DOI QR코드

DOI QR Code

Methylated-UHRF1 and PARP1 interaction is critical for homologous recombination

  • Hahm, Ja Young (Department of Life Science, College of Natural Sciences, Chung-Ang University) ;
  • Kang, Joo-Young (Department of Life Science, College of Natural Sciences, Chung-Ang University) ;
  • Park, Jin Woo (Department of Life Science, College of Natural Sciences, Chung-Ang University) ;
  • Jung, Hyeonsoo (Department of Life Science, College of Natural Sciences, Chung-Ang University) ;
  • Seo, Sang-Beom (Department of Life Science, College of Natural Sciences, Chung-Ang University)
  • Received : 2019.10.30
  • Accepted : 2019.12.15
  • Published : 2020.02.29

Abstract

A recent study suggested that methylation of ubiquitin-like with PHD and RING finger domain 1 (UHRF1) is regulated by SET7 and lysine-specific histone demethylase 1A (LSD1) and is essential for homologous recombination (HR). The study demonstrated that SET7-mediated methylation of UHRF1 promotes polyubiquitination of proliferating cell nuclear antigen (PCNA), inducing HR. However, studies on mediators that interact with and recruit UHRF1 to damaged lesions are needed to elucidate the mechanism of UHRF1 methylation-induced HR. Here, we identified that poly [ADP-ribose] polymerase 1 (PARP1) interacts with damage-induced methylated UHRF1 specifically and mediates UHRF1 to induce HR progression. Furthermore, cooperation of UHRF1-PARP1 is essential for cell viability, suggesting the importance of the interaction of UHRF1-PARP1 for damage tolerance in response to damage. Our data revealed that PARP1 mediates the HR mechanism, which is regulated by UHRF1 methylation. The data also indicated the significant role of PARP1 as a mediator of UHRF1 methylation-correlated HR pathway.

Keywords

References

  1. Bergink S and Jentsch S (2009) Principles of ubiquitin and SUMO modifications in DNA repair. Nature 458, 461-467 https://doi.org/10.1038/nature07963
  2. Zhang J, Lee YR, Dang F et al (2019) PTEN Methylation by NSD2 Controls Cellular Sensitivity to DNA Damage. Cancer Discov 9, 1306-1323 https://doi.org/10.1158/2159-8290.CD-18-0083
  3. Liu X, Gao Q, Li P et al (2013) UHRF1 targets DNMT1 for DNA methylation through cooperative binding of hemi-methylated DNA and methylated H3K9. Nat Commun 4, 1563 https://doi.org/10.1038/ncomms2562
  4. Chen H, Ma H, Inuzuka H et al (2013) DNA damage regulates UHRF1 stability via the SCF(beta-TrCP) E3 ligase. Mol Cell Biol 33, 1139-1148 https://doi.org/10.1128/MCB.01191-12
  5. Yang J, Liu K, Yang J et al (2017) PIM1 induces cellular senescence through phosphorylation of UHRF1 at Ser311. Oncogene 36, 4828-4842 https://doi.org/10.1038/onc.2017.96
  6. Hahm JY, Kim JY, Park JW et al (2019) Methylation of UHRF1 by SET7 is essential for DNA double-strand break repair. Nucleic Acids Res 47, 184-196 https://doi.org/10.1093/nar/gky975
  7. Ray Chaudhuri A and Nussenzweig A (2017) The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol 18, 610-621 https://doi.org/10.1038/nrm.2017.53
  8. Bunting SF, Callen E, Wong N et al (2010) 53BP1 inhibits homologous recombination in Brca1-deficient cells by blocking resection of DNA breaks. Cell 141, 243-254 https://doi.org/10.1016/j.cell.2010.03.012
  9. Li M and Yu X (2013) Function of BRCA1 in the DNA damage response is mediated by ADP-ribosylation. Cancer Cell 23, 693-704 https://doi.org/10.1016/j.ccr.2013.03.025
  10. De Vos M, El Ramy R, Quenet D et al (2014) Poly(ADP-ribose) polymerase 1 (PARP1) associates with E3 ubiquitin-protein ligase UHRF1 and modulates UHRF1 biological functions. J Biol Chem 289, 16223-16238 https://doi.org/10.1074/jbc.M113.527424
  11. Young LM, Marzio A, Perez-Duran P et al (2015) TIMELESS Forms a Complex with PARP1 Distinct from Its Complex with TIPIN and Plays a Role in the DNA Damage Response. Cell Rep 13, 451-459 https://doi.org/10.1016/j.celrep.2015.09.017
  12. Jelinic P and Levine DA (2014) New insights into PARP inhibitors' effect on cell cycle and homology-directed DNA damage repair. Mol Cancer Ther 13, 1645-1654 https://doi.org/10.1158/1535-7163.MCT-13-0906-T
  13. Clements KE, Thakar T, Nicolae CM, Liang X, Wang HG and Moldovan GL (2018) Loss of E2F7 confers resistance to poly-ADP-ribose polymerase (PARP) inhibitors in BRCA2-deficient cells. Nucleic Acids Res 46, 8898-8907 https://doi.org/10.1093/nar/gky657
  14. Rimar KJ, Tran PT, Matulewicz RS, Hussain M and Meeks JJ (2017) The emerging role of homologous recombination repair and PARP inhibitors in genitourinary malignancies. Cancer 123, 1912-1924 https://doi.org/10.1002/cncr.30631
  15. Thorn T, Gniadecki R, Petersen AB, Vicanova J and Wulf HC (2001) Differences in activation of G2/M checkpoint in keratinocytes after genotoxic stress induced by hydrogen peroxide and ultraviolet A radiation. Free Radic Res 35, 405-416 https://doi.org/10.1080/10715760100300921
  16. Santa-Gonzalez GA, Gomez-Molina A, Arcos-Burgos M, Meyer JN and Camargo M (2016) Distinctive adaptive response to repeated exposure to hydrogen peroxide associated with upregulation of DNA repair genes and cell cycle arrest. Redox Biol 9, 124-133 https://doi.org/10.1016/j.redox.2016.07.004
  17. Liu C, Vyas A, Kassab MA, Singh AK and Yu X (2017) The role of poly ADP-ribosylation in the first wave of DNA damage response. Nucleic Acids Res 45, 8129-8141 https://doi.org/10.1093/nar/gkx565
  18. Wang J, Hevi S, Kurash JK et al (2009) The lysine demethylase LSD1 (KDM1) is required for maintenance of global DNA methylation. Nat Genet 41, 125-129 https://doi.org/10.1038/ng.268
  19. Zhang H, Liu H, Chen Y et al (2016) A cell cycledependent BRCA1-UHRF1 cascade regulates DNA doublestrand break repair pathway choice. Nat Commun 7, 10201 https://doi.org/10.1038/ncomms10201
  20. Yang Q, Zhang R, Wang XW et al (2004) The mismatch DNA repair heterodimer, hMSH2/6, regulates BLM helicase. Oncogene 23, 3749-3756 https://doi.org/10.1038/sj.onc.1207462
  21. Chen G, Chen J, Qiao Y et al (2018) ZNF830 mediates cancer chemoresistance through promoting homologousrecombination repair. Nucleic Acids Res 46, 1266-1279 https://doi.org/10.1093/nar/gkx1258