2
Views
2
CrossRef citations to date
0
Altmetric
Review

Advances in understanding the genetic causes and mechanisms of female germ cell aneuploidy

&
Pages 687-706 | Published online: 10 Jan 2014

References

  • Hassold T, Hunt P. To err (meiotically) is human: the genesis of human aneuploidy. Nat. Rev. Genet.2, 280–291 (2001).
  • Hassold T, Hall H, Hunt P. The origin of human aneuploidy: where we have been, where we are going. Hum. Mol. Genet.16(Spec. No. 2), R203–R208 (2007).
  • Magli MC, Gianaroli L, Ferraretti AP. Chromosomal abnormalities in embryos. Mol. Cell. Endocrinol.183(Suppl. 1), S29–S34 (2001).
  • Munne S. Preimplantation genetic diagnosis of numerical and structural chromosome abnormalities. Reprod. Biomed. Online4, 183–196 (2002).
  • Kuliev A, Cieslak J, Ilkevitch Y, Verlinsky Y. Chromosomal abnormalities in a series of 6,733 human oocytes in preimplantation diagnosis for age-related aneuploidies. Reprod. Biomed. Online6, 54–59 (2003).
  • Yanagida M. Basic mechanism of eukaryotic chromosome segregation. Philos. Trans. R. Soc. Lond. B. Biol. Sci.360, 609–621 (2005).
  • Petronczki M, Siomos MF, Nasmyth K. Un menage a quatre: the molecular biology of chromosome segregation in meiosis. Cell112, 423–440 (2003).
  • Anderiesz C, Ferraretti A, Magli C et al. Effect of recombinant human gonadotrophins on human, bovine and murine oocyte meiosis, fertilization and embryonic development in vitro. Hum. Reprod.15, 1140–1148 (2000).
  • Hillier SG. Gonadotropic control of ovarian follicular growth and development. Mol. Cell. Endocrinol.179, 39–46 (2001).
  • Hecht NB. Molecular mechanisms of male germ cell differentiation. Bioessays20, 555–561 (1998).
  • Hunt PA, Hassold TJ. Sex matters in meiosis. Science296, 2181–2183 (2002).
  • Pacchierotti F, Adler ID, Eichenlaub-Ritter U, Mailhes JB. Gender effects on the incidence of aneuploidy in mammalian germ cells. Environ. Res.104, 46–69 (2007).
  • Warburton D. Biological aging and the etiology of aneuploidy. Cytogenet. Genome Res.111, 266–272 (2005).
  • Wolstenholme J, Angell RR. Maternal age and trisomy – a unifying mechanism of formation. Chromosoma109, 435–438 (2000).
  • Rosenbusch B. The incidence of aneuploidy in human oocytes assessed by conventional cytogenetic analysis. Hereditas141, 97–105 (2004).
  • Nicolaidis P, Petersen MB. Origin and mechanisms of non-disjunction in human autosomal trisomies. Hum. Reprod.13, 313–319 (1998).
  • Gianaroli L, Magli MC, Cavallini G et al. Predicting aneuploidy in human oocytes: key factors which affect the meiotic process. Hum. Reprod.25, 2374–2386 (2010).
  • Mailhes JB, Marchetti F. Mechanisms and chemical induction of aneuploidy in rodent germ cells. Cytogenet. Genome Res.111, 384–391 (2005).
  • Handel MA, Schimenti JC. Genetics of mammalian meiosis: regulation, dynamics and impact on fertility. Nat. Rev. Genet.11, 124–136 (2010).
  • Garcia-Cruz R, Roig I, Caldes MG. Maternal origin of the human aneuploidies. Are homolog synapsis and recombination to blame? Notes (learned) from the underbelly. Genome Dyn.5, 128–136 (2009).
  • Martin RH. Meiotic errors in human oogenesis and spermatogenesis. Reprod. Biomed. Online16, 523–531 (2008).
  • Angell RR. Predivision in human oocytes at meiosis I: a mechanism for trisomy formation in man. Hum. Genet.86, 383–387 (1991).
  • Angell RR. Aneuploidy in older women. Higher rates of aneuploidy in oocytes from older women. Hum. Reprod.9, 1199–1200 (1994).
  • Anahory T, Andreo B, Regnier-Vigouroux G et al. Sequential multiple probe fluorescence in-situ hybridization analysis of human oocytes and polar bodies by combining centromeric labelling and whole chromosome painting. Mol. Hum. Reprod.9, 577–585 (2003).
  • Pellestor F, Andreo B, Arnal F, Humeau C, Demaille J. Maternal aging and chromosomal abnormalities: new data drawn from in vitro unfertilized human oocytes. Hum. Genet.112, 195–203 (2003).
  • Plachot M. Genetic analysis of the oocyte – a review. Placenta24(Suppl. B), S66–S69 (2003).
  • Rodman TC. Chromatid disjunction in unfertilized ageing oocytes. Nature233, 191–193 (1971).
  • Cupisti S, Conn CM, Fragouli E et al. Sequential FISH analysis of oocytes and polar bodies reveals aneuploidy mechanisms. Prenat. Diagn.23, 663–668 (2003).
  • Pellestor F, Andreo B, Arnal F, Humeau C, Demaille J. Mechanisms of non-disjunction in human female meiosis: the co-existence of two modes of malsegregation evidenced by the karyotyping of 1397 in-vitro unfertilized oocytes. Hum. Reprod.17, 2134–2145 (2002).
  • Mailhes JB, Young D, London SN. 1,2-propanediol-induced premature centromere separation in mouse oocytes and aneuploidy in one-cell zygotes. Biol. Reprod.57, 92–98 (1997).
  • Mailhes JB, Young D, London SN. Postovulatory ageing of mouse oocytes in vivo and premature centromere separation and aneuploidy. Biol. Reprod.58, 1206–1210 (1998).
  • Yin H, Baart E, Betzendahl I, Eichenlaub-Ritter U. Diazepam induces meiotic delay, aneuploidy and predivision of homologues and chromatids in mammalian oocytes. Mutagenesis13, 567–580 (1998).
  • Jeffreys CA, Burrage PS, Bickel SE. A model system for increased meiotic nondisjunction in older oocytes. Curr. Biol.13, 498–503 (2003).
  • Dailey T, Dale B, Cohen J, Munne S. Association between nondisjunction and maternal age in meiosis-II human oocytes. Am. J. Hum. Genet.59, 176–184 (1996).
  • Pellestor F, Anahory T, Hamamah S. The chromosomal analysis of human oocytes. An overview of established procedures. Hum. Reprod. Update11, 15–32 (2005).
