2,062
Views
14
CrossRef citations to date
0
Altmetric
Research Paper

Destabilization of the MiniChromosome Maintenance (MCM) complex modulates the cellular response to DNA double strand breaks

, ORCID Icon, , , , ORCID Icon & ORCID Icon show all
Pages 2593-2609 | Received 20 Apr 2018, Accepted 24 Oct 2018, Published online: 10 Dec 2018

References

  • Blow JJ, Gillespie PJ. Replication licensing and cancer–a fatal entanglement? Nat Rev Cancer. 2008;8:799–806.
  • Blow JJ, Dutta A. Preventing re-replication of chromosomal DNA. Nat Rev Mol Cell Biol. 2005;6:476–486.
  • Bell SD, Botchan MR. The minichromosome maintenance replicative helicase. Cold Spring Harb Perspect Biol. 2013;5:a012807.
  • Sclafani RA, Fletcher RJ, Chen XS. Two heads are better than one: regulation of DNA replication by hexameric helicases. Genes Dev. 2004;18:2039–2045.
  • Bochman ML, Schwacha A. The Mcm complex: unwinding the mechanism of a replicative helicase. Microbiol Mol Biol Rev. 2009;73:652–683.
  • Arias EE, Walter JC. Strength in numbers: preventing rereplication via multiple mechanisms in eukaryotic cells. Genes Dev. 2007;21:497–518.
  • Allen C, Ashley AK, Hromas R, et al. More forks on the road to replication stress recovery. J Mol Cell Biol. 2011;3:4–12.
  • Nam EA, Cortez D. ATR signalling: more than meeting at the fork. Biochem J. 2011;436:527–536.
  • Bailis JM, Forsburg SL. MCM proteins: DNA damage, mutagenesis and repair. Curr Opin Genet Dev. 2004;14:17–21.
  • Forsburg SL. Eukaryotic MCM proteins: beyond replication initiation. Microbiol Mol Biol Rev. 2004;68:109–131.
  • Crevel I, Crevel G, Gostan T, et al. Decreased MCM2-6 in Drosophila S2 cells does not generate significant DNA damage or cause a marked increase in sensitivity to replication interference. PloS One. 2011;6:e27101.
  • Crevel G, Hashimoto R, Vass S, et al. Differential requirements for MCM proteins in DNA replication in Drosophila S2 cells. PloS One. 2007;2:e833.
  • Lei M, Kawasaki Y, Tye BK. Physical interactions among Mcm proteins and effects of Mcm dosage on DNA replication in Saccharomyces cerevisiae. Mol Cell Biol. 1996;16:5081–5090.
  • Oehlmann M, Score AJ, Blow JJ. The role of Cdc6 in ensuring complete genome licensing and S phase checkpoint activation. J Cell Biol. 2004;165:181–190.
  • Ibarra A, Schwob E, Mendez J. Excess MCM proteins protect human cells from replicative stress by licensing backup origins of replication. Pro Natl Acad Sci U S A. 2008;105:8956–8961.
  • Das M, Singh S, Pradhan S, et al. MCM paradox: abundance of eukaryotic replicative helicases and genomic integrity. Mol Biol Int. 2014;2014:1–11.
  • Painter RB, Young BR. Radiosensitivity in ataxia-telangiectasia: a new explanation. Proc Natl Acad Sci U S A. 1980;77:7315–7317.
  • Hartwell LH, Weinert TA. Checkpoints: controls that ensure the order of cell cycle events. Science. 1989;246:629–634.
  • Bartkova J, Horejsi Z, Koed K, et al. DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature. 2005;434:864–870.
  • Gorgoulis VG, Vassiliou LV, Karakaidos P, et al. Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature. 2005;434:907–913.
  • Smith J, Tho LM, Xu N, et al. The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Adv Cancer Res. 2010;108:73–112.
  • Ciccia A, Elledge SJ. The DNA damage response: making it safe to play with knives. Mol Cell. 2010;40:179–204.
  • Stracker TH, Usui T, Petrini JH. Taking the time to make important decisions: the checkpoint effector kinases Chk1 and Chk2 and the DNA damage response. DNA Repair (Amst). 2009;8:1047–1054.
