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.

  • Review Article
  • Published:

Aneuploidy as a promoter and suppressor of malignant growth

Abstract

Aneuploidy has been recognized as a hallmark of tumorigenesis for more than 100 years, but the connection between chromosomal errors and malignant growth has remained obscure. New evidence emerging from both basic and clinical research has illuminated a complicated relationship: despite its frequency in human tumours, aneuploidy is not a universal driver of cancer development and instead can exert substantial tumour-suppressive effects. The specific consequences of aneuploidy are highly context dependent and are influenced by a cell’s genetic and environmental milieu. In this Review, we discuss the diverse facets of cancer biology that are shaped by aneuploidy, including metastasis, drug resistance and immune recognition, and we highlight aneuploidy’s distinct roles as both a tumour promoter and an anticancer vulnerability.

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

Fig. 1: Generating aneuploidy in cell lines and in mouse models.
Fig. 2: Aneuploidy and tumorigenesis.
Fig. 3: Interrelationship between aneuploidy-associated and CIN-associated phenotypes.
Fig. 4: Aneuploidy can have protumorigenic and tumour-inhibiting effects on the immune system.

Similar content being viewed by others

References

  1. Biancotti, J. C. et al. The in vitro survival of human monosomies and trisomies as embryonic stem cells. Stem Cell Res. 9, 218–224 (2012).

    CAS  PubMed  Google Scholar 

  2. van den Berg, M. M. J., van Maarle, M. C., van Wely, M. & Goddijn, M. Genetics of early miscarriage. Biochim. Biophys. Acta 1822, 1951–1959 (2012).

    PubMed  Google Scholar 

  3. Weaver, B. A. & Cleveland, D. W. Does aneuploidy cause cancer? Curr. Opin. Cell Biol. 18, 658–667 (2006).

    CAS  PubMed  Google Scholar 

  4. Donne, R., Saroul-Aïnama, M., Cordier, P., Celton-Morizur, S. & Desdouets, C. Polyploidy in liver development, homeostasis and disease. Nat. Rev. Gastroenterol. Hepatol. 17, 391–405 (2020).

    CAS  PubMed  Google Scholar 

  5. Lens, S. M. A. & Medema, R. H. Cytokinesis defects and cancer. Nat. Rev. Cancer 19, 32–45 (2019).

    CAS  PubMed  Google Scholar 

  6. Tanaka, K. et al. Tetraploidy in cancer and its possible link to aging. Cancer Sci. 109, 2632–2640 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. de Smith, A. J., Trewick, A. L. & Blakemore, A. I. F. Implications of copy number variation in people with chromosomal abnormalities: potential for greater variation in copy number state may contribute to variability of phenotype. HUGO J. 4, 1–9 (2010).

    PubMed  PubMed Central  Google Scholar 

  8. Tang, Y.-C. & Amon, A. Gene copy-number alterations: a cost-benefit analysis. Cell 152, 394–405 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Hardy, P. A. & Zacharias, H. Reappraisal of the Hansemann-Boveri hypothesis on the origin of tumors. Cell Biol. Int. 29, 983–992 (2005).

    PubMed  Google Scholar 

  10. Boveri, T. Concerning the origin of malignant tumours by Theodor Boveri. Translated and annotated by Henry Harris. J. Cell Sci. 121 (Suppl. 1), 1–84 (2008).

    PubMed  Google Scholar 

  11. Taylor, A. M. et al. Genomic and functional approaches to understanding cancer aneuploidy. Cancer Cell 33, 676–689.e3 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Watkins, T. B. K. et al. Pervasive chromosomal instability and karyotype order in tumour evolution. Nature 587, 126–132 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Ben-David, U. & Amon, A. Context is everything: aneuploidy in cancer. Nat. Rev. Genet. 21, 44–62 (2020).

    CAS  PubMed  Google Scholar 

  14. Crowell, R. E. et al. Detection of trisomy 7 in nonmalignant bronchial epithelium from lung cancer patients and individuals at risk for lung cancer. Cancer Epidemiol. Biomarkers Prev. 5, 631–637 (1996).

    CAS  PubMed  Google Scholar 

  15. Aldaz, C. M., Conti, C. J., Klein-Szanto, A. J. & Slaga, T. J. Progressive dysplasia and aneuploidy are hallmarks of mouse skin papillomas: relevance to malignancy. Proc. Natl Acad. Sci. USA 84, 2029–2032 (1987).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Aldaz, C. M., Trono, D., Larcher, F., Slaga, T. J. & Conti, C. J. Sequential trisomization of chromosomes 6 and 7 in mouse skin premalignant lesions. Mol. Carcinog. 2, 22–26 (1989).

    CAS  PubMed  Google Scholar 

  17. Longy, M. et al. Chromosomal analysis of colonic adenomatous polyps. Cancer Genet. Cytogenet. 49, 249–257 (1990).

    CAS  PubMed  Google Scholar 

  18. Garewal, H. S., Sampliner, R., Liu, Y. & Trent, J. M. Chromosomal rearrangements in Barrett’s esophagus. A premalignant lesion of esophageal adenocarcinoma. Cancer Genet. Cytogenet. 42, 281–286 (1989).

    CAS  PubMed  Google Scholar 

  19. Mian, C. et al. Fluorescence in situ hybridization in cervical smears: detection of numerical aberrations of chromosomes 7, 3, and X and relationship to HPV infection. Gynecol. Oncol. 75, 41–46 (1999).

    CAS  PubMed  Google Scholar 

  20. Gerstung, M. et al. The evolutionary history of 2,658 cancers. Nature 578, 122–128 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Upender, M. B. et al. Chromosome transfer induced aneuploidy results in complex dysregulation of the cellular transcriptome in immortalized and cancer cells. Cancer Res. 64, 6941–6949 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Ben-David, U. et al. Aneuploidy induces profound changes in gene expression, proliferation and tumorigenicity of human pluripotent stem cells. Nat. Commun. 5, 4825 (2014). This article reveals the effects of trisomy 12, which is the most common genomic alteration in human pluripotent stem cells. Trisomy 12 causes transcriptional changes similar to germ cell tumours and increases proliferation and tumorigenicity.

    CAS  PubMed  Google Scholar 

  23. FitzPatrick, D. R. Transcriptome analysis of human autosomal trisomy. Hum. Mol. Genet. 11, 3249–3256 (2002).

    CAS  PubMed  Google Scholar 

  24. Stingele, S. et al. Global analysis of genome, transcriptome and proteome reveals the response to aneuploidy in human cells. Mol. Syst. Biol. 8, 608 (2012). This study performs transcriptome and proteome analysis to understand how aneuploidy alters cellular physiology. Although transcription levels of genes on extra chromosomes scale with chromosome copy number, the expression of certain proteins shows dosage compensation.

    PubMed  PubMed Central  Google Scholar 

  25. Pavelka, N. et al. Aneuploidy confers quantitative proteome changes and phenotypic variation in budding yeast. Nature 468, 321–325 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Braun, R. et al. Single chromosome aneuploidy induces genome-wide perturbation of nuclear organization and gene expression. Neoplasia 21, 401–412 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Torres, E. M., Williams, B. R. & Amon, A. Aneuploidy: cells losing their balance. Genetics 179, 737–746 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Sheltzer, J. M., Torres, E. M., Dunham, M. J. & Amon, A. Transcriptional consequences of aneuploidy. Proc. Natl Acad. Sci. USA 109, 12644–12649 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Schukken, K. M. & Foijer, F. CIN and aneuploidy: different concepts, different consequences. Bioessays 40, 1700147 (2018).

