Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Human aneuploidy: mechanisms and new insights into an age-old problem

Key Points

  • Aneuploidy is extraordinarily common in humans, occurring in an estimated 20–40% of all conceptions. It is the most common cause of miscarriages and congenital defects in our species and is a leading impediment to the treatment of infertility.

  • Most aneuploidy results from maternal meiotic nondisjunctional errors. However, there is remarkable variation among chromosomes in the way in which these errors originate, indicating that there are multiple mechanisms by which human aneuploidy occurs.

  • Studies of human fetal oocytes indicate a high level of recombination errors, indicating that some oocytes are predisposed to nondisjoin because of events that occurred before birth.

  • Cell cycle control checkpoints that operate in meiotic prophase and at the metaphase–anaphase transition are less stringent in females than in males. Consequently, abnormal cells that are eliminated in spermatogenesis may escape detection in the female, ultimately leading to aneuploid eggs.

  • Studies from mice suggest that loss of cohesin proteins over the reproductive life of the female contribute to the maternal age effect on human trisomy.

  • Exposure to endocrine disruptors (for example, bisphenol A) disrupts oogenesis at multiple stages and predisposes the oocyte to aneuploidy.

Abstract

Trisomic and monosomic (aneuploid) embryos account for at least 10% of human pregnancies and, for women nearing the end of their reproductive lifespan, the incidence may exceed 50%. The errors that lead to aneuploidy almost always occur in the oocyte but, despite intensive investigation, the underlying molecular basis has remained elusive. Recent studies of humans and model organisms have shed new light on the complexity of meiotic defects, providing evidence that the age-related increase in errors in the human female is not attributable to a single factor but to an interplay between unique features of oogenesis and a host of endogenous and exogenous factors.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Oogenesis and the female meiotic cycle.
Figure 2: Releasing sisters: normal and premature loss of cohesion.

Similar content being viewed by others

References

  1. Lejeune, J., Turpin, R. & Gautier, M. Mongolism; a chromosomal disease (trisomy). Bull. Acad. Natl Med. 143, 256–265 (1959).

    CAS  PubMed  Google Scholar 

  2. Jacobs, P. A., Baikie, A. G., Court Brown, W. M. & Strong, J. A. The somatic chromosomes in mongolism. Lancet 1, 710 (1959).

    Article  CAS  PubMed  Google Scholar 

  3. Hassold, T. & Hunt, P. To err (meiotically) is human: the genesis of human aneuploidy. Nature Rev. Genet. 2, 280–291 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Gaulden, M. E. Maternal age effect: the enigma of Down syndrome and other trisomic conditions. Mutat. Res. 296, 69–88 (1992).

    Article  CAS  PubMed  Google Scholar 

  5. Jacobs, P. A. The chromosome complement of human gametes. Oxf. Rev. Reprod. Biol. 14, 47–72 (1992).

    CAS  PubMed  Google Scholar 

  6. Jamieson, M. E., Coutts, J. R. & Connor, J. M. The chromosome constitution of human preimplantation embryos fertilized in vitro. Hum. Reprod. 9, 709–715 (1994).

    Article  CAS  PubMed  Google Scholar 

  7. Pellestor, F., Andreo, B., Anahory, T. & Hamamah, S. The occurrence of aneuploidy in human: lessons from the cytogenetic studies of human oocytes. Eur. J. Med. Genet. 49, 103–116 (2006).

    Article  PubMed  Google Scholar 

  8. Pacchierotti, F., Adler, I. D., Eichenlaub-Ritter, U. & Mailhes, J. B. Gender effects on the incidence of aneuploidy in mammalian germ cells. Environ. Res. 104, 46–69 (2007).

    Article  CAS  PubMed  Google Scholar 

  9. Fragouli, E. et al. Comprehensive molecular cytogenetic analysis of the human blastocyst stage. Hum. Reprod. 23, 2596–2608 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Fragouli, E. et al. The cytogenetics of polar bodies: insights into female meiosis and the diagnosis of aneuploidy. Mol. Hum. Reprod. 17, 286–295 (2011).

    Article  PubMed  Google Scholar 

  11. Gabriel, A. S. et al. Array comparative genomic hybridisation on first polar bodies suggests that non-disjunction is not the predominant mechanism leading to aneuploidy in humans. J. Med. Genet. 48, 433–437 (2011).

