1,574
Views
10
CrossRef citations to date
0
Altmetric
Extra View

Interrogating cell division errors using random and chromosome-specific missegregation approaches

ORCID Icon &
Pages 1252-1258 | Received 21 Apr 2017, Accepted 24 Apr 2017, Published online: 26 Jun 2017

References

  • Weaver BA, Cleveland DW. The aneuploidy paradox in cell growth and tumorigenesis. Cancer Cell 2008; 14:431-3; PMID:19061834; https://doi.org/10.1016/j.ccr.2008.11.011
  • Janssen A, van der Burg M, Szuhai K, Kops GJ, Medema RH. Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science 2011; 333:1895-8; PMID:21960636; https://doi.org/10.1126/science.1210214
  • Musacchio A, Desai A. A molecular view of kinetochore assembly and function. Biology (Basel) 2017; 6:pii: E5; PMID:28125021
  • Walczak CE, Cai S, Khodjakov A. Mechanisms of chromosome behaviour during mitosis. Nat Rev Mol Cell Biol 2010; 11:91-102; PMID:20068571
  • Thompson SL, Compton DA. Examining the link between chromosomal instability and aneuploidy in human cells. J Cell Biol 2008; 180:665-72; PMID:18283116; https://doi.org/10.1083/jcb.200712029
  • Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR, Mudie LJ, Pleasance ED, Lau KW, Beare D, Stebbings LA, et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 2011; 144:27-40; PMID:21215367; https://doi.org/10.1016/j.cell.2010.11.055
  • Zhang CZ, Spektor A, Cornils H, Francis JM, Jackson EK, Liu S, Meyerson M, Pellman D. Chromothripsis from DNA damage in micronuclei. Nature 2015; 522:179-84; PMID:26017310; https://doi.org/10.1038/nature14493
  • Tan EH, Henry IM, Ravi M, Bradnam KR, Mandakova T, Marimuthu MP, Korf I, Lysak MA, Comai L, Chan SW. Catastrophic chromosomal restructuring during genome elimination in plants. Elife 2015; 4:pii: e06516
  • Santaguida S, Amon A. Short- and long-term effects of chromosome mis-segregation and aneuploidy. Nat Rev Mol Cell Biol 2015; 16:473-85; PMID:26204159; https://doi.org/10.1038/nrm4025
  • Ly P, Teitz LS, Kim DH, Shoshani O, Skaletsky H, Fachinetti D, Page DC, Cleveland DW. Selective Y centromere inactivation triggers chromosome shattering in micronuclei and repair by non-homologous end joining. Nat Cell Biol 2017; 19:68-75; PMID:27918550; https://doi.org/10.1038/ncb3450
  • Khodjakov A, Cole RW, Oakley BR, Rieder CL. Centrosome-independent mitotic spindle formation in vertebrates. Curr Biol 2000; 10:59-67; PMID:10662665; https://doi.org/10.1016/S0960-9822(99)00276-6
  • Schiff PB, Horwitz SB. Taxol stabilizes microtubules in mouse fibroblast cells. Proc Natl Acad Sci U S A 1980; 77:1561-5; PMID:6103535; https://doi.org/10.1073/pnas.77.3.1561
  • Kirschner M, Mitchison T. Beyond self-assembly: from microtubules to morphogenesis. Cell 1986; 45:329-42; PMID:3516413; https://doi.org/10.1016/0092-8674(86)90318-1
  • Mayer TU, Kapoor TM, Haggarty SJ, King RW, Schreiber SL, Mitchison TJ. Small molecule inhibitor of mitotic spindle bipolarity identified in a phenotype-based screen. Science 1999; 286:971-4; PMID:10542155; https://doi.org/10.1126/science.286.5441.971
  • DeBonis S, Skoufias DA, Lebeau L, Lopez R, Robin G, Margolis RL, Wade RH, Kozielski F. In vitro screening for inhibitors of the human mitotic kinesin Eg5 with antimitotic and antitumor activities. Mol Cancer Ther 2004; 3:1079-90; PMID:15367702