    Google Scholar 

  30. Zhu, J., Tsai, H.-J., Gordon, M. R. & Li, R. Cellular stress associated with aneuploidy. Dev. Cell 44, 420–431 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Bakhoum, S. F. & Cantley, L. C. The multifaceted role of chromosomal instability in cancer and its microenvironment. Cell 174, 1347–1360 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Sheltzer, J. M. et al. Single-chromosome gains commonly function as tumor suppressors. Cancer Cell 31, 240–255 (2017). This study reveals that many single-chromosome trisomies suppress cellular transformation. At the same time, aneuploidy may provide evolutionary flexibility, promoting the aggressive growth of tumours with complex karyotypes.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Passerini, V. et al. The presence of extra chromosomes leads to genomic instability. Nat. Commun. 7, 10754 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Vasudevan, A. et al. Single-chromosomal gains can function as metastasis suppressors and promoters in colon cancer. Dev. Cell 52, 413–428.e6 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhang, M. et al. Aneuploid embryonic stem cells exhibit impaired differentiation and increased neoplastic potential. EMBO J. 35, 2285–2300 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Epstein, C. J. Mouse monosomies and trisomies as experimental systems for studying mammalian aneuploidy. Trends Genet. 1, 129–134 (1985).

    Google Scholar 

  37. Hernandez, D. & Fisher, E. M. Mouse autosomal trisomy: two’s company, three’s a crowd. Trends Genet. 15, 241–247 (1999).

    CAS  PubMed  Google Scholar 

  38. Williams, B. R. et al. Aneuploidy affects proliferation and spontaneous immortalization in mammalian cells. Science 322, 703–709 (2008). This study characterizes the effects of trisomies in mouse embryonic fibroblasts. These aneuploidies increase the expression of genes on the aneuploid chromosome while also impairing proliferation and causing certain metabolic alterations.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Pfau, S. J., Silberman, R. E., Knouse, K. A. & Amon, A. Aneuploidy impairs hematopoietic stem cell fitness and is selected against in regenerating tissues in vivo. Genes Dev. 30, 1395–1408 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Bolton, H. et al. Mouse model of chromosome mosaicism reveals lineage-specific depletion of aneuploid cells and normal developmental potential. Nat. Commun. 7, 11165 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Singla, S., Iwamoto-Stohl, L. K., Zhu, M. & Zernicka-Goetz, M. Autophagy-mediated apoptosis eliminates aneuploid cells in a mouse model of chromosome mosaicism. Nat. Commun. 11, 2958 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Herbst, E. W., Pluznik, D. H., Gropp, A. & Uthgennant, H. Trisomic hemopoietic stem cells of fetal origin restore hemopoiesis in lethally irradiated mice. Science 211, 1175–1177 (1981).

    CAS  PubMed  Google Scholar 

  43. Davisson, M. et al. Segmental trisomy as a mouse model for down syndrome. Prog. Clin. Biol. Res. 384, 117–133 (1993).

    CAS  PubMed  Google Scholar 

  44. Kazuki, Y. et al. A non-mosaic transchromosomic mouse model of down syndrome carrying the long arm of human chromosome 21. eLife 9, e56223 (2020).

    PubMed  PubMed Central  Google Scholar 

  45. Alford, K. A. et al. Perturbed hematopoiesis in the Tc1 mouse model of down syndrome. Blood 115, 2928–2937 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Dobles, M., Liberal, V., Scott, M. L., Benezra, R. & Sorger, P. K. Chromosome missegregation and apoptosis in mice lacking the mitotic checkpoint protein Mad2. Cell 101, 635–645 (2000).

    CAS  PubMed  Google Scholar 

  47. Hudson, D. F. et al. Centromere protein b null mice are mitotically and meiotically normal but have lower body and testis weights. J. Cell Biol. 141, 309–319 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Howman, E. V. et al. Early disruption of centromeric chromatin organization in centromere protein A (Cenpa) null mice. Proc. Natl Acad. Sci. USA 97, 1148–1153 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Diaz-Rodríguez, E., Sotillo, R., Schvartzman, J.-M. & Benezra, R. Hec1 overexpression hyperactivates the mitotic checkpoint and induces tumor formation in vivo. Proc. Natl Acad. Sci. USA 105, 16719–16724 (2008).

    PubMed  PubMed Central  Google Scholar 

  50. Kabeche, L. & Compton, D. A. Checkpoint-independent stabilization of kinetochore-microtubule attachments by Mad2 in human cells. Curr. Biol. 22, 638–644 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhang, D. et al. Cre-loxP-controlled periodic Aurora-A overexpression induces mitotic abnormalities and hyperplasia in mammary glands of mouse models. Oncogene 23, 8720–8730 (2004).

    CAS  PubMed  Google Scholar 

  52. Marthiens, V. et al. Centrosome amplification causes microcephaly. Nat. Cell Biol. 15, 731–740 (2013).

    CAS  PubMed  Google Scholar 

  53. Mukherjee, M. et al. separase loss of function cooperates with the loss of p53 in the initiation and progression of T- and B-cell lymphoma, leukemia and aneuploidy in mice. PLoS ONE 6, (2011).

  54. Remeseiro, S. et al. Cohesin-SA1 deficiency drives aneuploidy and tumourigenesis in mice due to impaired replication of telomeres. EMBO J. 31, 2076–2089 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Torres, E. M. et al. Effects of aneuploidy on cellular physiology and cell division in haploid yeast. Science 317, 916–924 (2007).

    CAS  PubMed  Google Scholar 

  56. Sheltzer, J. M. A transcriptional and metabolic signature of primary aneuploidy is present in chromosomally unstable cancer cells and informs clinical prognosis. Cancer Res. 73, 6401–6412 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Hasaart, K. A. L. et al. Mutation accumulation and developmental lineages in normal and Down syndrome human fetal haematopoiesis. Sci. Rep. 10, 12991 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Chunduri, N. K. & Storchová, Z. The diverse consequences of aneuploidy. Nat. Cell Biol. 21, 54–62 (2019).

    CAS  PubMed  Google Scholar 

  59. Andriani, G. A. et al. Whole chromosome instability induces senescence and promotes SASP. Sci. Rep. 6, 1–17 (2016). This study reveals that CIN induces DNA damage and oxidative stress that leads to premature cellular senescence in human fibroblasts. The senescent cells resulting from CIN secrete high levels of cytokines, chemokines and growth factors, characteristic of the SASP.

    Google Scholar 

  60. Torres, E. M. et al. Identification of aneuploidy-tolerating mutations. Cell 143, 71–83 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Thompson, S. L. & Compton, D. A. Proliferation of aneuploid human cells is limited by a p53-dependent mechanism. J. Cell Biol. 188, 369–381 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Soto, M. et al. p53 prohibits propagation of chromosome segregation errors that produce structural aneuploidies. Cell Rep. 19, 2423–2431 (2017).

    CAS  PubMed  Google Scholar 

  63. Giam, M. et al. p53 induces senescence in the unstable progeny of aneuploid cells. Cell Cycle https://doi.org/10.1080/15384101.2020.1850968 (2019).