  • Vialard F, Petit C, Bergere M et al. Evidence of a high proportion of premature unbalanced separation of sister chromatids in the first polar bodies of women of advanced age. Hum. Reprod.21, 1172–1178 (2006).
  • Sonoda E, Matsusaka T, Morrison C et al. Scc1/Rad21/Mcd1 is required for sister chromatid cohesion and kinetochore function in vertebrate cells. Dev. Cell1, 759–770 (2001).
  • Hoque MT, Ishikawa F. Cohesin defects lead to premature sister chromatid separation, kinetochore dysfunction, and spindle-assembly checkpoint activation. J. Biol. Chem.277, 42306–42314 (2002).
  • Uhlmann F. Chromosome cohesion and separation: from men and molecules. Curr. Biol.13, R104–R114 (2003).
  • Kerrebrock AW, Miyazaki WY, Birnby D, Orr-Weaver TL. The Drosophilamei-S332 gene promotes sister-chromatid cohesion in meiosis following kinetochore differentiation. Genetics130, 827–841 (1992).
  • Miyazaki WY, Orr-Weaver TL. Sister-chromatid misbehavior in Drosophila ord mutants. Genetics132, 1047–1061 (1992).
  • Hodges CA, Revenkova E, Jessberger R, Hassold TJ, Hunt PA. SMC1β-deficient female mice provide evidence that cohesins are a missing link in age-related nondisjunction. Nat. Genet.37, 1351–1355 (2005).
  • Hartman T, Stead K, Koshland D, Guacci V. Pds5p is an essential chromosomal protein required for both sister chromatid cohesion and condensation in Saccharomyces cerevisiae. J. Cell. Biol.151, 613–626 (2000).
  • Panizza S, Tanaka T, Hochwagen A, Eisenhaber F, Nasmyth K. Pds5 cooperates with cohesin in maintaining sister chromatid cohesion. Curr. Biol.10, 1557–1564 (2000).
  • Thein KH, Kleylein-Sohn J, Nigg EA, Gruneberg U. Astrin is required for the maintenance of sister chromatid cohesion and centrosome integrity. J. Cell. Biol.178, 345–354 (2007).
  • Cukurcam S, Hegele-Hartung C, Eichenlaub-Ritter U. Meiosis-activating sterol protects oocytes from precocious chromosome segregation. Hum. Reprod.18, 1908–1917 (2003).
  • Losada A, Yokochi T, Kobayashi R, Hirano T. Identification and characterization of SA/Scc3p subunits in the Xenopus and human cohesin complexes. J. Cell. Biol.150, 405–416 (2000).
  • Tomonaga T, Nagao K, Kawasaki Y et al. Characterization of fission yeast cohesin: essential anaphase proteolysis of Rad21 phosphorylated in the S phase. Genes Dev.14, 2757–2770 (2000).
  • Lee BH, Amon A. Role of Polo-like kinase CDC5 in programming meiosis I chromosome segregation. Science300, 482–486 (2003).
  • Yu HG, Koshland D. Chromosome morphogenesis: condensin-dependent cohesin removal during meiosis. Cell123, 397–407 (2005).
  • Peter M, Castro A, Lorca T et al. The APC is dispensable for first meiotic anaphase in Xenopus oocytes. Nat. Cell. Biol.3, 83–87 (2001).
  • O’Neill GT, Kaufman MH. Influence of postovulatory aging on chromosome segregation during the second meiotic division in mouse oocytes: a parthenogenetic analysis. J. Exp. Zool.248, 125–131 (1988).
  • Steuerwald NM, Steuerwald MD, Mailhes JB. Post-ovulatory aging of mouse oocytes leads to decreased Mad2 transcripts and increased frequencies of premature centromere separation and anaphase. Mol. Hum. Reprod.11, 623–630 (2005).
  • Kitajima TS, Sakuno T, Ishiguro K et al. Shugoshin collaborates with protein phosphatase 2A to protect cohesin. Nature441, 46–52 (2006).
  • Riedel CG, Katis VL, Katou Y et al. Protein phosphatase 2A protects centromeric sister chromatid cohesion during meiosis I. Nature441, 53–61 (2006).
  • Tang Z, Shu H, Qi W, Mahmood NA, Mumby MC, Yu H. PP2A is required for centromeric localization of Sgo1 and proper chromosome segregation. Dev. Cell10, 575–585 (2006).
  • Sluder G, McCollum D. Molecular biology. The mad ways of meiosis. Science289, 254–255 (2000).
  • Schultz RM, Montgomery RR, Belanoff JR. Regulation of mouse oocyte meiotic maturation: implication of a decrease in oocyte cAMP and protein dephosphorylation in commitment to resume meiosis. Dev. Biol.97, 264–273 (1983).
  • Dekel N. Regulation of oocyte maturation. The role of cAMP. Ann. N. Y. Acad. Sci.541, 211–216 (1988).
  • Yamashita M, Mita K, Yoshida N, Kondo T. Molecular mechanisms of the initiation of oocyte maturation: general and species-specific aspects. Prog. Cell. Cycle Res.4, 115–129 (2000).
  • Dekel N. Cellular, biochemical and molecular mechanisms regulating oocyte maturation. Mol. Cell. Endocrinol.234, 19–25 (2005).
  • Dunphy WG, Kumagai A. The Cdc25 protein contains an intrinsic phosphatase activity. Cell67, 189–196 (1991).
  • Gautier J, Solomon MJ, Booher RN, Bazan JF, Kirschner MW. Cdc25 is a specific tyrosine phosphatase that directly activates p34Cdc2. Cell67, 197–211 (1991).
  • Strausfeld U, Labbe JC, Fesquet D et al. Dephosphorylation and activation of a p34Cdc2/cyclin B complex in vitro by human CDC25 protein. Nature351, 242–245 (1991).
  • Arion D, Meijer L, Brizuela L, Beach D. Cdc2 is a component of the M phase-specific histone H1 kinase: evidence for identity with MPF. Cell55, 371–378 (1988).
  • Draetta G, Beach D. Activation of Cdc2 protein kinase during mitosis in human cells: cell cycle-dependent phosphorylation and subunit rearrangement. Cell54, 17–26 (1988).
  • Choi T, Aoki F, Mori M, Yamashita M, Nagahama Y, Kohmoto K. Activation of p34Cdc2 protein kinase activity in meiotic and mitotic cell cycles in mouse oocytes and embryos. Development113, 789–795 (1991).
  • Fulka J Jr, Jung T, Moor RM. The fall of biological maturation promoting factor (MPF) and histone H1 kinase activity during anaphase and telophase in mouse oocytes. Mol. Reprod. Dev.32, 378–382 (1992).