  • Sawin KE, LeGuellec K, Philippe M, Mitchison TJ. Mitotic spindle organization by a plus-end-directed microtubule motor. Nature 1992; 359:540-3; PMID:1406972; https://doi.org/10.1038/359540a0
  • Qian X, McDonald A, Zhou HJ, Adams ND, Parrish CA, Duffy KJ, Fitch DM, Tedesco R, Ashcraft LW, Yao B, et al. Discovery of the First Potent and Selective Inhibitor of Centromere-Associated Protein E: GSK923295. ACS Med Chem Lett 2010; 1:30-4; PMID:24900171; https://doi.org/10.1021/ml900018m
  • Yen TJ, Li G, Schaar BT, Szilak I, Cleveland DW. CENP-E is a putative kinetochore motor that accumulates just before mitosis. Nature 1992; 359:536-9; PMID:1406971; https://doi.org/10.1038/359536a0
  • Orth JD, Loewer A, Lahav G, Mitchison TJ. Prolonged mitotic arrest triggers partial activation of apoptosis, resulting in DNA damage and p53 induction. Mol Biol Cell 2012; 23:567-76; PMID:22171325; https://doi.org/10.1091/mbc.E11-09-0781
  • Uetake Y, Sluder G. Prolonged prometaphase blocks daughter cell proliferation despite normal completion of mitosis. Curr Biol 2010; 20:1666-71; PMID:20832310; https://doi.org/10.1016/j.cub.2010.08.018
  • Santaguida S, Tighe A, D'Alise AM, Taylor SS, Musacchio A. Dissecting the role of MPS1 in chromosome biorientation and the spindle checkpoint through the small molecule inhibitor reversine. J Cell Biol 2010; 190:73-87; PMID:20624901; https://doi.org/10.1083/jcb.201001036
  • Hewitt L, Tighe A, Santaguida S, White AM, Jones CD, Musacchio A, Green S, Taylor SS. Sustained Mps1 activity is required in mitosis to recruit O-Mad2 to the Mad1-C-Mad2 core complex. J Cell Biol 2010; 190:25-34; PMID:20624899; https://doi.org/10.1083/jcb.201002133
  • Sansregret L, Patterson JO, Dewhurst S, Lopez-Garcia C, Koch A, McGranahan N, Chao WC, Barry DJ, Rowan A, Instrell R, et al. APC/C dysfunction limits excessive cancer chromosomal instability. Cancer Discov 2017; 7(2):218-33; PMID:28069571
  • Robinett CC, Straight A, Li G, Willhelm C, Sudlow G, Murray A, Belmont AS. In vivo localization of DNA sequences and visualization of large-scale chromatin organization using lac operator/repressor recognition. J Cell Biol 1996; 135:1685-700; PMID:8991083; https://doi.org/10.1083/jcb.135.6.1685
  • Ma H, Naseri A, Reyes-Gutierrez P, Wolfe SA, Zhang S, Pederson T. Multicolor CRISPR labeling of chromosomal loci in human cells. Proc Natl Acad Sci U S A 2015; 112:3002-7; PMID:25713381; https://doi.org/10.1073/pnas.1420024112
  • Ma H, Tu LC, Naseri A, Huisman M, Zhang S, Grunwald D, Pederson T. Multiplexed labeling of genomic loci with dCas9 and engineered sgRNAs using CRISPRainbow. Nat Biotechnol 2016; 34:528-30; PMID:27088723; https://doi.org/10.1038/nbt.3526
  • Anton T, Leonhardt H, Markaki Y. Visualization of genomic loci in living cells with a fluorescent CRISPR/Cas9 system. Methods Mol Biol 2016; 1411:407-17; PMID:27147056
  • Chen B, Gilbert LA, Cimini BA, Schnitzbauer J, Zhang W, Li GW, Park J, Blackburn EH, Weissman JS, Qi LS, et al. Dynamic imaging of genomic loci in living human cells by an optimized CRISPR/Cas system. Cell 2013; 155:1479-91; PMID:24360272; https://doi.org/10.1016/j.cell.2013.12.001