  • Cortez D, Glick G, Elledge SJ. Minichromosome maintenance proteins are direct targets of the ATM and ATR checkpoint kinases. Proc Natl Acad Sci U S A. 2004;101:10078–10083.
  • Ishimi Y, Komamura-Kohno Y, Kwon HJ, et al. Identification of MCM4 as a target of the DNA replication block checkpoint system. J Biol Chem. 2003;278:24644–24650.
  • Sheu YJ, Stillman B. The Dbf4-Cdc7 kinase promotes S phase by alleviating an inhibitory activity in Mcm4. Nature. 2010;463:113–117.
  • Yoo HY, Shevchenko A, Shevchenko A, et al. Mcm2 is a direct substrate of ATM and ATR during DNA damage and DNA replication checkpoint responses. J Biol Chem. 2004;279:53353–53364.
  • Drissi R, Dubois ML, Douziech M, et al. Quantitative proteomics reveals dynamic interactions of the minichromosome maintenance complex (MCM) in the cellular response to etoposide induced DNA damage. Mol Cell Proteomics. 2015;14:2002–2013.
  • Ward IM, Wu X, Chen J. Threonine 68 of Chk2 is phosphorylated at sites of DNA strand breaks. J Biol Chem. 2001;276:47755–47758.
  • Liu Q, Guntuku S, Cui XS, et al. Chk1 is an essential kinase that is regulated by Atr and required for the G(2)/M DNA damage checkpoint. Genes Dev. 2000;14:1448–1459.
  • Boisvert FM, Ahmad Y, Gierlinski M, et al. A quantitative spatial proteomics analysis of proteome turnover in human cells. Mol Cell Proteomics. 2012;11:M111 011429.
  • Ge XQ, Jackson DA, Blow JJ. Dormant origins licensed by excess Mcm2-7 are required for human cells to survive replicative stress. Genes Dev. 2007;21:3331–3341.
  • Bochman ML, Bell SP, Schwacha A. Subunit organization of Mcm2-7 and the unequal role of active sites in ATP hydrolysis and viability. Mol Cell Biol. 2008;28:5865–5873.
  • Davey MJ, Indiani C, O’Donnell M. Reconstitution of the Mcm2-7p heterohexamer, subunit arrangement, and ATP site architecture. J Biol Chem. 2003;278:4491–4499.
  • Kimura H, Ohtomo T, Yamaguchi M, et al. Mouse MCM proteins: complex formation and transportation to the nucleus. Genes Cells. 1996;1:977–993.
  • Liku ME, Nguyen VQ, Rosales AW, et al. CDK phosphorylation of a novel NLS-NES module distributed between two subunits of the Mcm2-7 complex prevents chromosomal rereplication. Mol Biol Cell. 2005;16:5026–5039.
  • Han X, Aslanian A, Fu K, et al. The interaction between checkpoint kinase 1 (Chk1) and the minichromosome maintenance (MCM) complex is required for DNA damage-induced Chk1 phosphorylation. J Biol Chem. 2014;289:24716–24723.
  • Yan H, Tammaro M, Liao S. Collision of trapped topoisomerase 2 with transcription and replication: generation and repair of DNA double-strand breaks with 5ʹ adducts. Genes (Basel). 2016;7:32.
  • Shukla A, Navadgi VM, Mallikarjuna K, et al. Interaction of hRad51 and hRad52 with MCM complex: a cross-talk between recombination and replication proteins. Biochem Biophys Res Commun. 2005;329:1240–1245.
  • Bailis JM, Luche DD, Hunter T, et al. Minichromosome maintenance proteins interact with checkpoint and recombination proteins to promote s-phase genome stability. Mol Cell Biol. 2008;28:1724–1738.
  • Shechter D, Gautier J. MCM proteins and checkpoint kinases get together at the fork. Proc Natl Acad Sci U S A. 2004;101:10845–10846.
  • Vidi PA, Liu J, Salles D, et al. NuMA promotes homologous recombination repair by regulating the accumulation of the ISWI ATPase SNF2h at DNA breaks. Nucleic Acids Res. 2014;42:6365–6379.