    Article  Google Scholar 

  64. Li, M. et al. The ATM-p53 pathway suppresses aneuploidy-induced tumorigenesis. Proc. Natl Acad. Sci. USA 107, 14188–14193 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Simões-Sousa, S. et al. The p38α stress kinase suppresses aneuploidy tolerance by inhibiting Hif-1α. Cell Rep. 25, 749–760.e6 (2018).

    PubMed  PubMed Central  Google Scholar 

  66. López-García, C. et al. BCL9L dysfunction impairs caspase-2 expression permitting aneuploidy tolerance in colorectal cancer. Cancer Cell 31, 79–93 (2017).

    PubMed  PubMed Central  Google Scholar 

  67. Rutledge, S. D. et al. Selective advantage of trisomic human cells cultured in non-standard conditions. Sci. Rep. 6, 22828 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Lukow, D. A. et al. Chromosomal instability accelerates the evolution of resistance to anti-cancer therapies. Preprint at bioRxiv https://doi.org/10.1101/2020.09.25.314229 (2020).

    Article  Google Scholar 

  69. Ippolito, M. R. et al. Aneuploidy-driven genome instability triggers resistance to chemotherapy. Preprint at bioRxiv https://doi.org/10.1101/2020.09.25.313924 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Shukla, A. et al. Chromosome arm aneuploidies shape tumour evolution and drug response. Nat. Commun. 11, 449 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Selmecki, A. M., Dulmage, K., Cowen, L. E., Anderson, J. B. & Berman, J. Acquisition of aneuploidy provides increased fitness during the evolution of antifungal drug resistance. PLoS Genet. 5, e1000705 (2009).

    PubMed  PubMed Central  Google Scholar 

  72. Selmecki, A., Gerami-Nejad, M., Paulson, C., Forche, A. & Berman, J. An isochromosome confers drug resistance in vivo by amplification of two genes, ERG11 and TAC1. Mol. Microbiol. 68, 624–641 (2008).

    CAS  PubMed  Google Scholar 

  73. Salgueiro, L. et al. Acquisition of chromosome instability is a mechanism to evade oncogene addiction. EMBO Mol. Med. 12, e10941 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Liu, G. et al. Gene essentiality is a quantitative property linked to cellular evolvability. Cell 163, 1388–1399 (2015).

    CAS  PubMed  Google Scholar 

  75. Draper, J. S. et al. Recurrent gain of chromosomes 17q and 12 in cultured human embryonic stem cells. Nat. Biotechnol. 22, 53–54 (2004).

    CAS  PubMed  Google Scholar 

  76. Baker, D. E. C. et al. Adaptation to culture of human embryonic stem cells and oncogenesis in vivo. Nat. Biotechnol. 25, 207–215 (2007).

    CAS  PubMed  Google Scholar 

  77. Olariu, V. et al. Modeling the evolution of culture-adapted human embryonic stem cells. Stem Cell Res. 4, 50–56 (2010).

    PubMed  Google Scholar 

  78. Smith, J. C. & Sheltzer, J. M. Systematic identification of mutations and copy number alterations associated with cancer patient prognosis. eLife 7, e39217 (2018).

    PubMed  PubMed Central  Google Scholar 

  79. Bakhoum, S. F. et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 553, 467–472 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Barlogie, B. et al. Flow cytometry in clinical cancer research. Cancer Res. 43, 3982–3997 (1983).

    CAS  PubMed  Google Scholar 

  81. Stopsack, K. H. et al. Aneuploidy drives lethal progression in prostate cancer. Proc. Natl Acad. Sci. USA 116, 11390–11395 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Kheir, S. M. et al. Prognostic significance of DNA aneuploidy in stage I cutaneous melanoma. Ann. Surg. 207, 455–461 (1988).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Xu, J., Huang, L. & Li, J. DNA aneuploidy and breast cancer: a meta-analysis of 141,163 cases. Oncotarget 7, 60218–60229 (2016).

    PubMed  PubMed Central  Google Scholar 

  84. Gao, C. et al. Chromosome instability drives phenotypic switching to metastasis. Proc. Natl Acad. Sci. USA 113, 14793–14798 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Sotillo, R. et al. Mad2 overexpression promotes aneuploidy and tumorigenesis in mice. Cancer Cell 11, 9–23 (2007).

    CAS  PubMed  Google Scholar 

  86. Kalitsis, P. et al. Increased chromosome instability but not cancer predisposition in haploinsufficient Bub3 mice. Genes Chromosomes Cancer 44, 29–36 (2005).

    CAS  PubMed  Google Scholar 

  87. Jeganathan, K., Malureanu, L., Baker, D. J., Abraham, S. C. & van Deursen, J. M. Bub1 mediates cell death in response to chromosome missegregation and acts to suppress spontaneous tumorigenesis. J. Cell Biol. 179, 255–267 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Baker, D. J., Jin, F. & van Deursen, J. M. The yin and yang of the Cdkn2a locus in senescence and aging. Cell Cycle 7, 2795–2802 (2008).

    CAS  PubMed  Google Scholar 

  89. Baker, D. J., Jin, F., Jeganathan, K. B. & van Deursen, J. M. Whole chromosome instability caused by Bub1 insufficiency drives tumorigenesis through tumor suppressor gene loss of heterozygosity. Cancer Cell 16, 475–486 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Baker, D. J., Weaver, R. L. & van Deursen, J. M. p21 both attenuates and drives senescence and aging in BubR1 progeroid mice. Cell Rep. 3, 1164–1174 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Weaver, B. A. A., Silk, A. D., Montagna, C., Verdier-Pinard, P. & Cleveland, D. W. Aneuploidy acts both oncogenically and as a tumor suppressor. Cancer Cell 11, 25–36 (2007). This article establishes that CIN has context-dependent effects on tumorigenesis depending on the methods used to induce tumour formation. While CIN increases the rates of spontaneous lymphomas and lung tumours in aged animals, it inhibits tumorigenesis resulting from certain chemical and genetic perturbations.

    CAS  PubMed  Google Scholar 

  92. Zasadil, L. M. et al. High rates of chromosome missegregation suppress tumor progression but do not inhibit tumor initiation. Mol. Biol. Cell 27, 1981–1989 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Silk, A. D. et al. Chromosome missegregation rate predicts whether aneuploidy will promote or suppress tumors. Proc. Natl Acad. Sci. USA 110, E4134–E4141 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. García-Higuera, I. et al. Genomic stability and tumour suppression by the APC/C cofactor Cdh1. Nat. Cell Biol. 10, 802–811 (2008).

    PubMed  Google Scholar 

  95. Funk, L. C., Zasadil, L. M. & Weaver, B. A. Living in CIN: mitotic infidelity and its consequences for tumor promotion and suppression. Dev. Cell 39, 638–652 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Hoevenaar, W. H. M. et al. Degree and site of chromosomal instability define its oncogenic potential. Nat. Commun. 11, 1501 (2020). This study demonstrates that differing levels of CIN can have opposing effects in different tissues. Moderate CIN increases tumour formation in the intestine and distal part of the colon of a mouse model, but higher CIN increases tumour development in the distal part of the colon without any effect on the small intestine.

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Schvartzman, J.-M., Sotillo, R. & Benezra, R. Mitotic chromosomal instability and cancer: mouse modelling of the human disease. Nat. Rev. Cancer 10, 102–115 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Wozniak, R., Burke, B. & Doye, V. Nuclear transport and the mitotic apparatus: an evolving relationship. Cell. Mol. Life Sci. 67, 2215–2230 (2010).