    Article  CAS  PubMed  Google Scholar 

  12. Gutierrez-Mateo, C. et al. Validation of microarray comparative genomic hybridization for comprehensive chromosome analysis of embryos. Fertil. Steril. 95, 953–958 (2011).

    Article  CAS  PubMed  Google Scholar 

  13. Treff, N. R. et al. Single nucleotide polymorphism microarray-based concurrent screening of 24-chromosome aneuploidy and unbalanced translocations in preimplantation human embryos. Fertil. Steril. 95, 1606–1612.e2 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Treff, N. R. et al. SNP microarray-based 24 chromosome aneuploidy screening is significantly more consistent than FISH. Mol. Hum. Reprod. 16, 583–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Geraedts, J. et al. Polar body array CGH for prediction of the status of the corresponding oocyte. Part I: clinical results. Hum. Reprod. 26, 3173–3180 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Lamb, N. E. et al. Susceptible chiasmate configurations of chromosome 21 predispose to non-disjunction in both maternal meiosis I and meiosis II. Nature Genet. 14, 400–405 (1996).

    Article  CAS  PubMed  Google Scholar 

  17. Lynn, A. et al. Covariation of synaptonemal complex length and mammalian meiotic exchange rates. Science 296, 2222–2225 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Cheng, E. Y. et al. Meiotic recombination in human oocytes. PLoS Genet. 5, e1000661 (2009). This is a study of human fetal oocytes that provides evidence that recombination errors occurring during fetal development set the stage for nondisjunction in the adult.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Baudat, F. et al. PRDM9 is a major determinant of meiotic recombination hotspots in humans and mice. Science 327, 836–840 (2010).

    Article  CAS  PubMed  Google Scholar 

  20. Kong, A. et al. Sequence variants in the RNF212 gene associate with genome-wide recombination rate. Science 319, 1398–1401 (2008).

    Article  CAS  PubMed  Google Scholar 

  21. Lynn, A., Ashley, T. & Hassold, T. Variation in human meiotic recombination. Annu. Rev. Genom. Hum. Genet. 5, 317–349 (2004).

  22. O'Connell, M. J., Walworth, N. C. & Carr, A. M. The G2-phase DNA-damage checkpoint. Trends Cell Biol. 10, 296–303 (2000).

    Article  CAS  PubMed  Google Scholar 

  23. Hochwagen, A. & Amon, A. Checking your breaks: surveillance mechanisms of meiotic recombination. Curr. Biol. 16, R217–R228 (2006).

    Article  CAS  PubMed  Google Scholar 

  24. Li, X. C. & Schimenti, J. C. Mouse pachytene checkpoint 2 (trip13) is required for completing meiotic recombination but not synapsis. PLoS Genet. 3, e130 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Hunt, P. A. & Hassold, T. J. Sex matters in meiosis. Science 296, 2181–2183 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Celeste, A. et al. Genomic instability in mice lacking histone H2AX. Science 296, 922–927 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bannister, L. A. et al. A dominant, recombination-defective allele of Dmc1 causing male-specific sterility. PLoS Biol. 5, e105 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Kuznetsov, S. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Herran, Y. et al. The cohesin subunit RAD21L functions in meiotic synapsis and exhibits sexual dimorphism in fertility. EMBO J. 30, 3091–3105 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. McKee, B. D. & Handel, M. A. Sex chromosomes, recombination, and chromatin conformation. Chromosoma 102, 71–80 (1993).

    Article  CAS  PubMed  Google Scholar 

  31. Turner, J. M. Meiotic sex chromosome inactivation. Development 134, 1823–1831 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Burgoyne, P. S., Mahadevaiah, S. K. & Turner, J. M. The consequences of asynapsis for mammalian meiosis. Nature Rev. Genet. 10, 207–216 (2009). This is an informative Review of the meiotic consequences of synaptic defects that emphasizes the mechanisms and consequences of transcriptional silencing of unsynapsed regions.

    Article  CAS  PubMed  Google Scholar 

  33. Cloutier, J. M. & Turner, J. M. Meiotic sex chromosome inactivation. Curr. Biol. 20, R962–963 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Baarends, W. M. et al. Silencing of unpaired chromatin and histone H2A ubiquitination in mammalian meiosis. Mol. Cell. Biol. 25, 1041–1053 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Turner, J. M. et al. Silencing of unsynapsed meiotic chromosomes in the mouse. Nature Genet. 37, 41–47 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Mahadevaiah, S. K. et al. Extensive meiotic asynapsis in mice antagonizes meiotic silencing of unsynapsed chromatin and consequently disrupts meiotic sex chromosome inactivation. J. Cell Biol. 182, 263–276 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Royo, H. et al. Evidence that meiotic sex chromosome inactivation is essential for male fertility. Curr. Biol. 20, 2117–2123 (2010).