  • Min S, Jo S, Lee HS, et al. ATM-dependent chromatin remodeler Rsf-1 facilitates DNA damage checkpoints and homologous recombination repair. Cell Cycle. 2014;13:666–677.
  • Luijsterburg MS, de Krijger I, Wiegant WW, et al. PARP1 links CHD2-mediated chromatin expansion and H3.3 deposition to DNA repair by non-homologous end-joining. Mol Cell. 2016;61:547–562.
  • Zhao Z, Oh S, Li D, et al. A dual role for UVRAG in maintaining chromosomal stability independent of autophagy. Dev Cell. 2012;22:1001–1016.
  • Robert F, Hardy S, Nagy Z, et al. The transcriptional histone acetyltransferase cofactor TRRAP associates with the MRN repair complex and plays a role in DNA double-strand break repair. Mol Cell Biol. 2006;26:402–412.
  • Lee YY, Yu YB, Gunawardena HP, et al. BCLAF1 is a radiation-induced H2AX-interacting partner involved in gammaH2AX-mediated regulation of apoptosis and DNA repair. Cell Death Dis. 2012;3:e359.
  • Kari V, Mansour WY, Raul SK, et al. Loss of CHD1 causes DNA repair defects and enhances prostate cancer therapeutic responsiveness. EMBO Rep. 2016;17:1609–1623.
  • Shenoy TR, Boysen G, Wang MY, et al. CHD1 loss sensitizes prostate cancer to DNA damaging therapy by promoting error-prone double-strand break repair. Ann Oncol. 2017;28:1495–1507.
  • Boddy MN, Gaillard PH, McDonald WH, et al. Mus81-Eme1 are essential components of a holliday junction resolvase. Cell. 2001;107:537–548.
  • Ray A, Milum K, Battu A, et al. NER initiation factors, DDB2 and XPC, regulate UV radiation response by recruiting ATR and ATM kinases to DNA damage sites. DNA Repair (Amst). 2013;12:273–283.
  • Hentges P, Waller H, Reis CC, et al. Cdk1 restrains NHEJ through phosphorylation of XRCC4-like factor Xlf1. Cell Rep. 2014;9:2011–2017.
  • Lee KY, Im JS, Shibata E, et al. MCM8-9 complex promotes resection of double-strand break ends by MRE11-RAD50-NBS1 complex. Nat Commun. 2015;6:7744.
  • Lutzmann M, Grey C, Traver S, et al. MCM8- and MCM9-deficient mice reveal gametogenesis defects and genome instability due to impaired homologous recombination. Mol Cell. 2012;47:523–534.
  • Nishimura K, Ishiai M, Horikawa K, et al. Mcm8 and Mcm9 form a complex that functions in homologous recombination repair induced by DNA interstrand crosslinks. Mol Cell. 2012;47:511–522.
  • Park J, Long DT, Lee KY, et al. The MCM8-MCM9 complex promotes RAD51 recruitment at DNA damage sites to facilitate homologous recombination. Mol Cell Biol. 2013;33:1632–1644.
  • Lutzmann M, Mechali M. MCM9 binds Cdt1 and is required for the assembly of prereplication complexes. Mol Cell. 2008;31:190–200.
  • Maiorano D, Cuvier O, Danis E, et al. MCM8 is an MCM2-7-related protein that functions as a DNA helicase during replication elongation and not initiation. Cell. 2005;120:315–328.
  • Natsume T, Nishimura K, Minocherhomji S, et al. Acute inactivation of the replicative helicase in human cells triggers MCM8-9-dependent DNA synthesis. Genes Dev. 2017;31:816–829.
  • Pierce AJ, Johnson RD, Thompson LH, et al. XRCC3 promotes homology-directed repair of DNA damage in mammalian cells. Genes Dev. 1999;13:2633–2638.
  • Bennardo N, Cheng A, Huang N, et al. Alternative-NHEJ is a mechanistically distinct pathway of mammalian chromosome break repair. PLoS Genet. 2008;4:e1000110.
  • Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol. 2008;26:1367–1372.
  • Cox J, Matic I, Hilger M, et al. A practical guide to the MaxQuant computational platform for SILAC-based quantitative proteomics. Nat Protoc. 2009;4:698–705.