    CAS  PubMed  Google Scholar 

  99. Balbás-Martínez, C. et al. Recurrent inactivation of STAG2 in bladder cancer is not associated with aneuploidy. Nat. Genet. 45, 1464–1469 (2013).

    PubMed  PubMed Central  Google Scholar 

  100. Solomon, D. A., Kim, J.-S. & Waldman, T. Cohesin gene mutations in tumorigenesis: from discovery to clinical significance. BMB Rep. 47, 299–310 (2014).

    PubMed  PubMed Central  Google Scholar 

  101. Cipressa, F. et al. A role for separase in telomere protection. Nat. Commun. 7, 10405 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Choi, E., Zhang, X., Xing, C. & Yu, H. Mitotic checkpoint regulators control insulin signaling and metabolic homeostasis. Cell 166, 567–581 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Tighe, A., Johnson, V. L., Albertella, M. & Taylor, S. S. Aneuploid colon cancer cells have a robust spindle checkpoint. EMBO Rep. 2, 609–614 (2001). This article demonstrates that colon cancer cell lines that display CIN nonetheless have a functional spindle-assembly checkpoint and undergo mitotic arrest on spindle damage.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Gascoigne, K. E. & Taylor, S. S. Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs. Cancer Cell 14, 111–122 (2008).

    CAS  PubMed  Google Scholar 

  105. Thompson, S. L., Bakhoum, S. F. & Compton, D. A. Mechanisms of chromosomal instability. Curr. Biol. 20, R285–R295 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Hasle, H., Clemmensen, I. H. & Mikkelsen, M. Risks of leukaemia and solid tumours in individuals with Down’s syndrome. Lancet 355, 165–169 (2000).

    CAS  PubMed  Google Scholar 

  107. Nižetić, D. & Groet, J. Tumorigenesis in Down’s syndrome: big lessons from a small chromosome. Nat. Rev. Cancer 12, 721–732 (2012).

    PubMed  Google Scholar 

  108. Hasle, H., Friedman, J. M., Olsen, J. H. & Rasmussen, S. A. Low risk of solid tumors in persons with Down syndrome. Genet. Med. 18, 1151–1157 (2016).

    PubMed  Google Scholar 

  109. Patja, K., Pukkala, E., Sund, R., Iivanainen, M. & Kaski, M. Cancer incidence of persons with down syndrome in Finland: a population-based study. Int. J. Cancer 118, 1769–1772 (2006).

    CAS  PubMed  Google Scholar 

  110. Nixon, D. W. Down syndrome, obesity, Alzheimer’s disease, and cancer: a brief review and hypothesis. Brain Sci. 8, 53 (2018).

    PubMed Central  Google Scholar 

  111. Mendonca, G. V., Pereira, F. D. & Fernhall, B. Reduced exercise capacity in persons with Down syndrome: cause, effect, and management. Ther. Clin. Risk Manag. 6, 601–610 (2010).

    PubMed  PubMed Central  Google Scholar 

  112. Patja, K., Eero, P. & Iivanainen, M. Cancer incidence among people with intellectual disability. J. Intellect. Disabil. Res. 45, 300–307 (2001).

    CAS  PubMed  Google Scholar 

  113. Demas, G. E., Nelson, R. J., Krueger, B. K. & Yarowsky, P. J. Spatial memory deficits in segmental trisomic Ts65Dn mice. Behav. Brain Res. 82, 85–92 (1996).

    CAS  PubMed  Google Scholar 

  114. Baxter, L. L., Moran, T. H., Richtsmeier, J. T., Troncoso, J. & Reeves, R. H. Discovery and genetic localization of Down syndrome cerebellar phenotypes using the Ts65Dn mouse. Hum. Mol. Genet. 9, 195–202 (2000).

    CAS  PubMed  Google Scholar 

  115. Richtsmeier, J. T., Baxter, L. L. & Reeves, R. H. Parallels of craniofacial maldevelopment in Down syndrome and Ts65Dn mice. Dev. Dyn. 217, 137–145 (2000).

    CAS  PubMed  Google Scholar 

  116. Belichenko, P. V. et al. Synaptic structural abnormalities in the Ts65Dn mouse model of Down Syndrome. J. Comp. Neurol. 480, 281–298 (2004).

    PubMed  Google Scholar 

  117. Kleschevnikov, A. M. et al. Hippocampal long-term potentiation suppressed by increased inhibition in the Ts65Dn mouse, a genetic model of Down syndrome. J. Neurosci. 24, 8153–8160 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Moore, C. S. Postnatal lethality and cardiac anomalies in the Ts65Dn Down syndrome mouse model. Mamm. Genome 17, 1005–1012 (2006).

    CAS  PubMed  Google Scholar 

  119. Sussan, T. E., Yang, A., Li, F., Ostrowski, M. C. & Reeves, R. H. Trisomy represses ApcMin-mediated tumours in mouse models of Down’s syndrome. Nature 451, 73–75 (2008).

    CAS  PubMed  Google Scholar 

  120. Baek, K.-H. et al. Down’s syndrome suppression of tumour growth and the role of the calcineurin inhibitor DSCR1. Nature 459, 1126–1130 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Yang, A. & Reeves, R. H. Increased survival following tumorigenesis in Ts65Dn mice that model Down syndrome. Cancer Res. 71, 3573–3581 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Kirsammer, G. et al. Highly penetrant myeloproliferative disease in the Ts65Dn mouse model of Down syndrome. Blood 111, 767–775 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. O’Doherty, A. et al. An aneuploid mouse strain carrying human chromosome 21 with Down syndrome phenotypes. Science 309, 2033–2037 (2005).

    PubMed  PubMed Central  Google Scholar 

  124. Reynolds, L. E. et al. Tumour angiogenesis is reduced in the Tc1 mouse model of Down syndrome. Nature 465, 813–817 (2010). This article reveals that transplanted tumour cells grow poorly when injected into Tc1 mice due to decreased angiogenesis. Depletion of putative antiangiogenic genes such as Adamts1 and Erg from the extra copy of chromosome 21 is sufficient to restore a normal angiogenic response.

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Nusinow, D. P. et al. Quantitative proteomics of the Cancer Cell Line Encyclopedia. Cell 180, 387–402.e16 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Dephoure, N. et al. Quantitative proteomic analysis reveals posttranslational responses to aneuploidy in yeast. eLife 3, e03023 (2014).

    PubMed  PubMed Central  Google Scholar 

  127. McShane, E. et al. Kinetic analysis of protein stability reveals age-dependent degradation. Cell 167, 803–815.e21 (2016).

    CAS  PubMed  Google Scholar 

  128. Gry, M. et al. Correlations between RNA and protein expression profiles in 23 human cell lines. BMC Genomics 10, 365 (2009).

    PubMed  PubMed Central  Google Scholar 

  129. Roizen, N. J. & Patterson, D. Down’s syndrome. Lancet 361, 1281–1289 (2003).

    PubMed  Google Scholar 

  130. Wu, J., Springett, A. & Morris, J. K. Survival of trisomy 18 (Edwards syndrome) and trisomy 13 (Patau syndrome) in England and Wales: 2004-2011. Am. J. Med. Genet. A 161A, 2512–2518 (2013).

