Skip to main content

Role of Protein Linked DNA Breaks in Cancer

  • Chapter
  • First Online:

Part of the book series: Advances in Experimental Medicine and Biology ((AEMB,volume 1007))

Abstract

Topoisomerases are a group of specialized enzymes that function to maintain DNA topology by introducing transient DNA breaks during transcription and replication. As a result of abortive topoisomerases activity, topoisomerases catalytic intermediates may be trapped on the DNA forming topoisomerase cleavage complexes (Topcc). Topoisomerases trapping on the DNA is the mode of action of several anticancer drugs, it lead to formation of protein linked DAN breaks (PDBs). PDBs are now considered as one of the most dangerous forms of endogenous DNA damage and a major threat to genomic stability. The repair of PDBs involves both the sensing and repair pathways. Unsuccessful repair of PDBs leads to different signs of genomic instabilities such as chromosomal rearrangements and cancer predisposition. In this chapter we will summarize the role of topoisomerases induced PDBs, identification and signaling, repair, role in transcription. We will also discuss the role of PDBs in cancer with a special focus on prostate cancer.

This is a preview of subscription content, log in via an institution.

Buying options

Chapter
USD   29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD   139.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD   179.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD   179.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Learn about institutional subscriptions

References

  1. Schilsky RL (1996) Methotrexate: an effective agent for treating cancer and building careers. The polyglutamate era. Stem Cells 14:29–32. doi:10.1002/stem.140029

    Article  CAS  PubMed  Google Scholar 

  2. Miura S, Yoshimura Y, Satoh H, Izuta S (2001) The antitumor mechanism of 1-(2-deoxy-2-fluoro-4-thio-beta-D-arabinofuranosyl)-cytosine: effects of its triphosphate on mammalian DNA polymerases. Jpn J Cancer Res 92:562–567

    Article  CAS  PubMed  Google Scholar 

  3. Povirk LF (1996) DNA damage and mutagenesis by radiomimetic DNA-cleaving agents: bleomycin, neocarzinostatin and other enediynes. Mutat Res 355:71–89

    Article  PubMed  Google Scholar 

  4. Zorbas H, Keppler BK (2005) Cisplatin damage: are DNA repair proteins saviors or traitors to the cell? Chembiochem 6:1157–1166. doi:10.1002/cbic.200400427

    Article  CAS  PubMed  Google Scholar 

  5. Chen T, Stephens PA, Middleton FK, Curtin NJ (2012) Targeting the S and G2 checkpoint to treat cancer. Drug Discov Today 17:194–202. doi:10.1016/j.drudis.2011.12.009

    Article  CAS  PubMed  Google Scholar 

  6. Biss M, Xiao W (2012) Selective tumor killing based on specific DNA-damage response deficiencies. Cancer Biol Ther 13:239–246. doi:10.4161/cbt.18921

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yang X, Wood PA, Hrushesky WJ (2010) Mammalian TIMELESS is required for ATM-dependent CHK2 activation and G2/M checkpoint control. J Biol Chem 285:3030–3034. doi:10.1074/jbc.M109.050237

    Article  CAS  PubMed  Google Scholar 

  8. Bakkenist CJ, Kastan MB (2003) DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 421:499–506. doi:10.1038/nature01368

    Article  CAS  PubMed  Google Scholar 

  9. Lee JH, Paull TT (2007) Activation and regulation of ATM kinase activity in response to DNA double-strand breaks. Oncogene 26:7741–7748. doi:10.1038/sj.onc.1210872

    Article  CAS  PubMed  Google Scholar 

  10. Buscemi G et al (2001) Chk2 activation dependence on Nbs1 after DNA damage. Mol Cell Biol 21:5214–5222. doi:10.1128/MCB.21.15.5214-5222.2001

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Nakanishi K et al (2002) Interaction of FANCD2 and NBS1 in the DNA damage response. Nat Cell Biol 4:913–920. doi:10.1038/ncb879

    Article  CAS  PubMed  Google Scholar 

  12. Uziel T et al (2003) Requirement of the MRN complex for ATM activation by DNA damage. EMBO J 22:5612–5621. doi:10.1093/emboj/cdg541

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Lee JH, Paull TT (2005) ATM activation by DNA double-strand breaks through the Mre11-Rad50-Nbs1 complex. Science 308:551–554. doi:10.1126/science.1108297

    Article  CAS  PubMed  Google Scholar 

  14. Woods D, Turchi JJ (2013) Chemotherapy induced DNA damage response: convergence of drugs and pathways. Cancer Biol Ther 14:379–389. doi:10.4161/cbt.23761

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Liu X et al (2012) Overlapping functions between XLF repair protein and 53BP1 DNA damage response factor in end joining and lymphocyte development. Proc Natl Acad Sci U S A 109:3903–3908. doi:10.1073/pnas.1120160109

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. McGowan CH, Russell P (2004) The DNA damage response: sensing and signaling. Curr Opin Cell Biol 16:629–633. doi:10.1016/j.ceb.2004.09.005

    Article  CAS  PubMed  Google Scholar 

  17. Harrison JC, Haber JE (2006) Surviving the breakup: the DNA damage checkpoint. Annu Rev Genet 40:209–235. doi:10.1146/annurev.genet.40.051206.105231

    Article  CAS  PubMed  Google Scholar 

  18. Stewart L, Redinbo MR, Qiu X, Hol WG, Champoux JJ (1998) A model for the mechanism of human topoisomerase I. Science 279:1534–1541

    Article  CAS  PubMed  Google Scholar 

  19. Staker BL et al (2002) The mechanism of topoisomerase I poisoning by a camptothecin analog. Proc Natl Acad Sci U S A 99:15387–15392. doi:10.1073/pnas.242259599