  • Collas P, Sullivan EJ, Barnes FL. Histone H1 kinase activity in bovine oocytes following calcium stimulation. Mol. Reprod. Dev.34, 224–231 (1993).
  • Kikuchi K, Izaike Y, Noguchi J et al. Decrease of histone H1 kinase activity in relation to parthenogenetic activation of pig follicular oocytes matured and aged in vitro. J. Reprod. Fertil.105, 325–330 (1995).
  • Swain JE, Smith GD. Reversible phosphorylation and regulation of mammalian oocyte meiotic chromatin remodeling and segregation. Soc. Reprod. Fertil. Suppl.63, 343–358 (2007).
  • Murray AW. MAP kinases in meiosis. Cell92, 157–159 (1998).
  • Takenaka K, Moriguchi T, Nishida E. Activation of the protein kinase p38 in the spindle assembly checkpoint and mitotic arrest. Science280, 599–602 (1998).
  • Lee J, Miyano T, Moor RM. Localisation of phosphorylated MAP kinase during the transition from meiosis I to meiosis II in pig oocytes. Zygote8, 119–125 (2000).
  • Fan HY, Sun QY. Involvement of mitogen-activated protein kinase cascade during oocyte maturation and fertilization in mammals. Biol. Reprod.70, 535–547 (2004).
  • Sobajima T, Aoki F, Kohmoto K. Activation of mitogen-activated protein kinase during meiotic maturation in mouse oocytes. J. Reprod. Fertil.97, 389–394 (1993).
  • Verlhac MH, Kubiak JZ, Clarke HJ, Maro B. Microtubule and chromatin behavior follow MAP kinase activity but not MPF activity during meiosis in mouse oocytes. Development120, 1017–1025 (1994).
  • Gordo AC, He CL, Smith S, Fissore RA. Mitogen activated protein kinase plays a significant role in metaphase II arrest, spindle morphology, and maintenance of maturation promoting factor activity in bovine oocytes. Mol. Reprod. Dev.59, 106–114 (2001).
  • Solc P, Schultz RM, Motlik J. Prophase I arrest and progression to metaphase I in mouse oocytes: comparison of resumption of meiosis and recovery from G2-arrest in somatic cells. Mol. Hum. Reprod.16, 654–664 (2010).
  • Paules RS, Buccione R, Moschel RC, Vande Woude GF, Eppig JJ. Mouse Mos protooncogene product is present and functions during oogenesis. Proc. Natl Acad. Sci. USA86, 5395–5399 (1989).
  • Sagata N. What does Mos do in oocytes and somatic cells? Bioessays19, 13–21 (1997).
  • Dekel N. Protein phosphorylation/dephosphorylation in the meiotic cell cycle of mammalian oocytes. Rev. Reprod.1, 82–88 (1996).
  • Hashimoto N. Role of c-mos proto-oncogene product in the regulation of mouse oocyte maturation. Horm. Res.46(Suppl. 1), 11–14 (1996).
  • Sagata N. Meiotic metaphase arrest in animal oocytes: its mechanisms and biological significance. Trends Cell. Biol.6, 22–28 (1996).
  • Colledge WH, Carlton MB, Udy GB, Evans MJ. Disruption of c-mos causes parthenogenetic development of unfertilized mouse eggs. Nature370, 65–68 (1994).
  • Hashimoto N, Watanabe N, Furuta Y et al. Parthenogenetic activation of oocytes in c-mos-deficient mice. Nature370, 68–71 (1994).
  • Hyman AA, Mitchison TJ. Two different microtubule-based motor activities with opposite polarities in kinetochores. Nature351, 206–211 (1991).
  • Karsenti E. Mitotic spindle morphogenesis in animal cells. Semin. Cell Biol.2, 251–260 (1991).
  • Doree M, Le Peuch C, Morin N. Onset of chromosome segregation at the metaphase to anaphase transition of the cell cycle. Prog. Cell. Cycle Res.1, 309–318 (1995).
  • Nasmyth K. Disseminating the genome: joining, resolving, and separating sister chromatids during mitosis and meiosis. Annu. Rev. Genet.35, 673–745 (2001).
  • Ma W, Baumann C, Viveiros MM. NEDD1 is crucial for meiotic spindle stability and accurate chromosome segregation in mammalian oocytes. Dev. Biol.339, 439–450 (2010).
  • Schatten G, Simerly C, Schatten H. Microtubule configurations during fertilization, mitosis, and early development in the mouse and the requirement for egg microtubule-mediated motility during mammalian fertilization. Proc. Natl Acad. Sci. USA82, 4152–4156 (1985).
  • Baudat F, Manova K, Yuen JP, Jasin M, Keeney S. Chromosome synapsis defects and sexually dimorphic meiotic progression in mice lacking Spo11. Mol. Cell6, 989–998 (2000).
  • Romanienko PJ, Camerini-Otero RD. The mouse Spo11 gene is required for meiotic chromosome synapsis. Mol. Cell6, 975–987 (2000).
  • Yuan L, Liu JG, Hoja MR, Wilbertz J, Nordqvist K, Hoog C. Female germ cell aneuploidy and embryo death in mice lacking the meiosis-specific protein SCP3. Science296, 1115–1118 (2002).
  • Wang H, Hoog C. Structural damage to meiotic chromosomes impairs DNA recombination and checkpoint control in mammalian oocytes. J. Cell. Biol.173, 485–495 (2006).
  • Kuznetsov S, Pellegrini M, Shuda K et al. RAD51C deficiency in mice results in early prophase I arrest in males and sister chromatid separation at metaphase II in females. J. Cell. Biol.176, 581–592 (2007).
  • Lipkin SM, Moens PB, Wang V et al. Meiotic arrest and aneuploidy in MLH3-deficient mice. Nat. Genet.31, 385–390 (2002).
  • Champoux JJ. DNA topoisomerases: structure, function, and mechanism. Annu. Rev. Biochem.70, 369–413 (2001).
  • Wang JC. Cellular roles of DNA topoisomerases: a molecular perspective. Nat. Rev. Mol. Cell. Biol.3, 430–440 (2002).
  • Cobb J, Reddy RK, Park C, Handel MA. Analysis of expression and function of topoisomerase I and II during meiosis in male mice. Mol. Reprod. Dev.46, 489–498 (1997).
  • Mailhes JB, Marchetti F, Phillips GL Jr, Barnhill DR. Preferential pericentric lesions and aneuploidy induced in mouse oocytes by the topoisomerase II inhibitor etoposide. Teratog. Carcinog. Mutagen.14, 39–51 (1994).
  • Marchetti F, Bishop JB, Lowe X, Generoso WM, Hozier J, Wyrobek AJ. Etoposide induces heritable chromosomal aberrations and aneuploidy during male meiosis in the mouse. Proc. Natl Acad. Sci. USA98, 3952–3957 (2001).
  • Hagstrom KA, Holmes VF, Cozzarelli NR, Meyer BJ. C. elegans condensin promotes mitotic chromosome architecture, centromere organization, and sister chromatid segregation during mitosis and meiosis. Genes Dev.16, 729–742 (2002).
  • Lavoie BD, Hogan E, Koshland D. In vivo dissection of the chromosome condensation machinery: reversibility of condensation distinguishes contributions of condensin and cohesin. J. Cell. Biol.156, 805–815 (2002).
  • Ono T, Fang Y, Spector DL, Hirano T. Spatial and temporal regulation of Condensins I and II in mitotic chromosome assembly in human cells. Mol. Biol. Cell.15, 3296–3308 (2004).
  • Marston AL, Amon A. Meiosis: cellcycle controls shuffle and deal. Nat. Rev. Mol. Cell. Biol.5, 983–997 (2004).
  • Nasmyth K, Schleiffer A. From a single double helix to paired double helices and back. Philos. Trans. R. Soc. Lond. B. Biol. Sci.359, 99–108 (2004).
  • van Heemst D, Heyting C. Sister chromatid cohesion and recombination in meiosis. Chromosoma109, 10–26 (2000).
  • Revenkova E, Jessberger R. Keeping sister chromatids together: cohesins in meiosis. Reproduction130, 783–790 (2005).
  • Prieto I, Pezzi N, Buesa JM et al. STAG2 and Rad21 mammalian mitotic cohesins are implicated in meiosis. EMBO Rep.3, 543–550 (2002).
  • Haering CH, Lowe J, Hochwagen A, Nasmyth K. Molecular architecture of SMC proteins and the yeast cohesin complex. Mol. Cell9, 773–788 (2002).
  • Parra MT, Viera A, Gomez R et al. Involvement of the cohesin Rad21 and SCP3 in monopolar attachment of sister kinetochores during mouse meiosis I. J. Cell. Sci.117, 1221–1234 (2004).
  • Eijpe M, Heyting C, Gross B, Jessberger R. Association of mammalian SMC1 and SMC3 proteins with meiotic chromosomes and synaptonemal complexes. J. Cell. Sci.113(Pt 4), 673–682 (2000).
  • Watanabe Y, Kitajima TS. Shugoshin protects cohesin complexes at centromeres. Philos. Trans. R. Soc. Lond. B. Biol. Sci.360, 515–521, discussion 521 (2005).
  • Parisi S, McKay MJ, Molnar M et al. Rec8p, a meiotic recombination and sister chromatid cohesion phosphoprotein of the Rad21p family conserved from fission yeast to humans. Mol. Cell Biol.19, 3515–3528 (1999).
  • Eijpe M, Offenberg H, Jessberger R, Revenkova E, Heyting C. Meiotic cohesin Rec8 marks the axial elements of rat synaptonemal complexes before cohesins SMC1β and SMC3. J. Cell. Biol.160, 657–670 (2003).
  • Prieto I, Suja JA, Pezzi N et al. Mammalian STAG3 is a cohesin specific to sister chromatid arms in meiosis I. Nat. Cell. Biol.3, 761–766 (2001).
  • Prieto I, Tease C, Pezzi N et al. Cohesin component dynamics during meiotic prophase I in mammalian oocytes. Chromosome Res.12, 197–213 (2004).
  • Revenkova E, Eijpe M, Heyting C, Gross B, Jessberger R. Novel meiosis-specific isoform of mammalian SMC1. Mol. Cell Biol.21, 6984–6998 (2001).
  • Alexandru G, Uhlmann F, Mechtler K, Poupart MA, Nasmyth K. Phosphorylation of the cohesin subunit Scc1 by Polo/Cdc5 kinase regulates sister chromatid separation in yeast. Cell105, 459–472 (2001).
  • Losada A, Hirano M, Hirano T. Cohesin release is required for sister chromatid resolution, but not for condensin-mediated compaction, at the onset of mitosis. Genes Dev.16, 3004–3016 (2002).
  • Sumara I, Vorlaufer E, Stukenberg PT et al. The dissociation of cohesin from chromosomes in prophase is regulated by Polo-like kinase. Mol. Cell9, 515–525 (2002).
  • Clyne RK, Katis VL, Jessop L et al. Polo-like kinase Cdc5 promotes chiasmata formation and cosegregation of sister centromeres at meiosis I. Nat. Cell. Biol.5, 480–485 (2003).
  • Hauf S, Roitinger E, Koch B, Dittrich CM, Mechtler K, Peters JM. Dissociation of cohesin from chromosome arms and loss of arm cohesion during early mitosis depends on phosphorylation of SA2. PLoS Biol.3, e69 (2005).
  • Waizenegger IC, Hauf S, Meinke A, Peters JM. Two distinct pathways remove mammalian cohesin from chromosome arms in prophase and from centromeres in anaphase. Cell103, 399–410 (2000).
  • Uhlmann F. Chromosome cohesion and segregation in mitosis and meiosis. Curr. Opin. Cell. Biol.13, 754–761 (2001).
  • Clarke AS, Tang TT, Ooi DL, Orr-Weaver TL. Polo kinase regulates the Drosophila centromere cohesion protein MEI-S332. Dev. Cell8, 53–64 (2005).
  • Lee JY, Hayashi-Hagihara A, Orr-Weaver TL. Roles and regulation of the Drosophila centromere cohesion protein MEI-S332 family. Philos. Trans. R. Soc. Lond. B. Biol. Sci.360, 543–552 (2005).
  • Pahlavan G, Polanski Z, Kalab P, Golsteyn R, Nigg EA, Maro B. Characterization of Polo-like kinase 1 during meiotic maturation of the mouse oocyte. Dev. Biol.220, 392–400 (2000).
  • Pasierbek P, Jantsch M, Melcher M, Schleiffer A, Schweizer D, Loidl J. A Caenorhabditis elegans cohesion protein with functions in meiotic chromosome pairing and disjunction. Genes Dev.15, 1349–1360 (2001).
  • Siomos MF, Badrinath A, Pasierbek P et al. Separase is required for chromosome segregation during meiosis I in Caenorhabditis elegans. Curr. Biol.11, 1825–1835 (2001).
  • Lee J, Okada K, Ogushi S, Miyano T, Miyake M, Yamashita M. Loss of Rec8 from chromosome arm and centromere region is required for homologous chromosome separation and sister chromatid separation, respectively, in mammalian meiosis. Cell Cycle5, 1448–1455 (2006).
  • Michaelis C, Ciosk R, Nasmyth K. Cohesins: chromosomal proteins that prevent premature separation of sister chromatids. Cell91, 35–45 (1997).
  • Tanaka T, Fuchs J, Loidl J, Nasmyth K. Cohesin ensures bipolar attachment of microtubules to sister centromeres and resists their precocious separation. Nat. Cell. Biol.2, 492–499 (2000).
  • Kamieniecki RJ, Shanks RM, Dawson DS. Slk19p is necessary to prevent separation of sister chromatids in meiosis I. Curr. Biol.10, 1182–1190 (2000).
  • Katis VL, Galova M, Rabitsch KP, Gregan J, Nasmyth K. Maintenance of cohesin at centromeres after meiosis I in budding yeast requires a kinetochore associated protein related to MEI-S332. Curr. Biol.14, 560–572 (2004).
  • Kitajima TS, Kawashima SA, Watanabe Y. The conserved kinetochore protein shugoshin protects centromeric cohesion during meiosis. Nature427, 510–517 (2004).
  • Salic A, Waters JC, Mitchison TJ. Vertebrate shugoshin links sister centromere cohesion and kinetochore microtubule stability in mitosis. Cell118, 567–578 (2004).
  • Rabitsch KP, Gregan J, Schleiffer A, Javerzat JP, Eisenhaber F, Nasmyth K. Two fission yeast homologs of DrosophilaMEI-S332 are required for chromosome segregation during meiosis I and II. Curr. Biol.14, 287–301 (2004).
  • Tang Z, Sun Y, Harley SE, Zou H, Yu H. Human Bub1 protects centromeric sister-chromatid cohesion through Shugoshin during mitosis. Proc. Natl Acad. Sci. USA101, 18012–18017 (2004).
  • McGuinness BE, Hirota T, Kudo NR, Peters JM, Nasmyth K. Shugoshin prevents dissociation of cohesin from centromeres during mitosis in vertebrate cells. PLoS Biol.3, e86 (2005).
  • Marchetti F, Venkatachalam S. The multiple roles of Bub1 in chromosome segregation during mitosis and meiosis. Cell Cycle9, 58–63 (2010).
  • Llano E, Gomez R, Gutierrez-Caballero C et al. Shugoshin-2 is essential for the completion of meiosis but not for mitotic cell division in mice. Genes Dev.22, 2400–2413 (2008).
  • Parra MT, Gomez R, Viera A et al. Sequential assembly of centromeric proteins in male mouse meiosis. PLoS Genet.5, e1000417 (2009).
  • Goulding SE, Earnshaw WC. Shugoshin: a centromeric guardian senses tension. Bioessays27, 588–591 (2005).
  • Wang X, Dai W. Shugoshin, a guardian for sister chromatid segregation. Exp. Cell. Res.310, 1–9 (2005).
  • Gregan J, Rabitsch PK, Sakem B et al. Novel genes required for meiotic chromosome segregation are identified by a high-throughput knockout screen in fission yeast. Curr. Biol.15, 1663–1669 (2005).
  • Cimini D. Detection and correction of merotelic kinetochore orientation by Aurora B and its partners. Cell Cycle6, 1558–1564 (2007).
  • Biggins S, Walczak CE. Captivating capture: how microtubules attach to kinetochores. Curr. Biol.13, R449–R460 (2003).
  • Rieder CL, Maiato H. Stuck in division or passing through: what happens when cells cannot satisfy the spindle assembly checkpoint. Dev. Cell7, 637–651 (2004).
  • Salmon ED, Cimini D, Cameron LA, DeLuca JG. Merotelic kinetochores in mammalian tissue cells. Philos. Trans. R. Soc. Lond. B. Biol. Sci.360, 553–568 (2005).
  • Cimini D. Merotelic kinetochore orientation, aneuploidy, and cancer. Biochim Biophys Acta1786, 32–40 (2008).
  • Amor DJ, Kalitsis P, Sumer H, Choo KH. Building the centromere: from foundation proteins to 3D organization. Trends Cell. Biol.14, 359–368 (2004).
  • Vagnarelli P, Earnshaw WC. Chromosomal passengers: the four-dimensional regulation of mitotic events. Chromosoma113, 211–222 (2004).
  • Simerly C, Balczon R, Brinkley BR, Schatten G. Microinjected centromere [corrected] kinetochore antibodies interfere with chromosome movement in meiotic and mitotic mouse oocytes. J. Cell. Biol.111, 1491–1504 (1990).
  • Rieder CL, Salmon ED. The vertebrate cell kinetochore and its roles during mitosis. Trends Cell. Biol.8, 310–318 (1998).
  • Craig JM, Earnshaw WC, Vagnarelli P. Mammalian centromeres: DNA sequence, protein composition, and role in cell cycle progression. Exp. Cell. Res.246, 249–262 (1999).
  • Earnshaw WC, Cooke CA. Analysis of the distribution of the INCENPs throughout mitosis reveals the existence of a pathway of structural changes in the chromosomes during metaphase and early events in cleavage furrow formation. J. Cell. Sci.98(Pt 4), 443–461 (1991).
  • Meraldi P, Honda R, Nigg EA. Aurora kinases link chromosome segregation and cell division to cancer susceptibility. Curr. Opin. Genet. Dev.14, 29–36 (2004).
  • Giet R, Petretti C, Prigent C. Aurora kinases, aneuploidy and cancer, a coincidence or a real link? Trends Cell. Biol.15, 241–250 (2005).
  • Miyazaki WY, Orr-Weaver TL. Sister-chromatid cohesion in mitosis and meiosis. Annu. Rev. Genet.28, 167–187 (1994).
  • Stern BM. Mitosis: Aurora gives chromosomes a healthy stretch. Curr. Biol.12, R316–R318 (2002).
  • Tanaka TU, Rachidi N, Janke C et al. Evidence that the Ipl1–Sli15 (Aurora kinase–INCENP) complex promotes chromosome bi-orientation by altering kinetochore–spindle pole connections. Cell108, 317–329 (2002).
  • Ahonen LJ, Kallio MJ, Daum JR et al. Polo-like kinase 1 creates the tension-sensing 3F3/2 phosphoepitope and modulates the association of spindle-checkpoint proteins at kinetochores. Curr. Biol.15, 1078–1089 (2005).
  • Kallio MJ, McCleland ML, Stukenberg PT, Gorbsky GJ. Inhibition of Aurora B kinase blocks chromosome segregation, overrides the spindle checkpoint, and perturbs microtubule dynamics in mitosis. Curr. Biol.12, 900–905 (2002).
  • Knowlton AL, Lan W, Stukenberg PT. Aurora B is enriched at merotelic attachment sites, where it regulates MCAK. Curr. Biol.16, 1705–1710 (2006).
  • Vogt E, Sanhaji M, Klein W, Seidel T, Wordeman L, Eichenlaub-Ritter U. MCAK is present at centromeres, midspindle and chiasmata and involved in silencing of the spindle assembly checkpoint in mammalian oocytes. Mol. Hum. Reprod.16(9), 665–684 (2010).
  • Lee BH, Kiburz BM, Amon A. Spo13 maintains centromeric cohesion and kinetochore coorientation during meiosis I. Curr. Biol.14, 2168–2182 (2004).
  • Katis VL, Matos J, Mori S, Shirahige K, Zachariae W, Nasmyth K. Spo13 facilitates monopolin recruitment to kinetochores and regulates maintenance of centromeric cohesion during yeast meiosis. Curr. Biol.14, 2183–2196 (2004).
  • Adams RR, Eckley DM, Vagnarelli P et al. Human INCENP colocalizes with the Aurora-B/AIRK2 kinase on chromosomes and is overexpressed in tumour cells. Chromosoma110, 65–74 (2001).
  • Shang C, Hazbun TR, Cheeseman IM et al. Kinetochore protein interactions and their regulation by the Aurora kinase Ipl1p. Mol. Biol. Cell.14, 3342–3355 (2003).
  • Dewar H, Tanaka K, Nasmyth K, Tanaka TU. Tension between two kinetochores suffices for their bi-orientation on the mitotic spindle. Nature428, 93–97 (2004).
  • Bolton MA, Lan W, Powers SE, McCleland ML, Kuang J, Stukenberg PT. Aurora B kinase exists in a complex with survivin and INCENP and its kinase activity is stimulated by survivin binding and phosphorylation. Mol. Biol. Cell.13, 3064–3077 (2002).
  • Tong C, Fan HY, Lian L et al. Polo-like kinase-1 is a pivotal regulator of microtubule assembly during mouse oocyte meiotic maturation, fertilization, and early embryonic mitosis. Biol. Reprod.67, 546–554 (2002).
  • Ditchfield C, Johnson VL, Tighe A et al. Aurora B couples chromosome alignment with anaphase by targeting BubR1, Mad2, and CenP-E to kinetochores. J. Cell. Biol.161, 267–280 (2003).
  • Goto H, Kiyono T, Tomono Y et al. Complex formation of Plk1 and INCENP required for metaphase–anaphase transition. Nat. Cell. Biol.8, 180–187 (2006).
  • Taylor SS, Hussein D, Wang Y, Elderkin S, Morrow CJ. Kinetochore localisation and phosphorylation of the mitotic checkpoint components Bub1 and BubR1 are differentially regulated by spindle events in human cells. J. Cell. Sci.114, 4385–4395 (2001).
  • Lens SM, Medema RH. The survivin/Aurora B complex: its role in coordinating tension and attachment. Cell Cycle2, 507–510 (2003).
  • Johnson VL, Scott MI, Holt SV, Hussein D, Taylor SS. Bub1 is required for kinetochore localization of BubR1, CenP-E, CenP-F and Mad2, and chromosome congression. J. Cell. Sci.117, 1577–1589 (2004).
  • Lampson MA, Renduchitala K, Khodjakov A, Kapoor TM. Correcting improper chromosome–spindle attachments during cell division. Nat. Cell. Biol.6, 232–237 (2004).
  • Taylor SS, Scott MI, Holland AJ. The spindle checkpoint: a quality control mechanism which ensures accurate chromosome segregation. Chromosome Res.12, 599–616 (2004).
  • Hauf S, Cole RW, LaTerra S et al. The small molecule Hesperadin reveals a role for Aurora B in correcting kinetochore–microtubule attachment and in maintaining the spindle assembly checkpoint. J. Cell. Biol.161, 281–294 (2003).
  • Nguyen HG, Chinnappan D, Urano T, Ravid K. Mechanism of Aurora-B degradation and its dependency on intact KEN and A-boxes: identification of an aneuploidy-promoting property. Mol. Cell Biol.25, 4977–4992 (2005).
  • Mailhes JB. Faulty spindle checkpoint and cohesion protein activities predispose oocytes to premature chromosome separation and aneuploidy. Environ. Mol. Mutagen.49, 642–658 (2008).
  • Rieder CL, Palazzo RE. Colcemid and the mitotic cycle. J. Cell. Sci.102(Pt 3), 387–392 (1992).
  • Rieder CL, Schultz A, Cole R, Sluder G. Anaphase onset in vertebrate somatic cells is controlled by a checkpoint that monitors sister kinetochore attachment to the spindle. J. Cell. Biol.127, 1301–1310 (1994).
  • Andreassen PR, Martineau SN, Margolis RL. Chemical induction of mitotic checkpoint override in mammalian cells results in aneuploidy following a transient tetraploid state. Mutat. Res.372, 181–194 (1996).
  • Lee JY, Orr-Weaver TL. The molecular basis of sister-chromatid cohesion. Annu. Rev. Cell. Dev. Biol.17, 753–777 (2001).
  • Fang G. Checkpoint protein BubR1 acts synergistically with Mad2 to inhibit anaphase-promoting complex. Mol. Biol. Cell.13, 755–766 (2002).
  • Vigneron S, Prieto S, Bernis C, Labbe JC, Castro A, Lorca T. Kinetochore localization of spindle checkpoint proteins: who controls whom? Mol. Biol. Cell.15, 4584–4596 (2004).
  • Li M, Li S, Yuan J et al. Bub3 is a spindle assembly checkpoint protein regulating chromosome segregation during mouse oocyte meiosis. PLoS One4, e7701 (2009).
  • Li Y, Benezra R. Identification of a human mitotic checkpoint gene: hsMAD2. Science274, 246–248 (1996).
  • Nicklas RB. How cells get the right chromosomes. Science275, 632–637 (1997).
  • Taylor SS, Ha E, McKeon F. The human homologue of Bub3 is required for kinetochore localization of Bub1 and a Mad3/Bub1-related protein kinase. J. Cell. Biol.142, 1–11 (1998).
  • Musacchio A, Hardwick KG. The spindle checkpoint: structural insights into dynamic signalling. Nat. Rev. Mol. Cell. Biol.3, 731–741 (2002).
  • Bharadwaj R, Yu H. The spindle checkpoint, aneuploidy, and cancer. Oncogene23, 2016–2027 (2004).
  • Howell BJ, Moree B, Farrar EM, Stewart S, Fang G, Salmon ED. Spindle checkpoint protein dynamics at kinetochores in living cells. Curr. Biol.14, 953–964 (2004).
  • Nasmyth K. How do so few control so many? Cell120, 739–746 (2005).
  • Wassmann K, Niault T, Maro B. Metaphase I arrest upon activation of the Mad2-dependent spindle checkpoint in mouse oocytes. Curr. Biol.13, 1596–1608 (2003).
  • Tsurumi C, Hoffmann S, Geley S, Graeser R, Polanski Z. The spindle assembly checkpoint is not essential for CSF arrest of mouse oocytes. J. Cell. Biol.167, 1037–1050 (2004).
  • Homer HA, McDougall A, Levasseur M, Yallop K, Murdoch AP, Herbert M. Mad2 prevents aneuploidy and premature proteolysis of cyclin B and securin during meiosis I in mouse oocytes. Genes Dev.19, 202–207 (2005).
  • Zhang D, Ma W, Li YH et al. Intra-oocyte localization of Mad2 and its relationship with kinetochores, microtubules, and chromosomes in rat oocytes during meiosis. Biol. Reprod.71, 740–748 (2004).
  • Kallio M, Eriksson JE, Gorbsky GJ. Differences in spindle association of the mitotic checkpoint protein Mad2 in mammalian spermatogenesis and oogenesis. Dev. Biol.225, 112–123 (2000).
  • Ma W, Zhang D, Hou Y et al. Reduced expression of Mad2, BCL2, and MAP kinase activity in pig oocytes after in vitro aging are associated with defects in sister chromatid segregation during meiosis II and embryo fragmentation after activation. Biol. Reprod.72, 373–383 (2005).
  • Zhang D, Li M, Ma W et al. Localization of mitotic arrest deficient 1 (Mad1) in mouse oocytes during the first meiosis and its functions as a spindle checkpoint protein. Biol. Reprod.72, 58–68 (2005).
  • Chung E, Chen RH. Spindle checkpoint requires Mad1-bound and Mad1-free Mad2. Mol. Biol. Cell.13, 1501–1511 (2002).
  • Brunet S, Pahlavan G, Taylor S, Maro B. Functionality of the spindle checkpoint during the first meiotic division of mammalian oocytes. Reproduction126, 443–450 (2003).
  • Leland S, Nagarajan P, Polyzos A et al. Heterozygosity for a Bub1 mutation causes female-specific germ cell aneuploidy in mice. Proc. Natl Acad. Sci. USA106, 12776–12781 (2009).
  • McGuinness BE, Anger M, Kouznetsova A et al. Regulation of APC/C activity in oocytes by a Bub1-dependent spindle assembly checkpoint. Curr. Biol.19, 369–380 (2009).
  • Reis A, Madgwick S, Chang HY, Nabti I, Levasseur M, Jones KT. Prometaphase APCcdh1 activity prevents non-disjunction in mammalian oocytes. Nat. Cell. Biol.9, 1192–1198 (2007).
  • Dai W, Wang Q, Liu T et al. Slippage of mitotic arrest and enhanced tumor development in mice with BubR1 haploinsufficiency. Cancer Res.64, 440–445 (2004).
  • Michel LS, Liberal V, Chatterjee A et al. Mad2 haplo-insufficiency causes premature anaphase and chromosome instability in mammalian cells. Nature409, 355–359 (2001).
  • Hanks S, Coleman K, Reid S et al. Constitutional aneuploidy and cancer predisposition caused by biallelic mutations in BUB1B. Nat. Genet.36, 1159–1161 (2004).
  • Shonn MA, McCarroll R, Murray AW. Requirement of the spindle checkpoint for proper chromosome segregation in budding yeast meiosis. Science289, 300–303 (2000).
  • Michel L, Diaz-Rodriguez E, Narayan G, Hernando E, Murty VV, Benezra R. Complete loss of the tumor suppressor Mad2 causes premature cyclin B degradation and mitotic failure in human somatic cells. Proc. Natl Acad. Sci. USA101, 4459–4464 (2004).
  • Dobles M, Liberal V, Scott ML, Benezra R, Sorger PK. Chromosome missegregation and apoptosis in mice lacking the mitotic checkpoint protein Mad2. Cell101, 635–645 (2000).
  • Yu H. Regulation of APC–Cdc20 by the spindle checkpoint. Curr. Opin. Cell. Biol.14, 706–714 (2002).
  • Zhang Y, Lees E. Identification of an overlapping binding domain on Cdc20 for Mad2 and anaphase-promoting complex: model for spindle checkpoint regulation. Mol. Cell Biol.21, 5190–5199 (2001).
  • Morrow CJ, Tighe A, Johnson VL, Scott MI, Ditchfield C, Taylor SS. Bub1 and Aurora B cooperate to maintain BubR1-mediated inhibition of APC/CCdc20. J. Cell. Sci.118, 3639–3652 (2005).
  • Kawashima SA, Yamagishi Y, Honda T, Ishiguro K, Watanabe Y. Phosphorylation of H2A by Bub1 prevents chromosomal instability through localizing shugoshin. Science327, 172–177 (2010).
  • Kitajima TS, Hauf S, Ohsugi M, Yamamoto T, Watanabe Y. Human Bub1 defines the persistent cohesion site along the mitotic chromosome by affecting Shugoshin localization. Curr. Biol.15, 353–359 (2005).
  • Meraldi P, Sorger PK. A dual role for Bub1 in the spindle checkpoint and chromosome congression. EMBO J.24, 1621–1633 (2005).
  • Yin S, Wang Q, Liu JH et al. Bub1 prevents chromosome misalignment and precocious anaphase during mouse oocyte meiosis. Cell Cycle5, 2130–2137 (2006).
  • Perera D, Tilston V, Hopwood JA, Barchi M, Boot-Handford RP, Taylor SS. Bub1 maintains centromeric cohesion by activation of the spindle checkpoint. Dev. Cell13, 566–579 (2007).
  • Kienitz A, Vogel C, Morales I, Muller R, Bastians H. Partial downregulation of Mad1 causes spindle checkpoint inactivation and aneuploidy, but does not confer resistance towards taxol. Oncogene24, 4301–4310 (2005).
  • Baker DJ, Jeganathan KB, Cameron JD et al. BubR1 insufficiency causes early onset of aging-associated phenotypes and infertility in mice. Nat. Genet.36, 744–749 (2004).
  • Kalitsis P, Earle E, Fowler KJ, Choo KH. Bub3 gene disruption in mice reveals essential mitotic spindle checkpoint function during early embryogenesis. Genes Dev.14, 2277–2282 (2000).
  • Ikui AE, Yang CP, Matsumoto T, Horwitz SB. Low concentrations of taxol cause mitotic delay followed by premature dissociation of p55CDC from Mad2 and BubR1 and abrogation of the spindle checkpoint, leading to aneuploidy. Cell Cycle4, 1385–1388 (2005).
  • Mailhes JB, Carabatsos MJ, Young D, London SN, Bell M, Albertini DF. Taxol-induced meiotic maturation delay, spindle defects, and aneuploidy in mouse oocytes and zygotes. Mutat. Res.423, 79–90 (1999).
  • Homer HA, McDougall A, Levasseur M, Murdoch AP, Herbert M. RNA interference in meiosis I human oocytes: towards an understanding of human aneuploidy. Mol. Hum. Reprod.11, 397–404 (2005).
  • Steuerwald N, Cohen J, Herrera RJ, Sandalinas M, Brenner CA. Association between spindle assembly checkpoint expression and maternal age in human oocytes. Mol. Hum. Reprod.7, 49–55 (2001).
  • Grondahl ML, Yding Andersen C, Bogstad J, Nielsen FC, Meinertz H, Borup R. Gene expression profiles of single human mature oocytes in relation to age. Hum. Reprod.25, 957–968 (2010).
  • Duncan FE, Chiang T, Schultz RM, Lampson MA. Evidence that a defective spindle assembly checkpoint is not the primary cause of maternal age-associated aneuploidy in mouse eggs. Biol. Reprod.81, 768–776 (2009).
  • Glickman MH, Ciechanover A. The ubiquitin–proteasome proteolytic pathway: destruction for the sake of construction. Physiol Rev.82, 373–428 (2002).
  • Craig JM, Choo KH. Kiss and break up – a safe passage to anaphase in mitosis and meiosis. Chromosoma114, 252–262 (2005).
  • Goldberg AL. Functions of the proteasome: the lysis at the end of the tunnel. Science268, 522–523 (1995).
  • Coux O, Tanaka K, Goldberg AL. Structure and functions of the 20S and 26S proteasomes. Annu. Rev. Biochem.65, 801–847 (1996).
  • Holt JE, Weaver J, Jones KT. Spatial regulation of APCCdh1-induced cyclin B1 degradation maintains G2 arrest in mouse oocytes. Development137, 1297–1304 (2010).
  • Glover DM, Hagan IM, Tavares AA. Polo-like kinases: a team that plays throughout mitosis. Genes Dev.12, 3777–3787 (1998).
  • Sumara I, Gimenez-Abian JF, Gerlich D et al. Roles of Polo-like kinase 1 in the assembly of functional mitotic spindles. Curr. Biol.14, 1712–1722 (2004).
  • Zachariae W, Nasmyth K. Whose end is destruction: cell division and the anaphase-promoting complex. Genes Dev.13, 2039–2058 (1999).
  • Peters JM. The anaphase-promoting complex: proteolysis in mitosis and beyond. Mol. Cell9, 931–943 (2002).
  • Cohen-Fix O, Peters JM, Kirschner MW, Koshland D. Anaphase initiation in Saccharomyces cerevisiae is controlled by the APC-dependent degradation of the anaphase inhibitor Pds1p. Genes Dev.10, 3081–3093 (1996).
  • Uhlmann F, Lottspeich F, Nasmyth K. Sister-chromatid separation at anaphase onset is promoted by cleavage of the cohesin subunit Scc1. Nature400, 37–42 (1999).
  • Salah SM, Nasmyth K. Destruction of the securin Pds1p occurs at the onset of anaphase during both meiotic divisions in yeast. Chromosoma109, 27–34 (2000).
  • Hagting A, Den Elzen N, Vodermaier HC, Waizenegger IC, Peters JM, Pines J. Human securin proteolysis is controlled by the spindle checkpoint and reveals when the APC/C switches from activation by Cdc20 to Cdh1. J. Cell. Biol.157, 1125–1137 (2002).
  • Nasmyth K. Segregating sister genomes: the molecular biology of chromosome separation. Science297, 559–565 (2002).
  • Waizenegger I, Gimenez-Abian JF, Wernic D, Peters JM. Regulation of human separase by securin binding and autocleavage. Curr. Biol.12, 1368–1378 (2002).
  • Buonomo SB, Clyne RK, Fuchs J, Loidl J, Uhlmann F, Nasmyth K. Disjunction of homologous chromosomes in meiosis I depends on proteolytic cleavage of the meiotic cohesin Rec8 by separin. Cell103, 387–398 (2000).
  • Jallepalli PV, Waizenegger IC, Bunz F et al. Securin is required for chromosomal stability in human cells. Cell105, 445–457 (2001).
  • Agarwal R, Cohen-Fix O. Mitotic regulation: the fine tuning of separase activity. Cell Cycle1, 255–257 (2002).
  • Herbert M, Levasseur M, Homer H, Yallop K, Murdoch A, McDougall A. Homologue disjunction in mouse oocytes requires proteolysis of securin and cyclin B1. Nat. Cell. Biol.5, 1023–1025 (2003).
  • Kirsch-Volders M, Cundari E, Verdoodt B. Towards a unifying model for the metaphase/anaphase transition. Mutagenesis13, 321–335 (1998).
  • Zhang D, Nicklas RB. ‘Anaphase’ and cytokinesis in the absence of chromosomes. Nature382, 466–468 (1996).
  • Nasmyth K, Peters JM, Uhlmann F. Splitting the chromosome: cutting the ties that bind sister chromatids. Science288, 1379–1385 (2000).
  • Lejeune J, Gautier M, Turpin R. [Study of somatic chromosomes from 9 mongoloid children.]. CR Hebd. Seances Acad. Sci.248, 1721–1722 (1959).
  • Penrose L. The relative effects of paternal and maternal age in mongolism. J. Genet.27, 219–224 (1933).
  • Niault T, Hached K, Sotillo R et al. Changing Mad2 levels affects chromosome segregation and spindle assembly checkpoint control in female mouse meiosis I. PLoS One2, e1165 (2007).
  • Schlecht U, Primig M. Mining meiosis and gametogenesis with DNA microarrays. Reproduction125, 447–456 (2003).
  • Hutchins JR, Toyoda Y, Hegemann B et al. Systematic analysis of human protein complexes identifies chromosome segregation proteins. Science328, 593–599 (2010).
  • Bettencourt-Dias M, Giet R, Sinka R et al. Genome-wide survey of protein kinases required for cell cycle progression. Nature432, 980–987 (2004).
  • Prawitt D, Brixel L, Spangenberg C et al. RNAi knock-down mice: an emerging technology for post-genomic functional genetics. Cytogenet. Genome Res.105, 412–421 (2004).
  • Stein P, Svoboda P, Schultz RM. Transgenic RNAi in mouse oocytes: a simple and fast approach to study gene function. Dev. Biol.256, 187–193 (2003).

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.