  • Nakano M, Cardinale S, Noskov VN, Gassmann R, Vagnarelli P, Kandels-Lewis S, Larionov V, Earnshaw WC, Masumoto H. Inactivation of a human kinetochore by specific targeting of chromatin modifiers. Dev Cell 2008; 14:507-22; PMID:18410728; https://doi.org/10.1016/j.devcel.2008.02.001
  • Kouprina N, Samoshkin A, Erliandri I, Nakano M, Lee HS, Fu H, Iida Y, Aladjem M, Oshimura M, Masumoto H, et al. Organization of synthetic alphoid DNA array in human artificial chromosome (HAC) with a conditional centromere. ACS Synth Biol 2012; 1:590-601; PMID:23411994; https://doi.org/10.1021/sb3000436
  • Crasta K, Ganem NJ, Dagher R, Lantermann AB, Ivanova EV, Pan Y, Nezi L, Protopopov A, Chowdhury D, Pellman D. DNA breaks and chromosome pulverization from errors in mitosis. Nature 2012; 482:53-8; PMID:22258507; https://doi.org/10.1038/nature10802
  • Lee HS, Lee NC, Grimes BR, Samoshkin A, Kononenko AV, Bansal R, Masumoto H, Earnshaw WC, Kouprina N, Larionov V. A new assay for measuring chromosome instability (CIN) and identification of drugs that elevate CIN in cancer cells. BMC Cancer 2013; 13:252; PMID:23694679; https://doi.org/10.1186/1471-2407-13-252
  • Markossian S, Arnaoutov A, Saba NS, Larionov V, Dasso M. Quantitative assessment of chromosome instability induced through chemical disruption of mitotic progression. Cell Cycle 2016; 15:1706-14; PMID:27104376; https://doi.org/10.1080/15384101.2016.1175796
  • Macnab S, Whitehouse A. Progress and prospects: human artificial chromosomes. Gene Ther 2009; 16:1180-8; PMID:19710706; https://doi.org/10.1038/gt.2009.102
  • Zahn LM, Riddihough G. Building on nature's design. Science 2017; 355:1038-9; PMID:28280198
  • Fournier RE, Ruddle FH. Microcell-mediated transfer of murine chromosomes into mouse, Chinese hamster, and human somatic cells. Proc Natl Acad Sci U S A 1977; 74:319-23; PMID:264685; https://doi.org/10.1073/pnas.74.1.319
  • Upender MB, Habermann JK, McShane LM, Korn EL, Barrett JC, Difilippantonio MJ, Ried T. Chromosome transfer induced aneuploidy results in complex dysregulation of the cellular transcriptome in immortalized and cancer cells. Cancer Res 2004; 64:6941-9; PMID:15466185; https://doi.org/10.1158/0008-5472.CAN-04-0474
  • Passerini V, Ozeri-Galai E, de Pagter MS, Donnelly N, Schmalbrock S, Kloosterman WP, Kerem B, Storchova Z. The presence of extra chromosomes leads to genomic instability. Nat Commun 2016; 7:10754; PMID:26876972; https://doi.org/10.1038/ncomms10754
  • Rutledge SD, Douglas TA, Nicholson JM, Vila-Casadesus M, Kantzler CL, Wangsa D, Barroso-Vilares M, Kale SD, Logarinho E, Cimini D. Selective advantage of trisomic human cells cultured in non-standard conditions. Sci Rep 2016; 6:22828; PMID:26956415; https://doi.org/10.1038/srep22828
  • Sheltzer JM, Ko JH, Replogle JM, Habibe Burgos NC, Chung ES, Meehl CM, Sayles NM, Passerini V, Storchova Z, Amon A. Single-chromosome gains commonly function as tumor suppressors. Cancer Cell 2016; 31(2):240-55
  • Williams BR, Prabhu VR, Hunter KE, Glazier CM, Whittaker CA, Housman DE, Amon A. Aneuploidy affects proliferation and spontaneous immortalization in mammalian cells. Science 2008; 322:703-9; PMID:18974345; https://doi.org/10.1126/science.1160058
  • Chlebowicz-Sledziewska E, Sledziewski AZ. Construction of multicopy yeast plasmids with regulated centromere function. Gene 1985; 39:25-31; PMID:2934294; https://doi.org/10.1016/0378-1119(85)90103-9
  • Hill A, Bloom K. Genetic manipulation of centromere function. Mol Cell Biol 1987; 7:2397-405; PMID:3302676; https://doi.org/10.1128/MCB.7.7.2397
  • Fachinetti D, Folco HD, Nechemia-Arbely Y, Valente LP, Nguyen K, Wong AJ, Zhu Q, Holland AJ, Desai A, Jansen LE, et al. A two-step mechanism for epigenetic specification of centromere identity and function. Nat Cell Biol 2013; 15:1056-66; PMID:23873148; https://doi.org/10.1038/ncb2805
  • Jansen LE, Black BE, Foltz DR, Cleveland DW. Propagation of centromeric chromatin requires exit from mitosis. J Cell Biol 2007; 176:795-805; PMID:17339380; https://doi.org/10.1083/jcb.200701066
  • Foltz DR, Jansen LE, Bailey AO, Yates JR 3rd, Bassett EA, Wood S, Black BE, Cleveland DW. Centromere-specific assembly of CENP-a nucleosomes is mediated by HJURP. Cell 2009; 137:472-84; PMID:19410544; https://doi.org/10.1016/j.cell.2009.02.039
  • Dunleavy EM, Roche D, Tagami H, Lacoste N, Ray-Gallet D, Nakamura Y, Daigo Y, Nakatani Y, Almouzni-Pettinotti G. HJURP is a cell-cycle-dependent maintenance and deposition factor of CENP-A at centromeres. Cell 2009; 137:485-97; PMID:19410545; https://doi.org/10.1016/j.cell.2009.02.040
  • Hoffmann S, Dumont M, Barra V, Ly P, Nechemia-Arbely Y, McMahon MA, Herve S, Cleveland DW, Fachinetti D. CENP-A is dispensable for mitotic centromere function after initial centromere/kinetochore assembly. Cell Rep 2016; 17:2394-404; PMID:27880912; https://doi.org/10.1016/j.celrep.2016.10.084
  • Skaletsky H, Kuroda-Kawaguchi T, Minx PJ, Cordum HS, Hillier L, Brown LG, Repping S, Pyntikova T, Ali J, Bieri T, et al. The male-specific region of the human Y chromosome is a mosaic of discrete sequence classes. Nature 2003; 423:825-37; PMID:12815422; https://doi.org/10.1038/nature01722
  • Torres EM, Sokolsky T, Tucker CM, Chan LY, Boselli M, Dunham MJ, Amon A. Effects of aneuploidy on cellular physiology and cell division in haploid yeast. Science 2007; 317:916-24; PMID:17702937; https://doi.org/10.1126/science.1142210
  • Hatch EM, Fischer AH, Deerinck TJ, Hetzer MW. Catastrophic nuclear envelope collapse in cancer cell micronuclei. Cell 2013; 154:47-60; PMID:23827674; https://doi.org/10.1016/j.cell.2013.06.007
  • Kato H, Sandberg AA. Chromosome pulverization in human cells with micronuclei. J Natl Cancer Inst 1968; 40:165-79; PMID:5635016
  • Scott KC, Sullivan BA. Neocentromeres: a place for everything and everything in its place. Trends Genet 2014; 30:66-74; PMID:24342629; https://doi.org/10.1016/j.tig.2013.11.003
  • Amor DJ, Bentley K, Ryan J, Perry J, Wong L, Slater H, Choo KH. Human centromere repositioning "in progress." Proc Natl Acad Sci U S A 2004; 101:6542-7; PMID:15084747; https://doi.org/10.1073/pnas.0308637101
  • Burrack LS, Berman J. Neocentromeres and epigenetically inherited features of centromeres. Chromosome Res 2012; 20:607-19; PMID:22723125; https://doi.org/10.1007/s10577-012-9296-x
  • Amor DJ, Choo KH. Neocentromeres: role in human disease, evolution, and centromere study. Am J Hum Genet 2002; 71:695-714; PMID:12196915; https://doi.org/10.1086/342730

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.