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wang JC (2002) Cellular roles of DNA topoisomerases: a molecular perspective. Nat Rev Mol Cell Biol 3:430–440. doi:10.1038/nrm831

    Article  CAS  PubMed  Google Scholar 

  21. Flatten K et al (2005) The role of checkpoint kinase 1 in sensitivity to topoisomerase I poisons. J Biol Chem 280:14349–14355. doi:10.1074/jbc.M411890200

    Article  CAS  PubMed  Google Scholar 

  22. Wu CS et al (2014) SUMOylation of ATRIP potentiates DNA damage signaling by boosting multiple protein interactions in the ATR pathway. Genes Dev 28:1472–1484. doi:10.1101/gad.238535.114

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wu CS, Zou L (2016) The SUMO (Small Ubiquitin-like Modifier) ligase PIAS3 primes ATR for checkpoint activation. J Biol Chem 291:279–290. doi:10.1074/jbc.M115.691170

    Article  CAS  PubMed  Google Scholar 

  24. Liu S et al (2013) PIAS3 promotes homology-directed repair and distal non-homologous end joining. Oncol Lett 6:1045–1048. doi:10.3892/ol.2013.1472

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Sakasai R, Teraoka H, Tibbetts RS (2010) Proteasome inhibition suppresses DNA-dependent protein kinase activation caused by camptothecin. DNA Repair (Amst) 9:76–82. doi:10.1016/j.dnarep.2009.10.008

    Article  CAS  Google Scholar 

  26. Kocher S, Spies-Naumann A, Kriegs M, Dahm-Daphi J, Dornreiter I (2013) ATM is required for the repair of Topotecan-induced replication-associated double-strand breaks. Radiother Oncol 108:409–414. doi:10.1016/j.radonc.2013.06.024

    Article  PubMed  CAS  Google Scholar 

  27. Serrano MA et al (2013) DNA-PK, ATM and ATR collaboratively regulate p53-RPA interaction to facilitate homologous recombination DNA repair. Oncogene 32:2452–2462. doi:10.1038/onc.2012.257

    Article  CAS  PubMed  Google Scholar 

  28. Das BB et al (2009) Optimal function of the DNA repair enzyme TDP1 requires its phosphorylation by ATM and/or DNA-PK. EMBO J 28:3667–3680. doi:10.1038/emboj.2009.302

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Chiang SC, Carroll J, El-Khamisy SF (2010) TDP1 serine 81 promotes interaction with DNA ligase IIIalpha and facilitates cell survival following DNA damage. Cell Cycle 9:588–595. doi:10.4161/cc.9.3.10598

    Article  CAS  PubMed  Google Scholar 

  30. Zhou Y et al (2017) Regulation of the DNA damage response by DNA-PKcs inhibitory phosphorylation of ATM. Mol Cell 65:91–104. doi:10.1016/j.molcel.2016.11.004

    Article  CAS  PubMed  Google Scholar 

  31. Sordet O et al (2009) Ataxia telangiectasia mutated activation by transcription- and topoisomerase I-induced DNA double-strand breaks. EMBO Rep 10:887–893. doi:10.1038/embor.2009.97

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Cristini A et al (2016) DNA-PK triggers histone ubiquitination and signaling in response to DNA double-strand breaks produced during the repair of transcription-blocking topoisomerase I lesions. Nucleic Acids Res 44:1161–1178. doi:10.1093/nar/gkv1196

    Article  CAS  PubMed  Google Scholar 

  33. Alagoz M, Chiang SC, Sharma A, El-Khamisy SF (2013) ATM deficiency results in accumulation of DNA-topoisomerase I covalent intermediates in neural cells. PLoS One 8:e58239. doi:10.1371/journal.pone.0058239

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Huelsenbeck SC et al (2012) Rac1 protein signaling is required for DNA damage response stimulated by topoisomerase II poisons. J Biol Chem 287:38590–38599. doi:10.1074/jbc.M112.377903

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Forrest RA et al (2012) Activation of DNA damage response pathways as a consequence of anthracycline-DNA adduct formation. Biochem Pharmacol 83:1602–1612. doi:10.1016/j.bcp.2012.02.026

    Article  CAS  PubMed  Google Scholar 

  36. Maede Y et al (2014) Differential and common DNA repair pathways for topoisomerase I- and II-targeted drugs in a genetic DT40 repair cell screen panel. Mol Cancer Ther 13:214–220. doi:10.1158/1535-7163.MCT-13-0551

    Article  CAS  PubMed  Google Scholar 

  37. Rossi R, Lidonnici MR, Soza S, Biamonti G, Montecucco A (2006) The dispersal of replication proteins after Etoposide treatment requires the cooperation of Nbs1 with the ataxia telangiectasia Rad3-related/Chk1 pathway. Cancer Res 66:1675–1683. doi:10.1158/0008-5472.CAN-05-2741

    Article  CAS  PubMed  Google Scholar 

  38. Quennet V, Beucher A, Barton O, Takeda S, Lobrich M (2011) CtIP and MRN promote non-homologous end-joining of etoposide-induced DNA double-strand breaks in G1. Nucleic Acids Res 39:2144–2152. doi:10.1093/nar/gkq1175

    Article  CAS  PubMed  Google Scholar 

  39. Alvarez-Quilon A et al (2014) ATM specifically mediates repair of double-strand breaks with blocked DNA ends. Nat Commun 5:3347. doi:10.1038/ncomms4347

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Korwek Z et al (2012) Inhibition of ATM blocks the etoposide-induced DNA damage response and apoptosis of resting human T cells. DNA Repair (Amst) 11:864–873. doi:10.1016/j.dnarep.2012.08.006

    Article  CAS  Google Scholar 

  41. Cliby WA, Lewis KA, Lilly KK, Kaufmann SH (2002) S phase and G2 arrests induced by topoisomerase I poisons are dependent on ATR kinase function. J Biol Chem 277:1599–1606. doi:10.1074/jbc.M106287200

    Article  CAS  PubMed  Google Scholar 

  42. Costanzo V et al (2003) An ATR- and Cdc7-dependent DNA damage checkpoint that inhibits initiation of DNA replication. Mol Cell 11:203–213

    Article  CAS  PubMed  Google Scholar 

  43. Siu WY et al (2004) Topoisomerase poisons differentially activate DNA damage checkpoints through ataxia-telangiectasia mutated-dependent and -independent mechanisms. Mol Cancer Ther 3:621–632

    CAS  PubMed  Google Scholar 

  44. Ho CC et al (2005) The relative contribution of CHK1 and CHK2 to Adriamycin-induced checkpoint. Exp Cell Res 304:1–15. doi:10.1016/j.yexcr.2004.10.016

    Article  CAS  PubMed  Google Scholar 

  45. Martensson S, Nygren J, Osheroff N, Hammarsten O (2003) Activation of the DNA-dependent protein kinase by drug-induced and radiation-induced DNA strand breaks. Radiat Res 160:291–301

    Article  CAS  PubMed  Google Scholar 

  46. Das BB et al (2014) PARP1-TDP1 coupling for the repair of topoisomerase I-induced DNA damage. Nucleic Acids Res 42:4435–4449. doi:10.1093/nar/gku088

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Das SK et al (2016) Poly(ADP-ribose) polymers regulate DNA topoisomerase I (Top1) nuclear dynamics and camptothecin sensitivity in living cells. Nucleic Acids Res 44:8363–8375. doi:10.1093/nar/gkw665

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Cuya SM, Comeaux EQ, Wanzeck K, Yoon KJ, van Waardenburg RC (2016) Dysregulated human Tyrosyl-DNA phosphodiesterase I acts as cellular toxin. Oncotarget 7:86660–86674. doi:10.18632/oncotarget.13528

    PubMed  PubMed Central  Google Scholar 

  49. Takashima H et al (2002) Mutation of TDP1, encoding a topoisomerase I-dependent DNA damage repair enzyme, in spinocerebellar ataxia with axonal neuropathy. Nat Genet 32:267–272. doi:10.1038/ng987

    Article  CAS  PubMed  Google Scholar 

  50. He X et al (2007) Mutation of a conserved active site residue converts tyrosyl-DNA phosphodiesterase I into a DNA topoisomerase I-dependent poison. J Mol Biol 372:1070–1081. doi:10.1016/j.jmb.2007.07.055

    Article  CAS  PubMed  Google Scholar 

  51. Huang SY et al (2013) TDP1 repairs nuclear and mitochondrial DNA damage induced by chain-terminating anticancer and antiviral nucleoside analogs. Nucleic Acids Res 41:7793–7803. doi:10.1093/nar/gkt483

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hudson JJ, Chiang SC, Wells OS, Rookyard C, El-Khamisy SF (2012) SUMO modification of the neuroprotective protein TDP1 facilitates chromosomal single-strand break repair. Nat Commun 3:733. doi:10.1038/ncomms1739

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. El-Khamisy SF, Hartsuiker E, Caldecott KW (2007) TDP1 facilitates repair of ionizing radiation-induced DNA single-strand breaks. DNA Repair 6:1485–1495. doi:10.1016/j.dnarep.2007.04.015

    Article  CAS  PubMed  Google Scholar 

  54. Alagoz M, Wells OS, El-Khamisy SF (2014) TDP1 deficiency sensitizes human cells to base damage via distinct topoisomerase I and PARP mechanisms with potential applications for cancer therapy. Nucleic Acids Res 42:3089–3103. doi:10.1093/nar/gkt1260

    Article  CAS  PubMed  Google Scholar 

  55. Davies DR, Interthal H, Champoux JJ, Hol WG (2002) The crystal structure of human tyrosyl-DNA phosphodiesterase, Tdp1. Structure 10:237–248

    Article  CAS  PubMed  Google Scholar 

  56. Comeaux EQ, van Waardenburg RC (2014) Tyrosyl-DNA phosphodiesterase I resolves both naturally and chemically induced DNA adducts and its potential as a therapeutic target. Drug Metab Rev 46:494–507. doi:10.3109/03602532.2014.971957

    Article  CAS  PubMed  Google Scholar 

  57. Malik M, Nitiss KC, Enriquez-Rios V, Nitiss JL (2006) Roles of nonhomologous end-joining pathways in surviving topoisomerase II-mediated DNA damage. Mol Cancer Ther 5:1405–1414. doi:10.1158/1535-7163.MCT-05-0263

    Article  CAS  PubMed  Google Scholar 

  58. Gomez-Herreros F et al (2013) TDP2-dependent non-homologous end-joining protects against topoisomerase II-induced DNA breaks and genome instability in cells and in vivo. PLoS Genet 9:e1003226. doi:10.1371/journal.pgen.1003226

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Zeng Z et al (2012) TDP2 promotes repair of topoisomerase I-mediated DNA damage in the absence of TDP1. Nucleic Acids Res 40:8371–8380. doi:10.1093/nar/gks622

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Bian K et al (2016) ERK3 regulates TDP2-mediated DNA damage response and chemoresistance in lung cancer cells. Oncotarget 7:6665–6675. doi:10.18632/oncotarget.6682

  61. Pommier Y et al (2014) Tyrosyl-DNA-phosphodiesterases (TDP1 and TDP2). DNA Repair 19:114–129. doi:10.1016/j.dnarep.2014.03.020

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Gao R, Huang SY, Marchand C, Pommier Y (2012) Biochemical characterization of human tyrosyl-DNA phosphodiesterase 2 (TDP2/TTRAP): a Mg(2+)/Mn(2+)-dependent phosphodiesterase specific for the repair of topoisomerase cleavage complexes. J Biol Chem 287:30842–30852. doi:10.1074/jbc.M112.393983

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Gao R et al (2014) Proteolytic degradation of topoisomerase II (Top2) enables the processing of Top2.DNA and Top2.RNA covalent complexes by tyrosyl-DNA-phosphodiesterase 2 (TDP2). J Biol Chem 289:17960–17969. doi:10.1074/jbc.M114.565374

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Heo J et al (2015) TDP1 promotes assembly of non-homologous end joining protein complexes on DNA. DNA Repair 30:28–37. doi:10.1016/j.dnarep.2015.03.003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Elsayed W, El-Shafie L, Hassan MK, Farag MA, El-Khamisy SF (2016) Isoeugenol is a selective potentiator of camptothecin cytotoxicity in vertebrate cells lacking TDP1. Sci Rep 6:26626. doi:10.1038/srep26626

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Beck DE et al (2016) Synthesis and biological evaluation of new fluorinated and chlorinated indenoisoquinoline topoisomerase I poisons. Bioorg Med Chem 24:1469–1479. doi:10.1016/j.bmc.2016.02.015

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Liao S, Tammaro M, Yan H (2016) The structure of ends determines the pathway choice and Mre11 nuclease dependency of DNA double-strand break repair. Nucleic Acids Res 44:5689–5701. doi:10.1093/nar/gkw274

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Aparicio T, Baer R, Gottesman M, Gautier J (2016) MRN, CtIP, and BRCA1 mediate repair of topoisomerase II-DNA adducts. J Cell Biol 212:399–408. doi:10.1083/jcb.201504005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Wang J et al (2016) Loss of CtIP disturbs homologous recombination repair and sensitizes breast cancer cells to PARP inhibitors. Oncotarget 7:7701–7714. doi:10.18632/oncotarget.6715

  70. Chanut P, Britton S, Coates J, Jackson SP, Calsou P (2016) Coordinated nuclease activities counteract Ku at single-ended DNA double-strand breaks. Nat Commun 7:12889. doi:10.1038/ncomms12889

    Article  PubMed  PubMed Central  Google Scholar 

  71. Guirouilh-Barbat J et al (2016) 53BP1 protects against CtIP-dependent capture of ectopic chromosomal sequences at the junction of distant double-strand breaks. PLoS Genet 12:e1006230. doi:10.1371/journal.pgen.1006230

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. Ahrabi S et al (2016) A role for human homologous recombination factors in suppressing microhomology-mediated end joining. Nucleic Acids Res 44:5743–5757. doi:10.1093/nar/gkw326

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Anand R, Ranjha L, Cannavo E, Cejka P (2016) Phosphorylated CtIP functions as a co-factor of the MRE11-RAD50-NBS1 endonuclease in DNA end resection. Mol Cell 64:940–950. doi:10.1016/j.molcel.2016.10.017

    Article  CAS  PubMed  Google Scholar 

  74. Chen YJ et al (2016) S. cerevisiae Mre11 recruits conjugated SUMO moieties to facilitate the assembly and function of the Mre11-Rad50-Xrs2 complex. Nucleic Acids Res 44:2199–2213. doi:10.1093/nar/gkv1523

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Deshpande RA, Lee JH, Arora S, Paull TT (2016) Nbs1 converts the human Mre11/Rad50 nuclease complex into an endo/exonuclease machine specific for protein-DNA adducts. Mol Cell 64:593–606. doi:10.1016/j.molcel.2016.10.010

    Article  CAS  PubMed  Google Scholar 

  76. Broderick R et al (2016) EXD2 promotes homologous recombination by facilitating DNA end resection. Nat Cell Biol 18:271–280. doi:10.1038/ncb3303

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Keyamura K, Arai K, Hishida T (2016) Srs2 and Mus81-Mms4 prevent accumulation of toxic inter-homolog recombination intermediates. PLoS Genet 12:e1006136. doi:10.1371/journal.pgen.1006136

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Higashide M, Shinohara M (2016) Budding yeast SLX4 contributes to the appropriate distribution of crossovers and meiotic double-strand break formation on bivalents during meiosis. G3 (Bethesda) 6:2033–2042. doi:10.1534/g3.116.029488

  79. Dibitetto D et al (2016) Slx4 and Rtt107 control checkpoint signalling and DNA resection at double-strand breaks. Nucleic Acids Res 44:669–682. doi:10.1093/nar/gkv1080

    Article  CAS  PubMed  Google Scholar 

  80. Guervilly JH, Gaillard PH (2016) SLX4 gains weight with SUMO in genome maintenance. Mol Cell Oncol 3:e1008297. doi:10.1080/23723556.2015.1008297

    Article  PubMed  CAS  Google Scholar 

  81. Regairaz M et al (2011) Mus81-mediated DNA cleavage resolves replication forks stalled by topoisomerase I-DNA complexes. J Cell Biol 195:739–749. doi:10.1083/jcb.201104003

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Sebesta M et al (2017) Esc2 promotes Mus81 complex-activity via its SUMO-like and DNA binding domains. Nucleic Acids Res 45:215–230. doi:10.1093/nar/gkw882

    Article  PubMed  Google Scholar 

  83. Zhang YW et al (2011) Poly(ADP-ribose) polymerase and XPF-ERCC1 participate in distinct pathways for the repair of topoisomerase I-induced DNA damage in mammalian cells. Nucleic Acids Res 39:3607–3620. doi:10.1093/nar/gkq1304

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. LoRusso PM et al (2016) Phase I safety, pharmacokinetic, and pharmacodynamic study of the poly(ADP-ribose) polymerase (PARP) inhibitor veliparib (ABT-888) in combination with irinotecan in patients with advanced solid tumors. Clin Cancer Res 22:3227–3237. doi:10.1158/1078-0432.CCR-15-0652

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Mullen JR, Chen CF, Brill SJ (2010) Wss1 is a SUMO-dependent isopeptidase that interacts genetically with the Slx5-Slx8 SUMO-targeted ubiquitin ligase. Mol Cell Biol 30:3737–3748. doi:10.1128/MCB.01649-09

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. O’Neill BM, Hanway D, Winzeler EA, Romesberg FE (2004) Coordinated functions of WSS1, PSY2 and TOF1 in the DNA damage response. Nucleic Acids Res 32:6519–6530. doi:10.1093/nar/gkh994

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  87. Stingele J, Schwarz MS, Bloemeke N, Wolf PG, Jentsch S (2014) A DNA-dependent protease involved in DNA-protein crosslink repair. Cell 158:327–338. doi:10.1016/j.cell.2014.04.053

    Article  CAS  PubMed  Google Scholar 

  88. Balakirev MY et al (2015) Wss1 metalloprotease partners with Cdc48/Doa1 in processing genotoxic SUMO conjugates. Elife 4. doi:10.7554/eLife.06763

  89. Duxin JP, Dewar JM, Yardimci H, Walter JC (2014) Repair of a DNA-protein crosslink by replication-coupled proteolysis. Cell 159:346–357. doi:10.1016/j.cell.2014.09.024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Stingele J et al (2016) Mechanism and regulation of DNA-protein crosslink repair by the DNA-dependent metalloprotease SPRTN. Mol Cell 64:688–703. doi:10.1016/j.molcel.2016.09.031

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Lopez-Mosqueda J et al (2016) SPRTN is a mammalian DNA-binding metalloprotease that resolves DNA-protein crosslinks. Elife 5. doi:10.7554/eLife.21491

  92. Lessel D et al (2014) Mutations in SPRTN cause early onset hepatocellular carcinoma, genomic instability and progeroid features. Nat Genet 46:1239–1244. doi:10.1038/ng.3103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Vaz B et al (2016) Metalloprotease SPRTN/DVC1 orchestrates replication-coupled DNA-protein crosslink repair. Mol Cell 64:704–719. doi:10.1016/j.molcel.2016.09.032

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Mosbech A et al (2012) DVC1 (C1orf124) is a DNA damage-targeting p97 adaptor that promotes ubiquitin-dependent responses to replication blocks. Nat Struct Mol Biol 19:1084–1092. doi:10.1038/nsmb.2395

    Article  CAS  PubMed  Google Scholar 

  95. Morocz M et al (2017) DNA-dependent protease activity of human Spartan facilitates replication of DNA-protein crosslink-containing DNA. Nucleic Acids Res. doi:10.1093/nar/gkw1315

  96. Ray Chaudhuri A et al (2012) Topoisomerase I poisoning results in PARP-mediated replication fork reversal. Nat Struct Mol Biol 19:417–423. doi:10.1038/nsmb.2258

    Article  CAS  PubMed  Google Scholar 

  97. Solier S et al (2013) Transcription poisoning by Topoisomerase I is controlled by gene length, splice sites, and miR-142-3p. Cancer Res 73:4830–4839. doi:10.1158/0008-5472.CAN-12-3504

    Article  CAS  PubMed  Google Scholar 

  98. Berti M et al (2013) Human RECQ1 promotes restart of replication forks reversed by DNA topoisomerase I inhibition. Nat Struct Mol Biol 20:347–354. doi:10.1038/nsmb.2501

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Zellweger R et al (2015) Rad51-mediated replication fork reversal is a global response to genotoxic treatments in human cells. J Cell Biol 208:563–579. doi:10.1083/jcb.201406099

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Hashimoto Y, Puddu F, Costanzo V (2011) RAD51- and MRE11-dependent reassembly of uncoupled CMG helicase complex at collapsed replication forks. Nat Struct Mol Biol 19:17–24. doi:10.1038/nsmb.2177

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Petermann E, Orta ML, Issaeva N, Schultz N, Helleday T (2010) Hydroxyurea-stalled replication forks become progressively inactivated and require two different RAD51-mediated pathways for restart and repair. Mol Cell 37:492–502. doi:10.1016/j.molcel.2010.01.021

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Hu J et al (2012) The intra-S phase checkpoint targets Dna2 to prevent stalled replication forks from reversing. Cell 149:1221–1232. doi:10.1016/j.cell.2012.04.030

    Article  CAS  PubMed  Google Scholar 

  103. Ying S, Hamdy FC, Helleday T (2012) Mre11-dependent degradation of stalled DNA replication forks is prevented by BRCA2 and PARP1. Cancer Res 72:2814–2821. doi:10.1158/0008-5472.CAN-11-3417

    Article  CAS  PubMed  Google Scholar 

  104. Schlacher K et al (2011) Double-strand break repair-independent role for BRCA2 in blocking stalled replication fork degradation by MRE11. Cell 145:529–542. doi:10.1016/j.cell.2011.03.041

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Shiotani B et al (2013) Two distinct modes of ATR activation orchestrated by Rad17 and Nbs1. Cell Rep 3:1651–1662. doi:10.1016/j.celrep.2013.04.018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Murina O et al (2014) FANCD2 and CtIP cooperate to repair DNA interstrand crosslinks. Cell Rep 7:1030–1038. doi:10.1016/j.celrep.2014.03.069

    Article  CAS  PubMed  Google Scholar 

  107. Yeo JE, Lee EH, Hendrickson EA, Sobeck A (2014) CtIP mediates replication fork recovery in a FANCD2-regulated manner. Hum Mol Genet 23:3695–3705. doi:10.1093/hmg/ddu078

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Kuo C, Nuang H, Campbell JL (1983) Isolation of yeast DNA replication mutants in permeabilized cells. Proc Natl Acad Sci U S A 80:6465–6469

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Budd ME, Campbell JL (1995) A yeast gene required for DNA replication encodes a protein with homology to DNA helicases. Proc Natl Acad Sci U S A 92:7642–7646

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Gravel S, Chapman JR, Magill C, Jackson SP (2008) DNA helicases Sgs1 and BLM promote DNA double-strand break resection. Genes Dev 22:2767–2772. doi:10.1101/gad.503108

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Zhu Z, Chung WH, Shim EY, Lee SE, Ira G (2008) Sgs1 helicase and two nucleases Dna2 and Exo1 resect DNA double-strand break ends. Cell 134:981–994. doi:10.1016/j.cell.2008.08.037

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Nicolette ML et al (2010) Mre11-Rad50-Xrs2 and Sae2 promote 5′ strand resection of DNA double-strand breaks. Nat Struct Mol Biol 17:1478–1485. doi:10.1038/nsmb.1957

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Nimonkar AV et al (2011) BLM-DNA2-RPA-MRN and EXO1-BLM-RPA-MRN constitute two DNA end resection machineries for human DNA break repair. Genes Dev 25:350–362. doi:10.1101/gad.2003811

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Peng G et al (2012) Human nuclease/helicase DNA2 alleviates replication stress by promoting DNA end resection. Cancer Res 72:2802–2813. doi:10.1158/0008-5472.CAN-11-3152

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Thangavel S et al (2015) DNA2 drives processing and restart of reversed replication forks in human cells. J Cell Biol 208:545–562. doi:10.1083/jcb.201406100

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Iannascoli C, Palermo V, Murfuni I, Franchitto A, Pichierri P (2015) The WRN exonuclease domain protects nascent strands from pathological MRE11/EXO1-dependent degradation. Nucleic Acids Res 43:9788–9803. doi:10.1093/nar/gkv836

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Shamanna RA et al (2016) WRN regulates pathway choice between classical and alternative non-homologous end joining. Nat Commun 7:13785. doi:10.1038/ncomms13785

    Article  PubMed  PubMed Central  Google Scholar 

  118. Shamanna RA et al. (2016) Camptothecin targets WRN protein: mechanism and relevance in clinical breast cancer. Oncotarget 7:13269–13284. doi:10.18632/oncotarget.7906

  119. Ribeyre C et al (2016) Nascent DNA proteomics reveals a chromatin remodeler required for topoisomerase I loading at replication forks. Cell Rep 15:300–309. doi:10.1016/j.celrep.2016.03.027

    Article  CAS  PubMed  Google Scholar 

  120. Pommier Y, Kohlhagen G, Wu C, Simmons DT (1998) Mammalian DNA topoisomerase I activity and poisoning by camptothecin are inhibited by simian virus 40 large T antigen. Biochemistry 37:3818–3823. doi:10.1021/bi972067d

    Article  CAS  PubMed  Google Scholar 

  121. Seinsoth S, Uhlmann-Schiffler H, Stahl H (2003) Bidirectional DNA unwinding by a ternary complex of T antigen, nucleolin and topoisomerase I. EMBO Rep 4:263–268. doi:10.1038/sj.embor.embor770

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Gavin I, Horn PJ, Peterson CL (2001) SWI/SNF chromatin remodeling requires changes in DNA topology. Mol Cell 7:97–104

    Article  CAS  PubMed  Google Scholar 

  123. Walfridsson J, Khorosjutina O, Matikainen P, Gustafsson CM, Ekwall K (2007) A genome-wide role for CHD remodelling factors and Nap1 in nucleosome disassembly. EMBO J 26:2868–2879. doi:10.1038/sj.emboj.7601728

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Durand-Dubief M, Svensson JP, Persson J, Ekwall K (2011) Topoisomerases, chromatin and transcription termination. Transcription 2:66–70. doi:10.4161/trns.2.2.14411

    Article  PubMed  PubMed Central  Google Scholar 

  125. Krawczyk C, Dion V, Schar P, Fritsch O (2014) Reversible Top1 cleavage complexes are stabilized strand-specifically at the ribosomal replication fork barrier and contribute to ribosomal DNA stability. Nucleic Acids Res 42:4985–4995. doi:10.1093/nar/gku148

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Dykhuizen EC et al (2013) BAF complexes facilitate decatenation of DNA by topoisomerase IIalpha. Nature 497:624–627. doi:10.1038/nature12146

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Husain A et al (2016) Chromatin remodeller SMARCA4 recruits topoisomerase 1 and suppresses transcription-associated genomic instability. Nat Commun 7:10549. doi:10.1038/ncomms10549

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Stoll G et al (2013) Deletion of TOP3beta, a component of FMRP-containing mRNPs, contributes to neurodevelopmental disorders. Nat Neurosci 16:1228–1237. doi:10.1038/nn.3484

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Xu D et al (2013) Top3beta is an RNA topoisomerase that works with fragile X syndrome protein to promote synapse formation. Nat Neurosci 16:1238–1247. doi:10.1038/nn.3479