    PubMed  Google Scholar 

  131. Sack, L. M. et al. Profound tissue specificity in proliferation control underlies cancer drivers and aneuploidy patterns. Cell 173, 499–514.e23 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Davoli, T. et al. Cumulative haploinsufficiency and triplosensitivity drive aneuploidy patterns and shape the cancer genome. Cell 155, 948–962 (2013). This study develops novel computational models to analyse mutational signatures during human tumorigenesis to classify genes as oncogenes or tumour suppressors. The distribution of these genes on chromosomes correlates with the complex patterns of copy number alterations found in cancer genomes.

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Auslander, N. et al. Cancer-type specific aneuploidies hard-wire chromosome-wide gene expression patterns of their tissue of origin. Preprint at bioRxiv https://doi.org/10.1101/563858 (2019).

    Article  Google Scholar 

  134. Sheltzer, J. M. et al. Aneuploidy drives genomic instability in yeast. Science 333, 1026–1030 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Jones, L. et al. Gain of MYC underlies recurrent trisomy of the MYC chromosome in acute promyelocytic leukemia. J. Exp. Med. 207, 2581–2594 (2010). This article demonstrates that the gain of mouse chromosome 15, harbouring the Myc oncogene, occurs spontaneously during the development of acute myeloid leukaemia. In a mouse model, the selection of trisomy 15 is due to the fitness advantages provided by MYC overexpression, and when MYC is overexpressed ectopically, selection for this trisomy is lost.

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Tursky, M. L. et al. Overexpression of ERG in cord blood progenitors promotes expansion and recapitulates molecular signatures of high ERG leukemias. Leukemia 29, 819–827 (2015).

    CAS  PubMed  Google Scholar 

  137. Carmichael, C. L. et al. Hematopoietic overexpression of the transcription factor Erg induces lymphoid and erythro-megakaryocytic leukemia. Proc. Natl Acad. Sci. USA 109, 15437–15442 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Stankiewicz, M. J. & Crispino, J. D. ETS2 and ERG promote megakaryopoiesis and synergize with alterations in GATA-1 to immortalize hematopoietic progenitor cells. Blood 113, 3337–3347 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Ng, A. P. et al. Trisomy of Erg is required for myeloproliferation in a mouse model of Down syndrome. Blood 115, 3966–3969 (2010). This article identifies a gene on chromosome 21 that can contribute to the haematopoietic disorders found in individuals with Down syndrome. The authors demonstrate that losing a single copy of the Erg gene reverses the pathological and haematological features of myeloproliferation such as megakaryocytosis and progenitor cell expansion in Ts65Dn mice.

    CAS  PubMed  Google Scholar 

  140. Shin, J., Lee, J. C. & Baek, K.-H. A single extra copy of Dscr1 improves survival of mice developing spontaneous lung tumors through suppression of tumor angiogenesis. Cancer Lett. 342, 70–81 (2014).

    CAS  PubMed  Google Scholar 

  141. O’Reilly, M. S. et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth. Cell 88, 277–285 (1997).

    PubMed  Google Scholar 

  142. Zorick, T. S. et al. High serum endostatin levels in down syndrome: implications for improved treatment and prevention of solid tumours. Eur. J. Hum. Genet. 9, 811–814 (2001).

    CAS  PubMed  Google Scholar 

  143. Obika, M. et al. Tumor growth inhibitory effect of ADAMTS1 is accompanied by the inhibition of tumor angiogenesis. Cancer Sci. 103, 1889–1897 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Oromendia, A. B., Dodgson, S. E. & Amon, A. Aneuploidy causes proteotoxic stress in yeast. Genes Dev. 26, 2696–2708 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Santaguida, S., Vasile, E., White, E. & Amon, A. Aneuploidy-induced cellular stresses limit autophagic degradation. Genes Dev. 29, 2010–2021 (2015). This article shows that aneuploid cells undergo a lysosomal stress response due to an increase in protein aggregation. This proteotoxic stress activates the transcription factor TFEB to upregulate the expression of genes required for autophagy-mediated protein degradation.

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Dodgson, S. E. et al. Chromosome-specific and global effects of aneuploidy in Saccharomyces cerevisiae. Genetics 202, 1395–1409 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Santaguida, S. & Amon, A. Aneuploidy triggers a TFEB-mediated lysosomal stress response. Autophagy 11, 2383–2384 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Donnelly, N., Passerini, V., Dürrbaum, M., Stingele, S. & Storchová, Z. HSF1 deficiency and impaired HSP90-dependent protein folding are hallmarks of aneuploid human cells. EMBO J. 33, 2374–2387 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Nicholson, J. M. et al. Chromosome mis-segregation and cytokinesis failure in trisomic human cells. eLife 4, e05068 (2015).

    PubMed Central  Google Scholar 

  150. Lentini, L., Barra, V., Schillaci, T. & Di Leonardo, A. MAD2 depletion triggers premature cellular senescence in human primary fibroblasts by activating a p53 pathway preventing aneuploid cells propagation. J. Cell. Physiol. 227, 3324–3332 (2012).

    CAS  PubMed  Google Scholar 

  151. Humbert, N. et al. Regulation of ploidy and senescence by the AMPK-related kinase NUAK1. EMBO J. 29, 376–386 (2010).

    CAS  PubMed  Google Scholar 

  152. Meena, J. K. et al. Telomerase abrogates aneuploidy-induced telomere replication stress, senescence and cell depletion. EMBO J. 34, 1371–1384 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Macedo, J. C. et al. FoxM1 repression during human aging leads to mitotic decline and aneuploidy-driven full senescence. Nat. Commun. 9, 2834 (2018).

    PubMed  PubMed Central  Google Scholar 

  154. Barroso-Vilares, M. et al. Small-molecule inhibition of aging-associated chromosomal instability delays cellular senescence. EMBO Rep. 21, e49248 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Collado, M. & Serrano, M. Senescence in tumours: evidence from mice and humans. Nat. Rev. Cancer 10, 51–57 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Prieur, A. & Peeper, D. S. Cellular senescence in vivo: a barrier to tumorigenesis. Curr. Opin. Cell Biol. 20, 150–155 (2008).

    CAS  PubMed  Google Scholar 

  157. Freund, A., Orjalo, A. V., Desprez, P.-Y. & Campisi, J. Inflammatory networks during cellular senescence: causes and consequences. Trends Mol. Med. 16, 238–246 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Pawlikowski, J. S., Adams, P. D. & Nelson, D. M. Senescence at a glance. J. Cell Sci. 126, 4061–4067 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Serrano, M. Final act of senescence. Nature 479, 481–482 (2011).

    CAS  PubMed  Google Scholar 

  160. Hoenicke, L. & Zender, L. Immune surveillance of senescent cells — biological significance in cancer- and non-cancer pathologies. Carcinogenesis 33, 1123–1126 (2012).

    CAS  PubMed  Google Scholar 

  161. Haugstetter, A. M. et al. Cellular senescence predicts treatment outcome in metastasised colorectal cancer. Br. J. Cancer 103, 505–509 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Coppé, J.-P. et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, 2853–2868 (2008).

    PubMed  Google Scholar 

  163. Rodier, F. et al. Persistent DNA damage signaling triggers senescence-associated inflammatory cytokine secretion. Nat. Cell Biol. 11, 973–979 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Acosta, J. C. et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 133, 1006–1018 (2008).

    CAS  PubMed  Google Scholar 

  165. Kuilman, T. et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 133, 1019–1031 (2008).

    CAS  PubMed  Google Scholar 

  166. Raulet, D. H. & Guerra, N. Oncogenic stress sensed by the immune system: role of NK cell receptors. Nat. Rev. Immunol. 9, 568–580 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Santaguida, S. et al. Chromosome mis-segregation generates cell-cycle-arrested cells with complex karyotypes that are eliminated by the immune system. Dev. Cell 41, 638–651.e5 (2017). This study describes a mechanism by which the immune system can detect and eliminate aneuploid cells. Aneuploid cells exhibit senescent features and secrete proinflammatory signals, which can promote their own immune clearance.

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Krtolica, A., Parrinello, S., Lockett, S., Desprez, P. Y. & Campisi, J. Senescent fibroblasts promote epithelial cell growth and tumorigenesis: a link between cancer and aging. Proc. Natl Acad. Sci. USA 98, 12072–12077 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. He, Q. et al. Chromosomal instability-induced senescence potentiates cell non-autonomous tumourigenic effects. Oncogenesis 7, 62 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Cortés-Ciriano, I. et al. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing. Nat. Genet. 52, 331–341 (2020).

    PubMed  PubMed Central  Google Scholar 

  171. Kneissig, M. et al. Micronuclei-based model system reveals functional consequences of chromothripsis in human cells. eLife 8, e50292 (2019).

    PubMed  PubMed Central  Google Scholar 

  172. Ly, P. & Cleveland, D. W. Rebuilding chromosomes after catastrophe: emerging mechanisms of chromothripsis. Trends Cell Biol. 27, 917–930 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Forment, J. V., Kaidi, A. & Jackson, S. P. Chromothripsis and cancer: causes and consequences of chromosome shattering. Nat. Rev. Cancer 12, 663–670 (2012).

    CAS  PubMed  Google Scholar 

  174. Hong, C., Tijhuis, A. E. & Foijer, F. The cGAS paradox: contrasting roles for cGAS-STING pathway in chromosomal instability. Cells 8, 1228 (2019).

    CAS  PubMed Central  Google Scholar 

  175. Levine, M. S. et al. Centrosome amplification is sufficient to promote spontaneous tumorigenesis in mammals. Dev. Cell 40, 313–322.e5 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Yarchoan, M., Johnson, B. A., Lutz, E. R., Laheru, D. A. & Jaffee, E. M. Targeting neoantigens to augment antitumour immunity. Nat. Rev. Cancer 17, 569 (2017).

    CAS  PubMed  Google Scholar 

  177. Swann, J. B. & Smyth, M. J. Immune surveillance of tumors. J. Clin. Invest. 117, 1137–1146 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  178. Davoli, T., Uno, H., Wooten, E. C. & Elledge, S. J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 355, eaaf8399 (2017).

    PubMed  PubMed Central  Google Scholar 

  179. Wang, R. W., Viganò, S., Ben-David, U., Amon, A. & Santaguida, S. Aneuploid cells activate NF-κB to promote their immune clearance by NK cells. Preprint at bioRxiv https://doi.org/10.1101/2020.06.25.172239 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  180. Senovilla, L. et al. An immunosurveillance mechanism controls cancer cell ploidy. Science 337, 1678–1684 (2012).

    CAS  PubMed  Google Scholar 

  181. Boilève, A. et al. Immunosurveillance against tetraploidization-induced colon tumorigenesis. Cell Cycle 12, 473–479 (2013).

    PubMed  PubMed Central  Google Scholar 

  182. Mackenzie, K. J. et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 548, 461–465 (2017). This study reveals how damaged DNA is detected in the cytosol by cGAS, leading to an inflammatory response. The detection of mis-segregated DNA in micronuclei by cGAS could serve as a cell-intrinsic immunosurveillance programme to clear potentially tumorigenic cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Deng, L. et al. STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors. Immunity 41, 843–852 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  184. Tripathi, R., Modur, V., Senovilla, L., Kroemer, G. & Komurov, K. Suppression of tumor antigen presentation during aneuploid tumor evolution contributes to immune evasion. Oncoimmunology 8, 1657374 (2019).

    PubMed  PubMed Central  Google Scholar 

  185. Duncan, A. W. et al. Frequent aneuploidy among normal human hepatocytes. Gastroenterology 142, 25–28 (2012).

    PubMed  Google Scholar 

  186. Pack, S. D. et al. Individual adult human neurons display aneuploidy: detection by fluorescence in situ hybridization and single neuron PCR. Cell Cycle 4, 1758–1760 (2005).

    CAS  PubMed  Google Scholar 

  187. Knouse, K. A., Wu, J., Whittaker, C. A. & Amon, A. Single cell sequencing reveals low levels of aneuploidy across mammalian tissues. Proc. Natl Acad. Sci. USA 111, 13409–13414 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  188. van den Bos, H. et al. Single-cell whole genome sequencing reveals no evidence for common aneuploidy in normal and Alzheimer’s disease neurons. Genome Biol. 17, 116 (2016). Knouse et al. (2014) and van den Bos et al. (2016) use single-cell sequencing to demonstrate that aneuploidy is rare in normal somatic tissue and in neurons from individuals with Alzheimer disease. These findings refute previous studies that reported high levels of aneuploidy in the brain and liver and that linked aneuploidy to the pathogenesis of Alzheimer disease.

    PubMed  PubMed Central  Google Scholar 

  189. Tang, Y.-C., Williams, B. R., Siegel, J. J. & Amon, A. Identification of aneuploidy-selective antiproliferation compounds. Cell 144, 499–512 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  190. Schukken, K. M. et al. Altering microtubule dynamics is synergistically toxic with spindle assembly checkpoint inhibition. Life Sci. Alliance 3, e201900499 (2020).

    PubMed  PubMed Central  Google Scholar 

  191. Tang, Y.-C. et al. Aneuploid cell survival relies upon sphingolipid homeostasis. Cancer Res. 77, 5272–5286 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  192. Janssen, A., Kops, G. J. P. L. & Medema, R. H. Elevating the frequency of chromosome mis-segregation as a strategy to kill tumor cells. Proc. Natl Acad. Sci. USA 106, 19108–19113 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Bakhoum, S. F., Thompson, S. L., Manning, A. L. & Compton, D. A. Genome stability is ensured by temporal control of kinetochore-microtubule dynamics. Nat. Cell Biol. 11, 27–35 (2009).

    CAS  PubMed  Google Scholar 

  194. Ertych, N. et al. Increased microtubule assembly rates influence chromosomal instability in colorectal cancer cells. Nat. Cell Biol. 16, 779–791 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  195. Maia, A. R. R. et al. Inhibition of the spindle assembly checkpoint kinase TTK enhances the efficacy of docetaxel in a triple-negative breast cancer model. Ann. Oncol. 26, 2180–2192 (2015).

    CAS  PubMed  Google Scholar 

  196. Kops, G. J. P. L., Foltz, D. R. & Cleveland, D. W. Lethality to human cancer cells through massive chromosome loss by inhibition of the mitotic checkpoint. Proc. Natl Acad. Sci. USA 101, 8699–8704 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  197. Wengner, A. M. et al. Novel Mps1 kinase inhibitors with potent antitumor activity. Mol. Cancer Ther. 15, 583–592 (2016).

    CAS  PubMed  Google Scholar 

  198. Cohen-Sharir, Y. et al. Selective vulnerability of aneuploid human cancer cells to inhibition of the spindle assembly checkpoint. Preprint at bioRxiv https://doi.org/10.1101/2020.06.18.159038 (2020).

    Article  Google Scholar 

  199. Quinton, R. J. et al. Whole genome doubling confers unique genetic vulnerabilities on tumor cells. Preprint at bioRxiv https://doi.org/10.1101/2020.06.18.159095 (2020).

    Article  Google Scholar 

  200. Marquis, C. et al. Chromosomally unstable tumor cells specifically require KIF18A for proliferation. Preprint at bioRxiv https://doi.org/10.1101/2020.06.18.159327 (2020). The preprints Cohen-Sharir et al. (2020), Quinton et al. (2020) and Marquis et al. (2020) demonstrate that highly aneuploid cells are dependent on the kinesin KIF18A for mitotic progression. KIF18A inhibition may therefore serve as a strategy to selectively target aneuploid tumours.

    Article  Google Scholar 

  201. Falor, W. H. & Ward, R. M. Prognosis in early carcinoma of the bladder based on chromosomal analysis. J. Urol. 119, 44–48 (1978).

    CAS  PubMed  Google Scholar 

  202. Alimena, G., Annino, L., Balestrazzi, P., Montuoro, A. & Dallapiccola, B. Cytogenetic studies in acute leukaemias. Prognostic implications of chromosome imbalances. Acta Haematol. 58, 234–239 (1977).

    CAS  PubMed  Google Scholar 

  203. Zetterberg, A. & Esposti, P. L. Prognostic significance of nuclear DNA levels in prostatic carcinoma. Scand. J. Urol. Nephrol. Suppl. 55, 53–58 (1980).

    CAS  PubMed  Google Scholar 

  204. Okagaki, T., Meyer, A. A. & Sciarra, J. J. Prognosis of irradiated carcinoma of cervix uteri and nuclear DNA in cytologic postirradiation dysplasia. Cancer 33, 647–652 (1974).

    CAS  PubMed  Google Scholar 

  205. Hieronymus, H. et al. Tumor copy number alteration burden is a pan-cancer prognostic factor associated with recurrence and death. eLife 7, e37294 (2018).

    PubMed  PubMed Central  Google Scholar 

  206. Ben-David, U. et al. The landscape of chromosomal aberrations in breast cancer mouse models reveals driver-specific routes to tumorigenesis. Nat. Commun. 7, 12160 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  207. Sanson, K. R. et al. Optimized libraries for CRISPR-Cas9 genetic screens with multiple modalities. Nat. Commun. 9, 5416 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Zhan, T., Rindtorff, N., Betge, J., Ebert, M. P. & Boutros, M. CRISPR/Cas9 for cancer research and therapy. Semin. Cancer Biol. 55, 106–119 (2019).

    CAS  PubMed  Google Scholar 

  209. Lin, A. & Sheltzer, J. M. Discovering and validating cancer genetic dependencies: approaches and pitfalls. Nat. Rev. Genet. 21, 671–682 (2020).

    CAS  PubMed  Google Scholar 

  210. Esfahani, K. et al. A review of cancer immunotherapy: from the past, to the present, to the future. Curr. Oncol. 27, S87–S97 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  211. Fournier, R. E. A general high-efficiency procedure for production of microcell hybrids. Proc. Natl Acad. Sci. USA 78, 6349–6353 (1981).

    CAS  PubMed  PubMed Central  Google Scholar 

  212. Bennett, A. et al. Cenp-E inhibitor GSK923295: Novel synthetic route and use as a tool to generate aneuploidy. Oncotarget 6, 20921–20932 (2015).

    PubMed  PubMed Central  Google Scholar 

  213. Thomas, R., Marks, D. H., Chin, Y. & Benezra, R. Whole chromosome loss and associated breakage-fusion-bridge cycles transform mouse tetraploid cells. EMBO J. 37, 201–218 (2018).

    CAS  PubMed  Google Scholar 

  214. Kimura, M. et al. Proliferation dynamics in cultured skin fibroblasts from Down syndrome subjects. Free Radic. Biol. Med. 39, 374–380 (2005).

    CAS  PubMed  Google Scholar 

  215. Hwang, S. et al. Suppressing aneuploidy-associated phenotypes improves the fitness of trisomy 21 cells. Cell Rep. 29, 2473–2488.e5 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  216. Zuo, E. et al. CRISPR/Cas9-mediated targeted chromosome elimination. Genome Biol. 18, 224 (2017).

    PubMed  PubMed Central  Google Scholar 

  217. Giuliano, C. J., Lin, A., Girish, V. & Sheltzer, J. M. Generating single cell–derived knockout clones in mammalian cells with CRISPR/Cas9. Curr. Protoc. Mol. Biol. 128, e100 (2019).

    PubMed  PubMed Central  Google Scholar 

  218. Fernández-Miranda, G. et al. Genetic disruption of aurora B uncovers an essential role for aurora C during early mammalian development. Dev. Camb. Engl. 138, 2661–2672 (2011).

    Google Scholar 

  219. Ricke, R. M., Jeganathan, K. B. & van Deursen, J. M. Bub1 overexpression induces aneuploidy and tumor formation through Aurora B kinase hyperactivation. J. Cell Biol. 193, 1049–1064 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  220. Rao, C. V. et al. Colonic tumorigenesis in BubR1+/–ApcMin/+ compound mutant mice is linked to premature separation of sister chromatids and enhanced genomic instability. Proc. Natl Acad. Sci. USA 102, 4365–4370 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Dai, W. et al. Slippage of mitotic arrest and enhanced tumor development in mice with BubR1 haploinsufficiency. Cancer Res. 64, 440–445 (2004).

    CAS  PubMed  Google Scholar 

  222. Baker, D. J. et al. Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by BubR1 insufficiency. Nat. Cell Biol. 10, 825–836 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  223. Nam, H.-J. & van Deursen, J. M. Cyclin B2 and p53 control proper timing of centrosome separation. Nat. Cell Biol. 16, 538–549 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  224. Li, M., Fang, X., Wei, Z., York, J. P. & Zhang, P. Loss of spindle assembly checkpoint–mediated inhibition of Cdc20 promotes tumorigenesis in mice. J. Cell Biol. 185, 983–994 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  225. Mukherjee, M. et al. MMTV-Espl1 transgenic mice develop aneuploid, estrogen receptor alpha (ERα)-positive mammary adenocarcinomas. Oncogene 33, 5511–5522 (2014).

    CAS  PubMed  Google Scholar 

  226. Iwanaga, Y. et al. Heterozygous deletion of mitotic arrest-deficient protein 1 (MAD1) increases the incidence of tumors in mice. Cancer Res. 67, 160–166 (2007).

    CAS  PubMed  Google Scholar 

  227. Michel, L. S. et al. MAD2 haplo-insufficiency causes premature anaphase and chromosome instability in mammalian cells. Nature 409, 355–359 (2001).

    CAS  PubMed  Google Scholar 

  228. Chi, Y.-H., Ward, J. M., Cheng, L. I., Yasunaga, J. & Jeang, K.-T. Spindle assembly checkpoint and p53 deficiencies cooperate for tumorigenesis in mice. Int. J. Cancer 124, 1483–1489 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  229. Foijer, F. et al. Deletion of the MAD2L1 spindle assembly checkpoint gene is tolerated in mouse models of acute T-cell lymphoma and hepatocellular carcinoma. eLife 6, e20873 (2017).

    PubMed  PubMed Central  Google Scholar 

  230. Foijer, F. et al. Chromosome instability induced by Mps1 and p53 mutation generates aggressive lymphomas exhibiting aneuploidy-induced stress. Proc. Natl Acad. Sci. USA 111, 13427–13432 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  231. Lu, L.-Y. et al. Polo-like kinase 1 is essential for early embryonic development and tumor suppression. Mol. Cell. Biol. 28, 6870–6876 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Ko, M. A. et al. Plk4 haploinsufficiency causes mitotic infidelity and carcinogenesis. Nat. Genet. 37, 883–888 (2005).

    CAS  PubMed  Google Scholar 

  233. Rosario, C. O. et al. Plk4 is required for cytokinesis and maintenance of chromosomal stability. Proc. Natl Acad. Sci. USA 107, 6888–6893 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  234. Coelho, P. A. et al. Over-expression of Plk4 induces centrosome amplification, loss of primary cilia and associated tissue hyperplasia in the mouse. Open Biol. 5, 150209 (2015).

    PubMed  PubMed Central  Google Scholar 

  235. Babu, J. R. et al. Rae1 is an essential mitotic checkpoint regulator that cooperates with Bub3 to prevent chromosome missegregation. J. Cell Biol. 160, 341–353 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  236. Aguirre-Portolés, C. et al. Tpx2 controls spindle integrity, genome stability, and tumor development. Cancer Res. 72, 1518–1528 (2012).

    PubMed  Google Scholar 

  237. van Ree, J. H., Jeganathan, K. B., Malureanu, L. & van Deursen, J. M. Overexpression of the E2 ubiquitin–conjugating enzyme UbcH10 causes chromosome missegregation and tumor formation. J. Cell Biol. 188, 83–100 (2010).

    PubMed  PubMed Central  Google Scholar 

  238. Zhang, C.-Z. et al. Chromothripsis from DNA damage in micronuclei. Nature 522, 179–184 (2015). This report demonstrates that chromothripsis can arise as a result of chromosome mis-segregation. The authors show that mis-segregated chromosomes can become trapped in micronuclei, where the DNA is subjected to genomic damage and may undergo a series of chromothriptic rearrangements.

    CAS  PubMed  PubMed Central  Google Scholar 

  239. Stephens, P. J. et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 144, 27–40 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  240. Huang, Y., Fenech, M. & Shi, Q. Micronucleus formation detected by live-cell imaging. Mutagenesis 26, 133–138 (2011).

    CAS  PubMed  Google Scholar 

  241. Hatch, E. M., Fischer, A. H., Deerinck, T. J. & Hetzer, M. W. Catastrophic nuclear envelope collapse in cancer cell micronuclei. Cell 154, 47–60 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  242. Storlazzi, C. T. et al. Gene amplification as double minutes or homogeneously staining regions in solid tumors: Origin and structure. Genome Res. 20, 1198–1206 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. de Oliveira Mann, C. C. & Kranzusch, P. J. cGAS conducts micronuclei DNA surveillance. Trends Cell Biol. 27, 697–698 (2017).

    PubMed  Google Scholar 

  244. Ohtani, N. Deciphering the mechanism for induction of senescence-associated secretory phenotype (SASP) and its role in ageing and cancer development. J. Biochem. 166, 289–295 (2019).

    CAS  Google Scholar 

  245. Zierhut, C. et al. The cytoplasmic DNA sensor cGAS promotes mitotic cell death. Cell 178, 302–315.e23 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank members of the Sheltzer laboratory for helpful comments on the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to all aspects of the article (researching data, substantially contributing to discussion of content, and writing, reviewing and editing the manuscript before submission).

Corresponding author

Correspondence to Jason M. Sheltzer.

Ethics declarations

Competing interests

J.M.S. is a co-founder of Meliora Therapeutics, is a member of the Scientific Advisory Board of Tyra Biosciences and has received consulting fees from Merck and Ono Pharmaceutical Co. The other authors declare no competing interests.

Additional information

Dedication

J.M.S. dedicates this article to the memory of Angelika Amon, for her outstanding mentorship, her passion for tackling unorthodox questions and her pioneering research into the consequences of aneuploidy.

Peer review information

Nature Reviews Cancer thanks A. Holland, G. Kops and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Polyploidy

Describing an abnormal number of chromosomes, such that a cell’s karyotype is an integer multiple of the haploid complement greater than 2.

Chromosomal instability

(CIN). An abnormally high rate of chromosomal mis-segregation that may result in aneuploidy.

Microcell-mediated chromosome transfer

Technique involving the transfer of specific donor chromosomes into recipient cell lines as a means of creating aneuploid cells.

Spindle-assembly checkpoint

Cell cycle checkpoint that maintains genome stability by ensuring proper chromosome attachment at the kinetochore via anchorage to the microtubule spindle apparatus before the initiation of anaphase.

Cre–lox-mediated recombination

A technique used to introduce site-specific deletions, insertions, translocations and inversions within chromosomes.

Robertsonian translocations

Translocations in which two acrocentric chromosomes fuse to share a single centromere.

Proteotoxic stress

Impaired cell function arising as a result of defective protein translation, folding and/or turnover.

Epithelial–mesenchymal transition

The process by which epithelial cells transition into mesenchymal cells through the loss of cell polarity and cell–cell adhesion and the subsequent gain of migratory and invasive abilities.

Mesenchymal–epithelial transition

The process by which mesenchymal cells lose their migratory and invasive properties and transition into epithelial cells capable of increased cell–cell adhesion.

Apc Min/+ mice

Mice that develop spontaneous intestinal adenomas due to a point mutation in the mouse homologue of the APC gene. They are frequently used as models for intestinal tumorigenesis.

Hypomorphic

A mutation resulting in the reduction, but not the complete loss, of gene function.

Cyclic GMP–AMP synthase (cGAS)–stimulator of interferon genes (STING)

Immune system pathway that detects cytosolic DNA and elicits a downstream inflammatory response to aid in immune clearance.

Down syndrome

Genetic disorder associated with developmental and intellectual delays due to the added presence of a third copy of chromosome 21.

Ts65Dn mouse

A commonly used genetic mouse model for Down syndrome that is segmentally trisomic for part of mouse chromosome 16, which encodes genes homologous to those found on human chromosome 21.

Pearson correlation coefficient

Statistic measuring the strength of association between two variables.

Replication stress

A cellular state in which the fidelity of DNA replication is compromised, often characterized by the frequent collapse of the replication fork.

Hyper-recombination

A cellular state defined by elevated levels of recombination.

Senescence

Process by which proliferating cells cease dividing due to extracellular or intracellular stress and enter a state of permanent cell cycle arrest.

Senescence-associated secretory phenotype

(SASP). Unique secretome consisting of chemokines, cytokines, growth factors and immune regulators that are released into the microenvironment by senescent cells.

MMTV–PyMT mice

Common model of metastatic breast cancer in which the mouse mammary tumour virus (MMTV) promotor drives the expression of the polyomavirus middle T antigen (PyMT), leading to spontaneous and rapid tumorigenesis in mouse mammary epithelial cells.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vasudevan, A., Schukken, K.M., Sausville, E.L. et al. Aneuploidy as a promoter and suppressor of malignant growth. Nat Rev Cancer 21, 89–103 (2021). https://doi.org/10.1038/s41568-020-00321-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-00321-1

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer