stress activates Rad53. Genes & Development 17: 1755–1767.

50. Burgers PM (2009) Polymerase dynamics at the eukaryotic DNA replication fork. Journal of Biological Chemistry 284: 4041–4045. doi: 10.1074/jbc.R800062200 18835809

51. Garg P, Stith CM, Sabouri N, Johansson E, Burgers PM (2004) Idling by DNA polymerase δ maintains a ligatable nick during lagging-strand DNA replication. Genes & Development 18: 2764–2773.

52. Budd ME, Reis CC, Smith S, Myung K, Campbell JL (2006) Evidence suggesting that Pif1 helicase functions in DNA replication with the Dna2 helicase/nuclease and DNA polymerase δ. Molecular and Cellular Biology 26: 2490–2500. 16537895

53. Rossi ML, Pike JE, Wang W, Burgers PM, Campbell JL, et al. (2008) Pif1 helicase directs eukaryotic Okazaki fragments toward the two-nuclease cleavage pathway for primer removal. Journal of Biological Chemistry 283: 27483–27493. doi: 10.1074/jbc.M804550200 18689797

54. Stewart JA, Miller AS, Campbell JL, Bambara RA (2008) Dynamic removal of replication protein A by Dna2 facilitates primer cleavage during Okazaki fragment processing in Saccharomyces cerevisiae. Journal of Biological Chemistry 283: 31356–31365. doi: 10.1074/jbc.M805965200 18799459

55. Gary R, Park MS, Nolan JP, Cornelius HL, Kozyreva OG, et al. (1999) A novel role in DNA metabolism for the binding of Fen1/Rad27 to PCNA and implications for genetic risk. Molecular and Cellular Biology 19: 5373–5382. 10409728

56. Tishkoff DX, Boerger AL, Bertrand P, Filosi N, Gaida GM, et al. (1997) Identification and characterization of Saccharomyces cerevisiae EXO1, a gene encoding an exonuclease that interacts with MSH2. Proceedings of the National Academy of Sciences 94: 7487–7492.

57. Sun X, Thrower D, Qiu J, Wu P, Zheng L, et al. (2003) Complementary functions of the Saccharomyces cerevisiae Rad2 family nucleases in Okazaki fragment maturation, mutation avoidance, and chromosome stability. DNA Repair 2: 925–940. 12893088

58. Lewis LK, Karthikeyan G, Westmoreland JW, Resnick MA (2002) Differential suppression of DNA repair deficiencies of yeast rad50, mre11 and xrs2 mutants by EXO1 and TLC1 (the RNA component of telomerase). Genetics 160: 49–62. 11805044

59. Moreau S, Morgan EA, Symington LS (2001) Overlapping functions of the Saccharomyces cerevisiae Mre11, Exo1 and Rad27 nucleases in DNA metabolism. Genetics 159: 1423–1433. 11779786

60. Tsutakawa SE, Classen S, Chapados BR, Arvai AS, Finger LD, et al. (2011) Human flap endonuclease structures, DNA double-base flipping, and a unified understanding of the FEN1 superfamily. Cell 145: 198–211. doi: 10.1016/j.cell.2011.03.004 21496641

61. Orans J, McSweeney EA, Iyer RR, Hast MA, Hellinga HW, et al. (2011) Structures of human exonuclease 1 DNA complexes suggest a unified mechanism for nuclease family. Cell 145: 212–223. doi: 10.1016/j.cell.2011.03.005 21496642

62. Miętus M, Nowak E, Jaciuk M, Kustosz P, Studnicka J, et al. (2014) Crystal structure of the catalytic core of Rad2: insights into the mechanism of substrate binding. Nucleic Acids Research 42: 10762–10775. doi: 10.1093/nar/gku729 25120270

63. Tran PT, Erdeniz N, Dudley S, Liskay RM (2002) Characterization of nuclease-dependent functions of Exo1p in Saccharomyces cerevisiae. DNA Repair 1: 895–912. 12531018

64. Gutiérrez PJ, Wang TS-F (2003) Genomic instability induced by mutations in Saccharomyces cerevisiae POL1. Genetics 165: 65–81. 14504218

65. Fumasoni M, Zwicky K, Vanoli F, Lopes M, Branzei D (2015) Error-free DNA damage tolerance and sister chromatid proximity during DNA replication rely on the Polα/Primase/Ctf4 complex. Molecular Cell 57: 812–823. doi: 10.1016/j.molcel.2014.12.038 25661486

66. Suzuki M, Niimi A, Limsirichaikul S, Tomida S, Huang QM, et al. (2009) PCNA mono-ubiquitination and activation of translesion DNA polymerases by DNA polymerase α. Journal of Biochemistry 146: 13–21. doi: 10.1093/jb/mvp043 19279190

67. Tong AHY, Evangelista M, Parsons AB, Xu H, Bader GD, et al. (2001) Systematic genetic analysis with ordered arrays of yeast deletion mutants. Science 294: 2364–2368. 11743205

68. Nagai S, Dubrana K, Tsai-Pflugfelder M, Davidson MB, Roberts TM, et al. (2008) Functional targeting of DNA damage to a nuclear pore-associated SUMO-dependent ubiquitin ligase. Science 322: 597–602. doi: 10.1126/science.1162790 18948542

69. Budd ME, Antoshechkin IA, Reis C, Wold BJ, Campbell JL (2011) Inviability of a DNA2 deletion mutant is due to the DNA damage checkpoint. Cell Cycle 10: 1690–1698. 21508669

70. Budd ME, Tong AHY, Polaczek P, Peng X, Boone C, et al. (2005) A network of multi-tasking proteins at the DNA replication fork preserves genome stability. PLoS Genetics 1: e61. 16327883

71. Freudenreich CH, Kantrow SM, Zakian VA (1998) Expansion and length-dependent fragility of CTG repeats in yeast. Science 279: 853–856. 9452383

72. Callahan JL, Andrews KJ, Zakian VA, Freudenreich CH (2003) Mutations in yeast replication proteins that increase CAG/CTG expansions also increase repeat fragility. Molecular and Cellular Biology 23: 7849–7860. 14560028

73. Daee DL, Mertz T, Lahue RS (2007) Postreplication repair inhibits CAG· CTG repeat expansions in Saccharomyces cerevisiae. Molecular and Cellular Biology 27: 102–110. 17060452

74. Collins NS, Bhattacharyya S, Lahue RS (2007) Rev1 enhances CAG· CTG repeat stability in Saccharomyces cerevisiae. DNA Repair 6: 38–44. 16979389

75. Ben-Aroya S, Koren A, Liefshitz B, Steinlauf R, Kupiec M (2003) ELG1, a yeast gene required for genome stability, forms a complex related to replication factor C. Proceedings of the National Academy of Sciences 100: 9906–9911.

76. Smith S, Hwang J-Y, Banerjee S, Majeed A, Gupta A, et al. (2004) Mutator genes for suppression of gross chromosomal rearrangements identified by a genome-wide screening in Saccharomyces cerevisiae. Proceedings of the National Academy of Sciences of the United States of America 101: 9039–9044. 15184655

77. Kanellis P, Agyei R, Durocher D (2003) Elg1 forms an alternative PCNA-interacting RFC complex required to maintain genome stability. Current Biology 13: 1583–1595. 13678589

78. Davidson MB, Katou Y, Keszthelyi A, Sing TL, Xia T, et al. (2012) Endogenous DNA replication stress results in expansion of dNTP pools and a mutator phenotype. The EMBO Journal 31: 895–907. doi: 10.1038/emboj.2011.485 22234187

79. Yu C, Gan H, Han J, Zhou Z-X, Jia S, et al. (2014) Strand-specific analysis shows protein binding at replication forks and PCNA unloading from lagging strands when forks stall. Molecular Cell 56: 551–563. doi: 10.1016/j.molcel.2014.09.017 25449133

80. Balakrishnan L, Stewart J, Polaczek P, Campbell JL, Bambara RA (2010) Acetylation of Dna2 endonuclease/helicase and flap endonuclease 1 by p300 promotes DNA stability by creating long flap intermediates. Journal of Biological Chemistry 285: 4398–4404. doi: 10.1074/jbc.M109.086397 20019387

81. Kao H-I, Veeraraghavan J, Polaczek P, Campbell JL, Bambara RA (2004) On the roles of Saccharomyces cerevisiae Dna2p and Flap endonuclease 1 in Okazaki fragment processing. Journal of Biological Chemistry 279: 15014–15024. 14747468

82. Rossi ML, Bambara RA (2006) Reconstituted Okazaki fragment processing indicates two pathways of primer removal. Journal of Biological Chemistry 281: 26051–26061. 16837458

83. Kucherlapati M, Yang K, Kuraguchi M, Zhao J, Lia M, et al. (2002) Haploinsufficiency of Flap endonuclease (Fen1) leads to rapid tumor progression. Proceedings of the National Academy of Sciences 99: 9924–9929.

84. Zheng L, Dai H, Zhou M, Li M, Singh P, et al. (2007) Fen1 mutations result in autoimmunity, chronic inflammation and cancers. Nature Medicine 13: 812–819. 17589521

85. Tran HT, Keen JD, Kricker M, Resnick MA, Gordenin DA (1997) Hypermutability of homonucleotide runs in mismatch repair and DNA polymerase proofreading yeast mutants. Molecular and Cellular Biology 17: 2859–2865. 9111358

86. Lorenz MC, Muir RS, Lim E, McElver J, Weber SC, et al. (1995) Gene disruption with PCR products in Saccharomyces cerevisiae. Gene 158: 113–117. 7789793

87. Parenteau J, Wellinger RJ (1999) Accumulation of single-stranded DNA and destabilization of telomeric repeats in yeast mutant strains carrying a deletion of RAD27. Molecular and Cellular Biology 19: 4143–4152. 10330154

88. Ulrich HD (2009) SUMO protocols: Springer.

89. Ricke RM, Bielinsky A-K (2006) A conserved Hsp10-like domain in Mcm10 is required to stabilize the catalytic subunit of DNA polymerase-α in budding yeast. Journal of Biological Chemistry 281: 18414–18425. 16675460

90. Whelan WL, Gocke E, Manney TR (1979) The CAN1 locus of Saccharomyces cerevisiae: fine-structure analysis and forward mutation rates. Genetics 91: 35–51. 372045

91. Drake JW (1991) A constant rate of spontaneous mutation in DNA-based microbes. Proceedings of the National Academy of Sciences 88: 7160–7164.

92. Foster PL (2006) Methods for determining spontaneous mutation rates. Methods in Enzymology 409: 195–213. 16793403

93. Ricke RM, Bielinsky A-K (2004) Mcm10 regulates the stability and chromatin association of DNA polymerase-α. Molecular Cell 16: 173–185. 15494305

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2015 Číslo 11
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#