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Yang Y et al (2014) Arginine methylation facilitates the recruitment of TOP3B to chromatin to prevent R loop accumulation. Mol Cell 53:484–497. doi:10.1016/j.molcel.2014.01.011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Ahmad M et al (2016) RNA topoisomerase is prevalent in all domains of life and associates with polyribosomes in animals. Nucleic Acids Res 44:6335–6349. doi:10.1093/nar/gkw508

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Siaw GE, Liu IF, Lin PY, Been MD, Hsieh TS (2016) DNA and RNA topoisomerase activities of Top3beta are promoted by mediator protein Tudor domain-containing protein 3. Proc Natl Acad Sci U S A 113:E5544–E5551. doi:10.1073/pnas.1605517113

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Meisenberg C et al. (2016) Epigenetic changes in histone acetylation underpin resistance to the topoisomerase I inhibitor irinotecan. Nucleic Acids Res. doi:10.1093/nar/gkw1026

  134. Maxwell A, Costenaro L, Mitelheiser S, Bates AD (2005) Coupling ATP hydrolysis to DNA strand passage in type IIA DNA topoisomerases. Biochem Soc Trans 33:1460–1464. doi:10.1042/BST20051460

    Article  CAS  PubMed  Google Scholar 

  135. Joshi RS, Pina B, Roca J (2012) Topoisomerase II is required for the production of long Pol II gene transcripts in yeast. Nucleic Acids Res 40:7907–7915. doi:10.1093/nar/gks626

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. King IF et al (2013) Topoisomerases facilitate transcription of long genes linked to autism. Nature 501:58–62. doi:10.1038/nature12504

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Roedgaard M, Fredsoe J, Pedersen JM, Bjergbaek L, Andersen AH (2015) DNA topoisomerases are required for preinitiation complex assembly during GAL gene activation. PLoS One 10:e0132739. doi:10.1371/journal.pone.0132739

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  138. Ju BG et al (2006) A topoisomerase IIbeta-mediated dsDNA break required for regulated transcription. Science 312:1798–1802. doi:10.1126/science.1127196

    Article  CAS  PubMed  Google Scholar 

  139. Puc J et al (2015) Ligand-dependent enhancer activation regulated by topoisomerase-I activity. Cell 160:367–380. doi:10.1016/j.cell.2014.12.023

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Bunch H et al (2015) Transcriptional elongation requires DNA break-induced signalling. Nat Commun 6:10191. doi:10.1038/ncomms10191

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Madabhushi R et al (2015) Activity-induced DNA breaks govern the expression of neuronal early-response genes. Cell 161:1592–1605. doi:10.1016/j.cell.2015.05.032

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Williams JS, Lujan SA, Kunkel TA (2016) Processing ribonucleotides incorporated during eukaryotic DNA replication. Nat Rev Mol Cell Biol 17:350–363. doi:10.1038/nrm.2016.37

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Williams JS et al (2013) Topoisomerase 1-mediated removal of ribonucleotides from nascent leading-strand DNA. Mol Cell 49:1010–1015. doi:10.1016/j.molcel.2012.12.021

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Sparks JL, Burgers PM (2015) Error-free and mutagenic processing of topoisomerase 1-provoked damage at genomic ribonucleotides. EMBO J 34:1259–1269. doi:10.15252/embj.201490868

  145. Kim N et al (2011) Mutagenic processing of ribonucleotides in DNA by yeast topoisomerase I. Science 332:1561–1564. doi:10.1126/science.1205016

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Williams JS, Kunkel TA (2014) Ribonucleotides in DNA: origins, repair and consequences. DNA Repair (Amst) 19:27–37. doi:10.1016/j.dnarep.2014.03.029

    Article  CAS  PubMed Central  Google Scholar 

  147. Potenski CJ, Niu H, Sung P, Klein HL (2014) Avoidance of ribonucleotide-induced mutations by RNase H2 and Srs2-Exo1 mechanisms. Nature 511:251–254. doi:10.1038/nature13292

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Lazzaro F et al (2012) RNase H and postreplication repair protect cells from ribonucleotides incorporated in DNA. Mol Cell 45:99–110. doi:10.1016/j.molcel.2011.12.019

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Sekiguchi J, Shuman S (1997) Site-specific ribonuclease activity of eukaryotic DNA topoisomerase I. Mol Cell 1:89–97

    Article  CAS  PubMed  Google Scholar 

  150. Huang SY, Ghosh S, Pommier Y (2015) Topoisomerase I alone is sufficient to produce short DNA deletions and can also reverse nicks at ribonucleotide sites. J Biol Chem 290:14068–14076. doi:10.1074/jbc.M115.653345

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Arana ME et al (2012) Transcriptional responses to loss of RNase H2 in Saccharomyces cerevisiae. DNA Repair (Amst) 11:933–941. doi:10.1016/j.dnarep.2012.09.006

    Article  CAS  PubMed Central  Google Scholar 

  152. Conover HN et al (2015) Stimulation of chromosomal rearrangements by ribonucleotides. Genetics 201:951–961. doi:10.1534/genetics.115.181149

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Epshtein A, Potenski CJ, Klein HL (2016) Increased spontaneous recombination in RNase H2-deficient cells arises from multiple contiguous rNMPs and not from single rNMP residues incorporated by DNA polymerase epsilon. Microb Cell 3:248–254. doi:10.15698/mic2016.06.506

  154. Niu H, Potenski CJ, Epshtein A, Sung P, Klein HL (2016) Roles of DNA helicases and Exo1 in the avoidance of mutations induced by Top1-mediated cleavage at ribonucleotides in DNA. Cell Cycle 15:331–336. doi:10.1080/15384101.2015.1128594

    Article  CAS  PubMed  Google Scholar 

  155. Pendleton M, Lindsey RH Jr, Felix CA, Grimwade D, Osheroff N (2014) Topoisomerase II and leukemia. Ann N Y Acad Sci 1310:98–110. doi:10.1111/nyas.12358

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Cowell IG et al (2012) Model for MLL translocations in therapy-related leukemia involving topoisomerase IIbeta-mediated DNA strand breaks and gene proximity. Proc Natl Acad Sci U S A 109:8989–8994. doi:10.1073/pnas.1204406109

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Azarova AM et al (2007) Roles of DNA topoisomerase II isozymes in chemotherapy and secondary malignancies. Proc Natl Acad Sci U S A 104:11014–11019. doi:10.1073/pnas.0704002104

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Lovett BD et al (2001) Near-precise interchromosomal recombination and functional DNA topoisomerase II cleavage sites at MLL and AF-4 genomic breakpoints in treatment-related acute lymphoblastic leukemia with t(4;11) translocation. Proc Natl Acad Sci U S A 98:9802–9807. doi:10.1073/pnas.171309898

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Mahler M, Silverman ED, Schulte-Pelkum J, Fritzler MJ (2010) Anti-Scl-70 (topo-I) antibodies in SLE: myth or reality? Autoimmun Rev 9:756–760. doi:10.1016/j.autrev.2010.06.005

    Article  CAS  PubMed  Google Scholar 

  160. Haffner MC et al (2010) Androgen-induced TOP2B-mediated double-strand breaks and prostate cancer gene rearrangements. Nat Genet 42:668–675. doi:10.1038/ng.613

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Tomlins SA et al (2005) Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310:644–648. doi:10.1126/science.1117679

    Article  CAS  PubMed  Google Scholar 

  162. Helgeson BE et al (2008) Characterization of TMPRSS2:ETV5 and SLC45A3:ETV5 gene fusions in prostate cancer. Cancer Res 68:73–80. doi:10.1158/0008-5472.CAN-07-5352

    Article  CAS  PubMed  Google Scholar 

  163. Hermans KG et al (2008) Truncated ETV1, fused to novel tissue-specific genes, and full-length ETV1 in prostate cancer. Cancer Res 68:7541–7549. doi:10.1158/0008-5472.CAN-07-5930

    Article  CAS  PubMed  Google Scholar 

  164. Lin C et al (2009) Nuclear receptor-induced chromosomal proximity and DNA breaks underlie specific translocations in cancer. Cell 139:1069–1083. doi:10.1016/j.cell.2009.11.030

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Kumar-Sinha C, Tomlins SA, Chinnaiyan AM (2008) Recurrent gene fusions in prostate cancer. Nat Rev Cancer 8:497–511. doi:10.1038/nrc2402

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Bastus NC et al (2010) Androgen-induced TMPRSS2:ERG fusion in nonmalignant prostate epithelial cells. Cancer Res 70:9544–9548. doi:10.1158/0008-5472.CAN-10-1638

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  167. Bowen C, Gelmann E (2010) P. NKX3.1 activates cellular response to DNA damage. Cancer Res 70:3089–3097. doi:10.1158/0008-5472.CAN-09-3138

    Article  CAS  PubMed  Google Scholar 

  168. Bowen C, Zheng T, Gelmann EP (2015) NKX3.1 suppresses TMPRSS2-ERG gene rearrangement and mediates repair of androgen receptor-induced DNA damage. Cancer Res 75:2686–2698. doi:10.1158/0008-5472.CAN-14-3387

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Zhang H, Zheng T, Chua CW, Shen M, Gelmann EP (2016) Nkx3.1 controls the DNA repair response in the mouse prostate. Prostate 76:402–408. doi:10.1002/pros.23131

    Article  CAS  PubMed  Google Scholar 

  170. Mani RS et al (2016) Inflammation-induced oxidative stress mediates gene fusion formation in prostate cancer. Cell Rep 17:2620–2631. doi:10.1016/j.celrep.2016.11.019

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Laxman B et al (2006) Noninvasive detection of TMPRSS2:ERG fusion transcripts in the urine of men with prostate cancer. Neoplasia 8:885–888. doi:10.1593/neo.06625

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Merdan S et al (2015) Assessment of long-term outcomes associated with urinary prostate cancer antigen 3 and TMPRSS2:ERG gene fusion at repeat biopsy. Cancer 121:4071–4079. doi:10.1002/cncr.29611

    Article  CAS  PubMed  Google Scholar 

  173. Geybels MS et al (2015) Epigenomic profiling of prostate cancer identifies differentially methylated genes in TMPRSS2:ERG fusion-positive versus fusion-negative tumors. Clin Epigenetics 7:128. doi:10.1186/s13148-015-0161-6

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  174. Walker C, Herranz-Martin S, Karyka E, Liao C, Lewis K, Elsayed W, Lukashchuk V, Chiang SC, Ray S, Mulcahy PJ, Jurga M, Tsagakis I, Iannitti T, Chandran J, Coldicott I, De Vos KJ, Hassan MK, Higginbottom A, Shaw PJ, Hautbergue GM, Azzouz M, El-Khamisy SF (2017, July) C9orf72 expansion disrupts ATM-mediated chromosomal break repair. Nat Neurosci. doi:10.1038/nn.4604. [Epub ahead of print]

Download references

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Walaa R. Allam or Sherif El-Khamisy .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2017 American Association of Pharmaceutical Scientists

About this chapter

Cite this chapter

Allam, W.R., Ashour, M.E., Waly, A.A., El-Khamisy, S. (2017). Role of Protein Linked DNA Breaks in Cancer. In: El-Khamisy, S. (eds) Personalised Medicine. Advances in Experimental Medicine and Biology, vol 1007. Springer, Cham. https://doi.org/10.1007/978-3-319-60733-7_3

Download citation

Publish with us

Policies and ethics