    Article  CAS  PubMed  Google Scholar 

  38. Homolka, D., Jansa, P. & Forejt, J. Genetically enhanced asynapsis of autosomal chromatin promotes transcriptional dysregulation and meiotic failure. Chromosoma 121, 91–104 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Burgoyne, P. S. & Baker, T. G. Perinatal oocyte loss in XO mice and its implications for the aetiology of gonadal dysgenesis in XO women. J. Reprod. Fertil. 75, 633–645 (1985).

    Article  CAS  PubMed  Google Scholar 

  40. de Boer, P. & de Jong, J. H. in Fertility and Chromosome Pairing: Recent Studies in Plants and Animals (ed. Gilles, C. B.) 77–107 (CRC Press, 1989).

    Google Scholar 

  41. Baudat, F., Manova, K., Yuen, J. P., Jasin, M. & Keeney, S. Chromosome synapsis defects and sexually dimorphic meiotic progression in mice lacking Spo11. Mol. Cell 6, 989–998 (2000).

    Article  CAS  PubMed  Google Scholar 

  42. Hassold, T. J. & Jacobs, P. A. Trisomy in man. Annu. Rev. Genet. 18, 69–97 (1984).

    Article  CAS  PubMed  Google Scholar 

  43. Hunt, P. & Hassold, T. Female meiosis: coming unglued with age. Curr. Biol. 20, R699–702 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Henderson, S. A. & Edwards, R. G. Chiasma frequency and maternal age in mammals. Nature 218, 22–28 (1968).

    Article  CAS  PubMed  Google Scholar 

  45. Koehler, K. E. et al. Spontaneous X chromosome MI and MII nondisjunction events in Drosophila melanogaster oocytes have different recombinational histories. Nature Genet. 14, 406–414 (1996).

    Article  CAS  PubMed  Google Scholar 

  46. Ross, L. O., Maxfield, R. & Dawson, D. Exchanges are not equally able to enhance meiotic chromosome segregation in yeast. Proc. Natl Acad. Sci. USA 93, 4979–4983 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Angell, R. R. Predivision in human oocytes at meiosis I: a mechanism for trisomy formation in man. Hum. Genet. 86, 383–387 (1991).

    Article  CAS  PubMed  Google Scholar 

  48. Jeffreys, C. A., Burrage, P. S. & Bickel, S. E. A model system for increased meiotic nondisjunction in older oocytes. Curr. Biol. 13, 498–503 (2003).

    Article  CAS  PubMed  Google Scholar 

  49. Hodges, C. A., Revenkova, E., Jessberger, R., Hassold, T. J. & Hunt, P. A. SMC1β-deficient female mice provide evidence that cohesins are a missing link in age-related nondisjunction. Nature Genet. 37, 1351–1355 (2005).

    Article  CAS  PubMed  Google Scholar 

  50. Liu, L. & Keefe, D. L. Defective cohesin is associated with age-dependent misaligned chromosomes in oocytes. Reprod. Biomed. Online 16, 103–112 (2008).

    Article  CAS  PubMed  Google Scholar 

  51. Chiang, T., Duncan, F. E., Schindler, K., Schultz, R. M. & Lampson, M. A. Evidence that weakened centromere cohesion is a leading cause of age-related aneuploidy in oocytes. Curr. Biol. 20, 1522–1528 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Lister, L. M. et al. Age-related meiotic segregation errors in Mammalian oocytes are preceded by depletion of cohesin and Sgo2. Curr. Biol. 20, 1511–1521 (2010).

    Article  CAS  PubMed  Google Scholar 

  53. Chiang, T., Schultz, R. M. & Lampson, M. Meiotic origins of maternal age-related aneuploidy. Biol. Reprod. 86, 1–7 (2011).

    Google Scholar 

  54. Revenkova, E., Herrmann, K., Adelfalk, C. & Jessberger, R. Oocyte cohesin expression restricted to predictyate stages provides full fertility and prevents aneuploidy. Curr. Biol. 20, 1529–1533 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Tachibana-Konwalski, K. et al. Rec8-containing cohesin maintains bivalents without turnover during the growing phase of mouse oocytes. Genes Dev. 24, 2505–2516 (2010). This series of recent papers focuses on meiotic cohesins during oogenesis in the mouse. References 50–53 link loss of cohesin proteins with maternal age-dependent aneuploidy. References 54 and 55 provide evidence that cohesin proteins loaded during fetal development are necessary and sufficient to orchestrate meiotic chromosome segregation in the adult.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Angell, R. R., Xian, J., Keith, J., Ledger, W. & Baird, D. T. First meiotic division abnormalities in human oocytes: mechanism of trisomy formation. Cytogenet. Cell Genet. 65, 194–202 (1994).

    Article  CAS  PubMed  Google Scholar 

  57. 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).

    PubMed  Google Scholar 

  58. Fisher, J. M., Harvey, J. F., Morton, N. E. & Jacobs, P. A. Trisomy 18: studies of the parent and cell division of origin and the effect of aberrant recombination on nondisjunction. Am. J. Hum. Genet. 56, 669–675 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Bugge, M. et al. Non-disjunction of chromosome 18. Hum. Mol. Genet. 7, 661–669 (1998).

    Article  CAS  PubMed  Google Scholar 

  60. Hassold, T., Merrill, M., Adkins, K., Freeman, S. & Sherman, S. Recombination and maternal age-dependent nondisjunction: molecular studies of trisomy 16. Am. J. Hum. Genet. 57, 867–874 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Oliver, T. R. et al. New insights into human nondisjunction of chromosome 21 in oocytes. PLoS Genet. 4, e1000033 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Bond, D. J. & Chandley, A. C. in Aneuploidy 83–90 (Oxford Univ. Press, 1983).

    Google Scholar 

  63. Garcia-Cruz, R. et al. Dynamics of cohesin proteins REC8, STAG3, SMC1 β and SMC3 are consistent with a role in sister chromatid cohesion during meiosis in human oocytes. Hum. Reprod. 25, 2316–2327 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Musacchio, A. & Salmon, E. D. The spindle-assembly checkpoint in space and time. Nature Rev. Mol. Cell Biol. 8, 379–393 (2007).

    Article  CAS  Google Scholar 

  65. Kot, M. C. & Handel, M. A. Spermatogenesis in XO,Sxr mice: role of the Y chromosome. J. Exp. Zool. 256, 92–105 (1990).

    Article  CAS  PubMed  Google Scholar 

  66. Sutcliffe, M. J., Darling, S. M. & Burgoyne, P. S. Spermatogenesis in XY, XYSxra and XOSxra mice: a quantitative analysis of spermatogenesis throughout puberty. Mol. Reprod. Dev. 30, 81–89 (1991).

    Article  CAS  PubMed  Google Scholar 

  67. LeMaire-Adkins, R., Radke, K. & Hunt, P. A. Lack of checkpoint control at the metaphase/anaphase transition: a mechanism of meiotic nondisjunction in mammalian females. J. Cell Biol. 139, 1611–1619 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kouznetsova, A., Lister, L., Nordenskjold, M., Herbert, M. & Hoog, C. Bi-orientation of achiasmatic chromosomes in meiosis I oocytes contributes to aneuploidy in mice. Nature Genet. 39, 966–968 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. LeMaire-Adkins, R. & Hunt, P. A. Nonrandom segregation of the mouse univalent X chromosome: evidence of spindle-mediated meiotic drive. Genetics 156, 775–783 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Nagaoka, S. I., Hodges, C. A., Albertini, D. F. & Hunt, P. A. Oocyte-specific differences in cell-cycle control create an innate susceptibility to meiotic errors. Curr. Biol. 21, 651–657 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Gui, L. & Homer, H. Spindle assembly checkpoint signalling is uncoupled from chromosomal position in mouse oocytes. Development 139, 1941–1946 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Kolano, A., Brunet, S., Silk, A. D., Cleveland, D. W. & Verlhac, M. H. Error prone mammalian female meiosis from silencing the spindle assembly checkpoint without normal interkinetochore tension. Proc. Natl Acad. Sci. USA 2 May 2012 (doi:10.1073/pnas.1204686109).

    Article  CAS  Google Scholar 

  73. Lane, S. I. R., Yun, Y. & Jones, K. T. Timing of anaphase promoting complex activation in mouse oocytes is predicted by microtubule–kinetochore attachment, but not by bivalent alignment or tension. Development 139, 1947–1955 (2012). The studies described in references 70–73 provide evidence that the spindle assembly checkpoint mechanism differs in the oocyte and that metaphase alignment of all chromosomes is not a prerequisite for anaphase onset. This difference provides a mechanism whereby various different factors can all lead to aneuploidy.

    Article  CAS  PubMed  Google Scholar 

  74. Steuerwald, N., Cohen, J., Herrera, R. J., Sandalinas, M. & Brenner, C. A. Association between spindle assembly checkpoint expression and maternal age in human oocytes. Mol. Hum. Reprod. 7, 49–55 (2001).

    Article  CAS  PubMed  Google Scholar 

  75. Brunet, S., Pahlavan, G., Taylor, S. & Maro, B. Functionality of the spindle checkpoint during the first meiotic division of mammalian oocytes. Reproduction 126, 443–450 (2003).

    Article  CAS  PubMed  Google Scholar 

  76. 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).

    Article  CAS  PubMed  Google Scholar 

  77. Homer, H. A., McDougall, A., Levasseur, M., Murdoch, A. P. & Herbert, M. Mad2 is required for inhibiting securin and cyclin B degradation following spindle depolymerisation in meiosis I mouse oocytes. Reproduction 130, 829–843 (2005).

    Article  CAS  PubMed  Google Scholar 

  78. McGuinness, B. E. et al. Regulation of APC/C activity in oocytes by a Bub1-dependent spindle assembly checkpoint. Curr. Biol. 19, 369–380 (2009).

    Article  CAS  PubMed  Google Scholar 

  79. Woods, L. M. et al. Chromosomal influence on meiotic spindle assembly: abnormal meiosis I in female Mlh1 mutant mice. J. Cell Biol. 145, 1395–1406 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Yin, H., Cukurcam, S., Betzendahl, I., Adler, I. D. & Eichenlaub-Ritter, U. Trichlorfon exposure, spindle aberrations and nondisjunction in mammalian oocytes. Chromosoma 107, 514–522 (1998).

    Article  CAS  PubMed  Google Scholar 

  81. Hodges, C. A. et al. Experimental evidence that changes in oocyte growth influence meiotic chromosome segregation. Hum. Reprod. 17, 1171–1180 (2002).

    Article  CAS  PubMed  Google Scholar 

  82. Hunt, P. A. et al. Bisphenol a exposure causes meiotic aneuploidy in the female mouse. Curr. Biol. 13, 546–553 (2003).

    Article  CAS  PubMed  Google Scholar 

  83. Jin, F. et al. Cdc20 is critical for meiosis I and fertility of female mice. PLoS Genet 6, e1001147 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Selesniemi, K., Lee, H. J., Muhlhauser, A. & Tilly, J. L. Prevention of maternal aging-associated oocyte aneuploidy and meiotic spindle defects in mice by dietary and genetic strategies. Proc. Natl Acad. Sci. USA 108, 12319–12324 (2011). This provocative report links caloric restriction with decreased levels of aneuploidy in the ageing female mouse.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Battaglia, D. E., Goodwin, P., Klein, N. A. & Soules, M. R. Influence of maternal age on meiotic spindle assembly in oocytes from naturally cycling women. Hum. Reprod. 11, 2217–2222 (1996).

    Article  CAS  PubMed  Google Scholar 

  86. Volarcik, K. et al. The meiotic competence of in-vitro matured human oocytes is influenced by donor age: evidence that folliculogenesis is compromised in the reproductively aged ovary. Hum. Reprod. 13, 154–160 (1998).

    Article  CAS  PubMed  Google Scholar 

  87. Reis, A. et al. Prometaphase APCcdh1 activity prevents non-disjunction in mammalian oocytes. Nature Cell Biol. 9, 1192–1198 (2007).

    Article  CAS  PubMed  Google Scholar 

  88. Homer, H., Gui, L. & Carroll, J. A spindle assembly checkpoint protein functions in prophase I arrest and prometaphase progression. Science 326, 991–994 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Can, A., Semiz, O. & Cinar, O. Bisphenol-A induces cell cycle delay and alters centrosome and spindle microtubular organization in oocytes during meiosis. Mol. Hum. Reprod. 11, 389–396 (2005).

    Article  CAS  PubMed  Google Scholar 

  90. Pacchierotti, F., Ranaldi, R., Eichenlaub-Ritter, U., Attia, S. & Adler, I. D. Evaluation of aneugenic effects of bisphenol A in somatic and germ cells of the mouse. Mutat. Res. 651, 64–70 (2008).

    Article  CAS  PubMed  Google Scholar 

  91. Eichenlaub-Ritter, U. et al. Exposure of mouse oocytes to bisphenol A causes meiotic arrest but not aneuploidy. Mutat. Res. 651, 82–92 (2008).

    Article  CAS  PubMed  Google Scholar 

  92. Lenie, S., Cortvrindt, R., Eichenlaub-Ritter, U. & Smitz, J. Continuous exposure to bisphenol A during in vitro follicular development induces meiotic abnormalities. Mutat. Res. 651, 71–81 (2008).

    Article  CAS  PubMed  Google Scholar 

  93. Peretz, J., Gupta, R. K., Singh, J., Hernandez-Ochoa, I. & Flaws, J. A. Bisphenol A impairs follicle growth, inhibits steroidogenesis, and downregulates rate-limiting enzymes in the estradiol biosynthesis pathway. Toxicol. Sci. 119, 209–217 (2011).

    Article  CAS  PubMed  Google Scholar 

  94. Bloom, M. S. et al. Bisphenol A exposure reduces the estradiol response to gonadotropin stimulation during in vitro fertilization. Fertil. Steril. 96, 672–677.e2 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Fujimoto, V. Y. et al. Serum unconjugated bisphenol A concentrations in women may adversely influence oocyte quality during in vitro fertilization. Fertil. Steril. 95, 1816–1819 (2011).

    Article  CAS  PubMed  Google Scholar 

  96. Susiarjo, M., Hassold, T. J., Freeman, E. & Hunt, P. A. Bisphenol A exposure in utero disrupts early oogenesis in the mouse. PLoS Genet. 3, e5 (2007).

  97. Allard, P. & Colaiacovo, M. P. Bisphenol A impairs the double-strand break repair machinery in the germline and causes chromosome abnormalities. Proc. Natl Acad. Sci. USA 107, 20405–20410 (2010). These studies provide evidence that exposure to the bisphenol A (BPA) disrupts the prophase events of meiosis in the ovaries of mice and worms, setting the stage for nondisjunctional events during the meiotic divisions.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Brieno-Enriquez, M. A. et al. Human meiotic progression and recombination are affected by bisphenol A exposure during in vitro human oocyte development. Hum. Reprod. 26, 2807–2818 (2011).

    Article  CAS  PubMed  Google Scholar 

  99. Doherty, A. S., Mann, M. R., Tremblay, K. D., Bartolomei, M. S. & Schultz, R. M. Differential effects of culture on imprinted H19 expression in the preimplantation mouse embryo. Biol. Reprod. 62, 1526–1535 (2000).

    Article  CAS  PubMed  Google Scholar 

  100. Khosla, S., Dean, W., Brown, D., Reik, W. & Feil, R. Culture of preimplantation mouse embryos affects fetal development and the expression of imprinted genes. Biol. Reprod. 64, 918–926 (2001).

    Article  CAS  PubMed  Google Scholar 

  101. Mann, M. R. et al. Selective loss of imprinting in the placenta following preimplantation development in culture. Development 131, 3727–3735 (2004).

    Article  CAS  PubMed  Google Scholar 

  102. Rivera, R. M. et al. Manipulations of mouse embryos prior to implantation result in aberrant expression of imprinted genes on day 9.5 of development. Hum. Mol. Genet. 17, 1–14 (2008).

    Article  CAS  PubMed  Google Scholar 

  103. Market-Velker, B. A., Zhang, L., Magri, L. S., Bonvissuto, A. C. & Mann, M. R. Dual effects of superovulation: loss of maternal and paternal imprinted methylation in a dose-dependent manner. Hum. Mol. Genet. 19, 36–51 (2010).

    Article  CAS  PubMed  Google Scholar 

  104. Denomme, M. M., Zhang, L. & Mann, M. R. Embryonic imprinting perturbations do not originate from superovulation-induced defects in DNA methylation acquisition. Fertil. Steril. 96, 734–738.e2 (2011).

    Article  CAS  PubMed  Google Scholar 

  105. Harlap, S. et al. Chromosome abnormalities in oral contraceptive breakthrough pregnancies. Lancet 1, 1342–1343 (1979).

    Article  CAS  PubMed  Google Scholar 

  106. Maudlin, I. & Fraser, L. R. The effect of PMSG dose on the incidence of chromosomal anomalies in mouse embryos fertilized in vitro. J. Reprod. Fertil. 50, 275–280 (1977).

    Article  CAS  PubMed  Google Scholar 

  107. Munne, S. et al. Treatment-related chromosome abnormalities in human embryos. Hum. Reprod. 12, 780–784 (1997).

    Article  CAS  PubMed  Google Scholar 

  108. Baart, E. B. et al. Milder ovarian stimulation for in-vitro fertilization reduces aneuploidy in the human preimplantation embryo: a randomized controlled trial. Hum. Reprod. 22, 980–988 (2007).

    Article  PubMed  Google Scholar 

  109. Rubio, C. et al. Prospective cohort study in high responder oocyte donors using two hormonal stimulation protocols: impact on embryo aneuploidy and development. Hum. Reprod. 25, 2290–2297 (2010).

    Article  CAS  PubMed  Google Scholar 

  110. Penrose, L. in The Early Conceptus, Normal and Abnormal 94–97 (Univ. St Andrews, 1964).

    Google Scholar 

  111. Bond, D. J. & Chandley, A. C. in Aneuploidy 67–75 (Oxford Univ. Press, 1983).

    Google Scholar 

  112. Hubner, K. et al. Derivation of oocytes from mouse embryonic stem cells. Science 300, 1251–1256 (2003).

    Article  PubMed  CAS  Google Scholar 

  113. Toyooka, Y., Tsunekawa, N., Akasu, R. & Noce, T. Embryonic stem cells can form germ cells in vitro. Proc. Natl Acad. Sci. USA 100, 11457–11462 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Geijsen, N. et al. Derivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 427, 148–154 (2004).

    Article  CAS  PubMed  Google Scholar 

  115. Nayernia, K. et al. In vitro-differentiated embryonic stem cells give rise to male gametes that can generate offspring in mice. Dev. Cell 11, 125–132 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. Aflatoonian, B. et al. In vitro post-meiotic germ cell development from human embryonic stem cells. Hum. Reprod. 24, 3150–3159 (2009).

    Article  CAS  PubMed  Google Scholar 

  117. Kee, K., Angeles, V. T., Flores, M., Nguyen, H. N. & Reijo Pera, R. A. Human DAZL, DAZ and BOULE genes modulate primordial germ-cell and haploid gamete formation. Nature 462, 222–225 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Nicholas, C. R., Haston, K. M., Grewall, A. K., Longacre, T. A. & Reijo Pera, R. A. Transplantation directs oocyte maturation from embryonic stem cells and provides a therapeutic strategy for female infertility. Hum. Mol. Genet. 18, 4376–4389 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. White, Y. A. et al. Oocyte formation by mitotically active germ cells purified from ovaries of reproductive-age women. Nature Med. 18, 413–421 (2012).

    Article  CAS  PubMed  Google Scholar 

  120. The Practice Committee of the Society for Assisted Reproductive Technology, Practice Committee of the American Society for Reproductive Medicine. Preimplantation genetic testing: a Practice Committee opinion. Fertil. Steril. 90, S136–S143 (2008).

  121. Committee on Genetics. ACOG Committee Opinion No. 430: preimplantation genetic screening for aneuploidy. Obstet. Gynecol. 113, 766–767 (2009).

  122. Munne, S., Wells, D. & Cohen, J. Technology requirements for preimplantation genetic diagnosis to improve assisted reproduction outcomes. Fertil. Steril. 94, 408–430 (2010).

    Article  PubMed  Google Scholar 

  123. Handyside, A. H. PGD and aneuploidy screening for 24 chromosomes by genome-wide SNP analysis: seeing the wood and the trees. Reprod. Biomed. Online 23, 686–691 (2011).

    Article  CAS  PubMed  Google Scholar 

  124. Lamb, N. E. et al. Characterization of susceptible chiasma configurations that increase the risk for maternal nondisjunction of chromosome 21. Hum. Mol. Genet. 6, 1391–1399 (1997).

    Article  CAS  PubMed  Google Scholar 

  125. Hassold, T. et al. Human aneuploidy: incidence, origin, and etiology. Environ. Mol. Mutagen. 28, 167–175 (1996).

    Article  CAS  PubMed  Google Scholar 

  126. Zenzes, M. T. & Casper, R. F. Cytogenetics of human oocytes, zygotes, and embryos after in vitro fertilization. Hum. Genet. 88, 367–375 (1992).

    Article  CAS  PubMed  Google Scholar 

  127. Magli, M. C., Gianaroli, L. & Ferraretti, A. P. Chromosomal abnormalities in embryos. Mol. Cell Endocrinol. 183, S29–S34 (2001).

    Article  CAS  PubMed  Google Scholar 

  128. Staessen, C. et al. Comparison of blastocyst transfer with or without preimplantation genetic diagnosis for aneuploidy screening in couples with advanced maternal age: a prospective randomized controlled trial. Hum. Reprod. 19, 2849–2858 (2004).

    Article  PubMed  Google Scholar 

  129. Munne, S. et al. Maternal age, morphology, development and chromosome abnormalities in over 6000 cleavage-stage embryos. Reprod. Biomed. Online 14, 628–634 (2007).

    Article  CAS  PubMed  Google Scholar 

  130. Ercelen, N. et al. Successful preimplantation genetic aneuploidy screening in Turkish patients. Genet. Mol. Res. 10, 4093–4103 (2011).

    Article  CAS  PubMed  Google Scholar 

  131. Obradors, A. et al. Whole-chromosome aneuploidy analysis in human oocytes: focus on comparative genomic hybridization. Cytogenet. Genome Res. 133, 119–126 (2011).

    Article  CAS  PubMed  Google Scholar 

  132. Martin, R. H. & Rademaker, A. The frequency of aneuploidy among individual chromosomes in 6,821 human sperm chromosome complements. Cytogenet. Cell Genet. 53, 103–107 (1990).

    Article  CAS  PubMed  Google Scholar 

  133. Martin, R. H., Ko, E. & Rademaker, A. Distribution of aneuploidy in human gametes: comparison between human sperm and oocytes. Am. J. Med. Genet. 39, 321–331 (1991).

    Article  CAS  PubMed  Google Scholar 

  134. Templado, C., Vidal, F. & Estop, A. Aneuploidy in human spermatozoa. Cytogenet. Genome Res. 133, 91–99 (2011).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Research conducted in the Hunt and Hassold laboratories and discussed in this Review was supported by US National Institutes of Health grants HD21341 (to T.J.H.) and ES013527 (to P.A.H.). In addition, the authors would gratefully like to acknowledge the three 'grand dames' of human aneuploidy research who sparked our interest and shaped our thinking: D. Warburton, M. Mikkelsen and, most of all, P. Jacobs.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Terry J. Hassold or Patricia A. Hunt.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Glossary

Aneuploidy

A chromosome abnormality in which the chromosome number is not a multiple of the haploid number.

Assisted reproductive technology

(ART). Clinical approaches that are used to help infertile couples achieve a normal pregnancy. These include ovarian stimulation protocols using exogenous hormones, in vitro fertilization, intracytoplasmic sperm injection and preimplantation genetic diagnosis.

Nondisjunction

The failure of chromosomes to segregate normally during cell division. Nondisjunction at meiosis I results in products with additional or missing whole chromosomes; nondisjunction at meiosis II results in products with additional or missing sister chromatids.

Pachytene

The stage of meiotic prophase characterized by complete synapsis of all homologues. Importantly, crossover sites can be visualized in pachytene stage cells using appropriate markers.

Bivalent

Paired homologous chromosomes that are tethered by a crossover (or crossovers).

Synapsis

The intimate pairing of homologous chromosomes that occurs during prophase of meiosis and is essential for meiotic recombination. Synapsis is facilitated by the formation of a meiosis-specific protein scaffold called the synaptonemal complex.

Pseudoautosomal region

(PAR). The small region of homology at the distal ends of the X and Y chromosomes that allows for synapsis and recombination.

Sister chromatid cohesion

Replicated chromosomes, or sister chromatids, are held together by cohesin, which is a protein complex that is loaded onto the chromosomes during S phase. In meiosis, sister chromatid cohesion is sequentially released from the chromosome arms at anaphase I and from sister centromeres at anaphase II, allowing for the orderly segregation of homologues and sister chromatids, respectively.

Univalents

Homologous chromosomes that are not associated with one another (for example, owing to failure to recombine).

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nagaoka, S., Hassold, T. & Hunt, P. Human aneuploidy: mechanisms and new insights into an age-old problem. Nat Rev Genet 13, 493–504 (2012). https://doi.org/10.1038/nrg3245

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3245

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing