Oncogene (2013) 32, 4459–4470 & 2013 Macmillan Publishers Limited All rights reserved 0950-9232/13 www.nature.com/onc

REVIEW Genetic instability: tipping the balance

A Janssen1,2 and RH Medema1

Tumor cells typically contain a genome that is highly divergent from the genome of normal, non-transformed cells. This genetic divergence is caused by a number of distinct changes that the tumor cell acquires during its transformation from a normal cell into a tumorigenic counterpart. Changes to the genome include mutations, deletions, insertions, and also gross chromosomal aberrations, such as translocations and whole chromosome gains or losses. This genetic disorder of the tumor cell has complicated the identification of crucial driver mutations that cause . Moreover, the large genetic divergence between different tumors causes them to behave very differently, and makes it difficult to predict response to therapy. In addition, tumor cells are genetically unstable and frequently acquire new mutations and/or gross chromosomal aberrations as they divide. This is beneficial for the overall capacity of a tumor to adapt to changes in its environment, but newly acquired genetic alterations can also compromise the genetic dominance of the tumor cell and thus affect tumor cell viability. Here, we review the mechanisms that can cause gross chromosomal aberrations, and discuss how these affect tumor cell viability.

Oncogene (2013) 32, 4459–4470; doi:10.1038/onc.2012.576; published online 17 December 2012 Keywords: chromosome instability; cancer; ; DNA damage; translocations;

STRUCTURAL CHROMOSOMAL INSTABILITY Breakage Syndrome, Bloom’s syndrome, ataxia telangiectasia or Translocations, one of the most prominent types of structural mutations in BRCA1 and 2, display an increased susceptibility to 18 chromosomal changes, have been found in many cancer types1 form structural chromosomal changes. Cells from these patients (Figure 1). In hematological malignancies, several translocations accumulate translocations due to mutations in DNA repair have been identified that contribute to specific gene fusions, which proteins, such as NBS1, BLM helicase, ATM kinase or the BRCA1 are thought to be drivers in the process of tumorigenesis.2 The first and BRCA2 proteins. Moreover, it was shown long ago that several identified translocation in human cancer was the Philadelphia DSB-inducing agents, such as ionizing irradiation, UV-light and 3 chemical mutagens, can also result in the formation of chromosome, which results in the formation of a fusion between 19 the BCR and Abl genes, and is causative in the development of chromosomal aberrations. chronic myeloid leukemia.4 Translocations found in cancer can be The structural instability of cancer cells is not merely caused by balanced, creating two reciprocal chromosomal fusions that are inherited genetic defects or damage induced by exogenous homogeneously present in all tumor cells.5 However, more often, agents. Cancer cells can also acquire new translocations through the breakage-fusion-bridge (BFB) cycle, a process first described in cancer cells within a tumor display mosaic structural changes, 20,21 indicating that continue to rearrange at a high rate in maize meiosis. In the BFB cycle, chromosomes that are broken 6–11 by DSBs first fuse with other broken chromosomal parts, for the established tumor. Generally, these chromosome structure 22 instabilities are thought to promote tumorigenesis by providing example, through fusion at dysfunctional telomeres. These telomere fusion events result in dicentric (two centromeres) continuous genetic diversification within the tumor that facilitates chromosomes, often found in tumors.22,23 The presence of two adaptation to environmental changes, for example through loss of centromeres on these aberrantly shaped chromosomes can result certain tumor suppressors or gain of specific oncogenes.7,12 in improper microtubule (MT) attachments in mitosis, such that Moreover, this continued genetic diversification could help the the two centromeres on a single chromatid become attached to tumor in acquiring drug resistance, cope with increased hypoxia, or opposite spindle poles. These attachments can induce chromatin escape challenges by the immune system. In line with this, an bridging in telophase, resulting in breakage of the fused increased occurrence of structural chromosomal aberrations 20,24 1,13,14 chromosome during cytokinesis, starting another BFB cycle. correlates with increased tumor grade. Such repetitive BFB events increase the number of aberrant Structural chromosomal instabilities arise through mis- or chromosomes in the offspring.20,21 BFB has indeed been unrepaired DNA double-stranded breaks (DSBs). Homologous correlated with an increase in intratumor heterogeneity and recombination and non-homologous end joining are thought to might therefore be an important factor in structural instability.25 be the two main repair pathways contributing to the formation of structural aberrations.15–17 Especially the error-prone non- homologous end joining pathway, which can ligate any two broken DNA ends together, is held responsible for the formation NUMERICAL CHROMOSOMAL INSTABILITY of structural aberrations.15 Indeed, individuals with genetic defects Another striking hallmark of cancer cells is the presence of an that affect repair of DSBs, such as patients with Nijmegen abnormal chromosome number8,26,27 (Figure 1), termed

1Division of Cell Biology, Netherlands Cancer Institute, Amsterdam, The Netherlands and 2Department of Medical Oncology and Cancer Genomics Center, University Medical Center Utrecht, Utrecht, The Netherlands. Correspondence: Dr R Medema, Netherlands Cancer Institute Division of Cell Biology, Plesmanlaan 121, Amsterdam, Holland 1066 CX, The Netherlands. E-mail: [email protected] Received 24 September 2012; revised 23 October 2012; accepted 24 October 2012; published online 17 December 2012 Genetic instability in tumors A Janssen and RH Medema 4460

Figure 1. Typical karyotype of a cancer cell. Karyotype of a human osteosarcoma cell line (U2OS) revealing a variety of numerical and structural chromosomal abnormalities.

aneuploidy, a state in which cells do not contain an exact multiple of the haploid DNA content. On average, 25 percent of the genome of a cancer cell is affected by numerical changes of either whole chromosomes or complete chromosomal arms.6 The aneuploid karyotype can be stably propagated in a population of (tumor) cells,6,28,29 but more often chromosome numbers continuously change between cancer cells and their offspring.30 This continuous change in chromosome number is termed whole chromosomal instability (CIN)30.31 and is correlated 32–38 Figure 2. Various mitotic defects can lead to CIN. Absence of mitotic with tumor grade, metastasis and poor prognosis. Moreover, checkpoint signaling allows initiation in the presence of CIN has been associated with resistance to chemotherapeutics, unattached kinetochores, whereas cohesion loss or merotelic such as the widely used MT-stabilizing agent Paclitaxel.39–41 attachments induce chromosomal segregation errors by incorrect Various hypotheses have been postulated on how CIN and kinetochore orientation or the induction of lagging chromatids aneuploidy could contribute to tumorigenesis.42–46 The general respectively. idea is that whole chromosome gains and losses during can, as suggested above for structural changes, provide a mode for cancer cells to adapt to their environment and and causes embryonic lethality in mouse,51–58 likely because it continuously divide.47,48 results in continuous chromosome missegregation. Thus, subsequent cell divisions of a viable aneuploid karyotype results in improper propagation of the genetic material, resulting in high CAUSES OF NUMERICAL CIN rates of cell death in the respective daughter cells. In contrast, Since the initial discovery of CIN in a variety of colon cancer cell partial checkpoint dysfunction results in a low frequency of lines in 1997,30 many researchers have investigated the underlying chromosome segregation errors, a low rate of cell death and the cause of this striking phenotype, found to be present in many potential to generate a new viable karyotype that is relatively other tumor types as well. Several mechanisms have been stable. Partial mitotic checkpoint activity could be responsible for CIN in tumor cells by allowing mitotic exit in the presence of one proposed and tested using a variety of cancer cell lines and 51,58–60 mouse models. It seems highly unlikely that one single or more unattached kinetochores (Figure 2). However, CIN mechanism can be held responsible for the CIN observed in the cancer cell lines that were initially thought to have a severely different types of tumors. Below we outline the different cellular impaired mitotic checkpoint due to mutations in the mitotic checkpoint kinase Bub1,61 were subsequently shown to have a causes that have been postulated and the genetic defects that 62 could underlie these phenotypes. very sturdy mitotic checkpoint. Nonetheless, altered expression levels and mutations in mitotic checkpoint genes have been identified in human cancer that compromise checkpoint Causes of numerical CIN: Mitotic checkpoint defects function.43,63–76 In one specific syndrome, mosaic-variegated The mitotic checkpoint has evolved to safeguard genetic stability, aneuploidy, which is linked to cancer predisposition at a very a function it performs by monitoring the attachment status of young age, mono- and biallelic mutations in the BUB1B gene have chromosomes to the mitotic spindle. During mitosis, sister been identified.63 BUB1B encodes for the mitotic checkpoint chromatid pairs need to bi-orient on the mitotic spindle, meaning protein BubR1 and these mutations can lead to the complete that sister chromatids within a pair are attached to opposite absence of BubR1 mRNA or single amino-acid substitutions in the spindle poles. Improper attachment of as much as a single sister BubR1 protein product.77 In all cases, these mutations lead to chromatid is sufficient to sustain the mitotic checkpoint and lower BubR1 protein levels and decreased mitotic checkpoint prevent the onset of anaphase. Biorientation is achieved through activity,64,77,78 linking the cancer predisposition of mosaic- binding of the MT attachment site of each sister chromatid, called variegated aneuploidy patients to defects in the mitotic kinetochore, to MTs coming from one spindle pole. Once all checkpoint. kinetochores in a cell are properly attached to the spindle, the The occurrence of mutations in mitotic checkpoint genes in mitotic checkpoint is satisfied and anaphase onset is no longer human remains extremely rare.79–82 Surprisingly, many inhibited.49 Defects in mitotic checkpoint signaling directly lead to CIN tumors actually display enhanced levels of mitotic checkpoint chromosome missegregations by allowing mitotic exit in the proteins.33,83–89 One explanation for this increase in expression of presence of unattached kinetochores (Figure 2). Indeed, the mitotic checkpoint genes lies in the dysfunction of the Rb hypothesis that has been investigated the most, postulates that pathway, resulting in overexpression of the mitotic checkpoint mitotic checkpoint defects could underlie CIN.50 Complete mitotic protein MAD2.90 Indeed, deregulation of the Rb pathway can checkpoint loss is lethal to all dividing cell types studied thus far result in CIN in several organisms.91,92 Notably, CIN induced in

Oncogene (2013) 4459 – 4470 & 2013 Macmillan Publishers Limited Genetic instability in tumors A Janssen and RH Medema 4461 Rb-negative cell lines was directly dependent on increased MAD2 causes of merotelic attachments have been hypothesized, of levels.90 In line with this, MAD2 overexpression in mice results in which the most significant ones are outlined below. aneuploidy and spontaneous tumor formation93 (See Table 1) and CIN in Rb-negative tumors depends on increased MAD2 levels.94 Causes of merotelic attachments: aberrant spindle morphology. In summary, decreased expression levels of checkpoint proteins One way in which merotelic attachments can arise is through as well as mutations in mitotic checkpoint genes have been abnormal spindle assembly in mitosis. Initial clues on this came observed in tumor tissues, but these are relatively rare. Instead, from the observation that multipolar spindle assembly in mere overexpression of mitotic checkpoint proteins, in particular kangaroo cells often led to kinetochore-binding to two spindle MAD2, seems to be the more prevalent cause in the induction of poles,110 producing merotelic attachments and lagging CIN in various tumor types. chromosomes.111 Thus, proper spindle polarity is important for the establishment of proper chromosome attachments. In Causes of numerical CIN: cohesion loss mammalian cells, spindle polarity is largely dictated by the . The two centrosomes that are normally present Duplicated sister chromatids are held together until anaphase by separate before mitosis, and subsequently organize the spindle the cohesin complex. Maintenance of cohesion between the two poles. Drugs that perturb separation also increase the sister centromeres is essential for chromosome biorientation and number of merotelic attachments and induce chromosome hence for proper chromosome segregation.95–97 Loss of cohesion segregation errors.107 before anaphase results in premature sister chromatid separation, Interestingly, multipolar spindles are often observed in CIN which eventually results in chromosome segregation errors and tumors,112–115 which indicates that aberrant spindle morphology aneuploidy (Figure 2). Various genes encoding proteins involved might be responsible for merotelic attachments and CIN induction in cohesion establishment or maintenance have been found to be in vivo as well. Multipolar spindles are often the result of mutated in aneuploid tumors.96,98 In addition, a recent report centrosome overduplication, a common trait of cancer cells.116 observed frequent deletion of part of the X chromosome in Persistence of multipolar spindles in anaphase results in severe genetically unstable tumor samples.99 This locus encodes for SA2, chromosome missegregations and the severely aneuploid which is a subunit of the cohesin complex. Interestingly, targeted daughter cells often die in the next G1 phase.117 However, inactivation of this locus, termed STAG2, resulted in CIN in tumor cells that harbor multiple centrosomes can progress otherwise chromosome-stable near-diploid cells. Moreover, through a transient multipolar state, after which the reconstitution of STAG2 in CIN tumor cells harboring a deletion supernumerary centrosomes cluster during mitosis and form a of the endogenous locus, reverted the CIN phenotype and seemingly normal bipolar spindle118,119 (Figure 3). The transient enhanced chromosomal stability in these lines.99 multipolar state that results from having too many centrosomes is Defective sister chromatid cohesion as a result of somatic very prone to attach single kinetochores to multiple spindle mutations may represent a major cause of CIN in human cancers. poles.110,111,118,119 These multipolar attachments are converted to In line with this, overexpression of Separase, the protease that merotelic attachments upon centrosome clustering and can cleaves cohesin upon mitotic exit, has been observed in breast produce chromosome segregation errors118,119 (Figure 3). cancer samples and transient overexpression of Separase can also Supernumerary centrosomes can arise through a number of produce aneuploidy in human cells.100 Besides its role in sister different mechanisms, such as centrosome overduplication, chromatid cohesion, cohesin has also been implicated in DNA cytokinesis failure or virus-induced cell fusion, with the last two replication, repair of DNA damage and protection of telomeres.101 also resulting in increased ploidy. Interestingly, tetraploidy is also Therefore, various other mechanisms might underlie the frequently observed in tumor cells and has been shown to aneuploidy observed in cells that carry mutations in one of the promote tumorigenesis in mice120 as well as transformation cohesin subunits102 and further research is needed to investigate in vitro and in vivo.121 It is currently unclear if the tumorigenic the contribution of cohesion defects in the establishment of CIN. nature of a tetraploid cell is due to the fact that it contains an extra copy of the genome or because it contains twice the amount of Causes of numerical CIN: Merotelic attachments centrosomes. The extra copy of the genome could set the stage Although CIN cell lines do not necessarily possess a weakened for subsequent chromosome loss to produce the genetic mitotic checkpoint, many CIN cell lines do have an increased imbalance that drives tumorigenesis. Alternatively, the extra occurrence of lagging chromosomes due to unresolved merotelic centrosomes could be the critical driving force to produce that attachments to the mitotic spindle103,104 (Figure 2). Merotelic genetic imbalance by enhancing the chance of creating merotelic attachments link a single kinetochore to two spindle poles, instead attachments. of one. If this merotelic attachment persists, the respective sister Very recently, a novel and remarkable mechanism to produce chromatid will end up in between the two packs of DNA during tetraploid cells was described. The process was named ‘entosis’: anaphase, producing a lagging chromosome105,106 (Figure 2). the invasion of a cell into another cell. Entosis has been described Most merotelic attachments are resolved by the Aurora B-depen- as a non-apoptotic cell death program of cells that have been dent error-correction machinery before anaphase.12,107–109 detached from the extracellular matrix.122 Internalization of these However, merotelic attachments are not actively being sensed detached cells into host cells most often results in cell death. by the mitotic checkpoint and therefore do not delay mitotic exit Entosis has been observed in tumors, and its occurrence correlates to allow for correction. Such merotelic attachments can therefore with tumor grade.122–124 Cells taken up by entosis are usually persist into anaphase and cause chromosome missegregations destroyed before the host divides, but if an intact invading cell and aneuploidy.103,106 Although not all lagging chromosomes ends up in the host cell’s cleavage plane, it can inhibit cytokinesis. necessarily lead to aneuploidy in the respective daughter cells,12 This produces a tetraploid cell that will build a multipolar spindle merotelic attachments are very prominent in CIN cells and in the next mitosis, a combination that is likely to produce therefore thought to be a major cause of aneuploidy. aneuploid daughter cells.124,125 This illustrates that one single As indicated in Figure 2, loss of cohesion between two sister cytokinesis failure could initiate chromosome segregation errors in chromatids could increase the occurrence of merotelic attach- the tumor cells’ offspring, ultimately leading to CIN. ments. In the absence of cohesion, kinetochores become more In addition to the abovementioned models for the induction of prone to bind MTs coming from two poles due to the absence of multiple centrosomes and subsequent aberrant spindle formation, the physical barrier normally created by the presence of another loss of the DNA damage checkpoint kinase Chk2 has also been sister chromatid. Many other cellular, genetic and molecular implicated in the induction of abnormal spindles through

& 2013 Macmillan Publishers Limited Oncogene (2013) 4459 – 4470 Genetic instability in tumors A Janssen and RH Medema 4462

Figure 3. Schematic representation of underlying causes of merotelic attachments. Centrosome coalescence creates merotelic attachments by allowing clustering of centrosomes, which had already established MT interactions with kinetochores. Impaired kinetochore MT dynamics inhibits the release of erroneous attachments, whereas the presence of multiple centromeres increases the chance of creating faulty attachments.

deregulation of MT dynamics.126 Chk2 is thought to be important when compared with that of tumor cells.131 Since MAD2 for proper spindle formation through phosphorylation of Brca1 on overexpression results in chromosome segregation errors and Serine 988. The fact that deregulated Chk2 functioning causes tumorigenesis in mice,90,93 aberrant Aurora B localization and numerical CIN through abnormal spindle formation is intriguing, activation could very well explain the CIN observed in MAD2- as the role of Chk2 in the maintenance of genetic stability has overexpressing tumors. mainly been ascribed to its function in DNA damage checkpoint One other interesting possibility that could explain the signaling.127 increased stability of kinetochore-MTs observed in CIN cells,128 is inactivation of the tumor-suppressor gene adenomatous polyposis coli (APC), which is frequently mutated in colon carcinomas.132 Causes of merotelic attachments: increased stability of kinetochore There is ample evidence that mutation of APC is an initiating microtubule attachments. The proper control of kinetochore–MT event in colon tumorigenesis, and that the effect of APC stability is another crucial factor that determines the fidelity of inactivation on tumorigenesis is mediated through the Wnt chromosome segregation (Figure 3). Increasing the stability of MTs pathway.133 Nonetheless, APC also affects MT dynamics and APC bound to kinetochores through depletion of MCAK or Kif2b, two loss and CIN are found to coincide in early cancer lesions.134 Also, kinetochore-localized MT depolymerases, results in increased APC loss-of-function has been shown to directly result in genetic occurrence of merotelic attachments and severe chromosome 135,136 128 instability. This latter effect is thought to, at least partially, be missegregations in human tumor cells. A follow-up study in dependent on the function of APC in kinetochore–MT stability.137–139 which kinetochore-MT turnover was compared between an APC localizes to centrosomes, kinetochores and MT plus-ends,134 untransformed cell line and several CIN lines, revealed that and depletion of APC results in reduced inter-kinetochore tension, kinetochore MTs are more stable in the CIN lines. This increased which is thought to be due to decreased kinetochore–MT kinetochore MT stability could impede error correction and result 137,138 129 dynamics. Although a very interesting hypothesis, future in misattachments that can persist until anaphase. Importantly, research will have to assess if APC loss produces CIN through increasing kinetochore-MT turnover in these CIN cell lines by altered MT dynamics or through activation of Wnt signaling, as the overexpressing Kif2b or MCAK could partially revert the CIN 129 activation of Wnt target genes could also result in the induction of phenotype. CIN.140,141 More recently an unexpected link was reported between MAD2 overexpression, often found in CIN tumors, and increased kinetochore-MT stability. It was shown that MAD2 over- Causes of merotelic attachments: multiple attachment sites. The expression causes a reduction in Aurora B levels and activity at presence of extra centromeres on chromosomes could be another the centromeres, indicating that MAD2 overexpression inhibits cause of lagging chromosomes (Figure 3). As discussed above, error correction, providing a plausible explanation for the aberrantly structured chromosomes, such as ring chromosomes or increased segregation errors seen in these cells.130 The authors fusion of multiple chromosomes have a high chance of ending up showed that the effect on Aurora B localization is independent of in the cleavage furrow during telophase. These chromosomes the checkpoint function of MAD2, but the exact mechanism often have multiple centromeres and kinetochores, creating responsible for the loss of Aurora B remains to be elucidated. The multiple MT attachment sites that complicate chromosome effect on Aurora B localization and activity is in line with another biorientation. The multiple kinetochores present on a single study, which suggested that the Aurora B-dependent error chromatid can attach to opposite poles, effectively creating correction machinery works more efficiently in healthy cells ‘merotelic’ attachments142–144 (Figure 3). Upon exit from mitosis,

Oncogene (2013) 4459 – 4470 & 2013 Macmillan Publishers Limited Genetic instability in tumors A Janssen and RH Medema 4463 this bipolarly attached chromatid would lag behind, causing Besides DNA damaging events that occur in the S phase following whole-chromosome gain or loss,20,24,142,144 or initiate a BFB cycle an aberrant mitosis, it has also been shown that lagging as discussed above. These data indicate that structural chromosomes can be damaged during telophase.155 Following chromosomal aberrations and numerical CIN are closely linked both artificially induced chromosome segregation errors and and show that DNA damage due to, for example, replication spontaneous missegregations in CIN cancer cells, chromatin was defects,145,146 telomere defects24,102,144,147 or DNA-repair found to be damaged in telophase and the next G1 phase.155 The defects148 could, besides structural CIN, eventually result in occurrence of these DSBs could be (partially) rescued by inhibiting numerical CIN as well. cytokinesis, suggesting that cleavage furrow ingression causes Another cause of multiple centromere formation could be the damage to the lagging chromosomes (Figure 4). random incorporation of CENP-A at heterochromatic sites, other How ingression of the furrow could impose sufficient force to than the core centromeric region. CENP-A is a Histone H3-like damage the lagging chromatin is unclear. It could be that the protein, which specifically integrates at centromeric nucleosomes mechanical force exerted through contraction of the actin–myosin and is required for the proper formation of centromeres and ring can break the lagging chromatid. In this respect it will be kinetochores.149 It has been shown in Drosophila (cells) that interesting to see whether the furrow is actually in direct contact overexpression of CENP-A can directly result in the localization of with the lagging chromatin and whether this physical contact CENP-A into noncentromeric regions.142 This leads to ectopic coincides with the observed DNA damage. kinetochore formation, resulting in an increase in erroneous MT An interesting hypothesis is that fragile sites present on lagging attachments upon mitotic entry and ultimately severe chromosomes could render them more prone to breakage when chromosome segregation errors. Enhanced levels of CENP-A and present in the cleavage furrow. It has been shown that fragile sites its targeting factor HJURP have been found in both breast and induced by replication defects are protected from further colon cancer.150,151 However, in human cells, contradictory data destabilization and breaking in G1 through the recruitment of have been obtained on the effects of ectopic localization of CENP- 53BP1.162,163 Hypothetically, cleavage furrow ingression in the A. It has been suggested that CENP-A presence is not sufficient to presence of these underreplicated hotspots could promote drive complete de novo kinetochore formation,152 whereas other breakage of lagging chromosomes before 53BP1 has studies have shown it can.153 Thus, it is unclear if mere accumulated to protect these loci. Another cause of overexpression of CENP-A is sufficient for the induction of CIN missegregation-induced DNA damage could be the presence of through creation of multiple attachment sites. an increased, centromere-localized force imposed by MTs coming from the two spindle poles. This has also been suggested by an earlier study, in which the authors observed centromere-localized NUMERICAL CIN AND DNA DAMAGE DNA damage foci in interphase CIN cells.164 To address this point, Both structural and numerical chromosomal changes arise in the future studies should determine whether inhibition of this majority of cancer cells.6,8 As described above, it has been known centromere-localized force can reduce the damage. for several decades that structural chromosomal aberrations can Nevertheless, these recent findings154–156 reveal new induce chromosome segregation errors upon cell division.20 mechanisms by which CIN and aneuploidy promote genomic However, it has only recently been revealed that the opposite instability in cancer cells (Figure 4). The reciprocal relationship also occurs: chromosome segregation errors can induce structural between numerical and structural chromosomal changes compli- chromosomal changes through generation of DSBs.154–156 cates analysis of the direct effects of either structural chromoso- Duesberg et al.157,158 initially observed that the severity of mal changes or numerical CIN on tumor formation and reveals aneuploidy coincides with an increase in structural chromosomal additional ways in which heterogeneity arises in tumors. changes, and several CIN model systems displayed structural chromosomal aberrations as well.8,33,93,120,159 However, the underlying mechanisms for this correlation remained unknown. CHROMOSOME SEGREGATION ERRORS AND TUMORIGENESIS By generating 13 aneuploid yeast strains that each harbor an extra Boveri165 was the first to postulate that chromosomal aberrations single chromosome in addition to the haploid content, it was could be a causal factor in the occurrence of cancer after von recently shown that aneuploidy can directly induce genomic Hansemann had observed the presence of chromosomal instability156 (Figure 4). Aneuploidy of a single chromosome in abnormalities and mitotic errors in cancer cells.166 At present, some cases resulted in chromosome missegregations, whereas in the impact of structural changes on cancer progression is quite others, the abnormal chromosome number induced a high well understood,2 whereas the effects of numerical changes mutation rate. This increased genomic instability is thought to on tumorigenesis remain highly debated.46,167 Numerical be due to both an increase in DSBs and defects in recombinational chromosomal instability has been observed in early neoplastic repair.156 The underlying cause of the increase in DSBs remains lesions168 and is thought to be able to cause transformation.169–171 largely unknown, but could be due to aneuploidy-induced In addition, aneuploidy, one of the common consequences of CIN, imbalances in protein stoichiometry.160,161 Indeed, diploid yeast can drive evolution by introducing phenotypic variation in several strains carrying a single extra chromosome did not display an budding yeast strains.172,173 In line with a role for CIN in increase in genomic instability.156 In addition to defects induced tumorigenesis, CIN has been associated with poor prognosis and by aneuploidy per se, it was also recently shown that single resistance to chemotherapeutics in human patients.32–41 Below, we chromosome missegregation events, the underlying cause of summarize some of the main findings on the consequences of CIN aneuploidy induction, can also directly lead to DNA damage154,155 on tumor cells and tumor formation. For a more comprehensive (Figure 4). summary of the most recent advances in our understanding of the Micronuclei, small nuclear compartments that are surrounded consequences of CIN, we refer to a set of recently published reviews, by a nuclear envelope, in general, contain one or two chromo- which specifically summarize this aspect of CIN.46,174,175 somes that missegregated in the preceding mitosis.12,24 These Several CIN mouse models have been generated with deletions micronuclei undergo replication defects in the next S and G2 or hypomorphic alleles of genes important for mitotic fidelity phase, due to incomplete recruitment of DNA replication factors, (Table 1). Homozygous deletion of genes required for faithful such as the DNA helicase components MCM2 and MCM3 and the mitotic progression leads to embryonic lethality.53,57,58,79,171,176–179 initiation factor Cdt1.154 These replication defects result in Haploinsufficiency of mitotic genes, however, is tolerated in all persistent DNA damage in micronuclei, providing a link between cases and is therefore used extensively to investigate in the effects chromosome segregation errors and genomic instability (Figure 4). of CIN in tumorigenesis. Following recent findings that mitotic

& 2013 Macmillan Publishers Limited Oncogene (2013) 4459 – 4470 Genetic instability in tumors A Janssen and RH Medema 4464

Figure 4. Schematic representation of the proposed links between chromosomal instability (CIN), aneuploidy and genomic instability. The presence of an extra chromosome enhances protein synthesis, which can (in)directly lead to chromosome segregation errors, recombination problems and defects in DNA repair. Chromosome segregation errors, associated with CIN, could lead to DNA damage through the formation of replication-defective micronuclei (MN) or cleavage furrow-induced chromatin breakage.

genes are more frequently upregulated than downregulated in spontaneous tumor formation does not always correlate with the CIN tumors, various mouse models have been generated that level of aneuploidy found in MEFs. For example, BubR1H/H MEFs57 overexpress genes involved in mitotic progression, such as the have similar levels of aneuploidy as MEFs of Bub1 À /H and Bub1H/H mitotic checkpoint genes Bub1180 and MAD293 and the E2 enzyme animals,178 but BubR1H/H animals do not display an increased UbcH10181 (Table 1). Below we discuss the insights that these tumor incidence, whereas Bub1 hypomorphic animals do. These various models have generated with respect to the contribution of discrepancies could be due to additional roles of these checkpoint CIN in tumor formation. proteins in other cellular processes. Indeed, BubR1 hypomorphic animals also develop severe ageing-associated phenotypes, indicating that BubR1 has additive functions in maintaining cell Tumor-promoting role of numerical CIN homeostasis, possibly through regulation of p16 and p19 protein Haploinsufficiency, hypomorphy or overexpression of genes levels.57,184 One striking similarity in the spectrum of involved in mitotic progression can result in CIN. Although spontaneously developing tumors in the various CIN models is aneuploidy levels vary between different mouse models,167 CIN the formation of lung tumors. This resemblance indicates that, for generally seems to be able to enhance tumorigenesis. Treatment unknown reasons, CIN specifically enhances tumorigenesis in lung with carcinogens, such as DMBA, leads to increased tumor epithelial cells. incidence in 6 out of 10 CIN models tested (see Table 1) and One single chromosome missegregation event results in the crossing some of the CIN models with tumor-prone mouse loss or gain of one complete chromosome and thereby affects a models, such as Em-myc93,180 or APCmin/ þ 182,183 mice, results in large number of genes. The fact that CIN tumor cells continuously enhanced or accelerated tumor formation (Table 1). Many CIN change their chromosome composition indicates that CIN causes mouse models also display an increased susceptibility to extensive phenotypic changes, as signified by the increased spontaneous tumor formation in lung, liver and lymph nodes, overall transcriptomic and proteomic activity in aneuploid yeast albeit at a relatively old age (Table 1). Interestingly, the extent of strains.160 These dramatic changes in genetic make-up make it

Oncogene (2013) 4459 – 4470 & 2013 Macmillan Publishers Limited Genetic instability in tumors A Janssen and RH Medema 4465 Table 1. Mouse models of numerical CIN

Mouse model Spontaneous (organ) Carcinogen induced Crossed with Tumorigenesis in References tumors (organ) compound mice (organ)

MAD2 þ / À Yes (lung)  p53 þ / À Increase 51,193 p53 þ / À /MAD1 þ / À Increase (lung, spleen, breast) MAD1 þ / À Yes (lung) Increase, vincristine p53 þ / À Normal 58,193 (lung, liver) p53 þ / À /MAD2 þ / À Increase (lung, spleen, breast) Bub3 þ / À No Normal, DMBA p53 þ / À Normal 55,60,177 Rb þ / À Normal Rae1 þ / À Increase, DMBA (lung) BubR1 þ / À No Increase, APCmin/ þ Increase (colon) 56,183 azoxymethane (lung, Decrease (small intestine) colon) BubR1 þ /H No  57 BubR1H/H No Increase, DMBA (lung)  57 Bub1 þ / À No Increase, DMBA (lung) p53 À / À Normal 178,182,188 APCmin/ þ Increase (colon) Bub1H/H Yes (liver, lung, lymph  p53 þ / À Increase (thymus) 178,182,188,194 nodes) p53 À / À Increase (thymus) Bub1 À /H Yes (liver, lung, lymph  p53 þ / À Increase (thymus) 178,182,188 nodes) p53 À / À Increase (thymus) APCmin/ þ Increase (colon) Rb þ / À Increase (pituitary) PTEN þ / À Decrease (prostate lesions) MAD2 ox Yes (liver, lung, lymph  Em-myc Increase (lymph nodes) 93,195 nodes, intestine) KRASG12D Increased relapse (lung) Bub1T85/T264 Yes (liver, lymph  Em-myc Increase (lymph nodes) 180 nodes and skin) CENP-E þ / À Yes (lung, colon, Decrease, DMBA p19 þ / À Decrease (lymph nodes) 171 spleen) (lung) Decrease (liver) Cdh1 þ / À Yes (breast, lung)  196 Hec1 ox Yes (lung, liver)  197 Cdc20 þ / À /H/H/ À /H No Normal, DMBA  198 Cdc20AAA/ þ Yes (lymph nodes,  p53 À / À Increase (thymus) 188,199 liver) ATM À / À Increase (thymus) UbcH10T1/T2 Yes (lung, liver, blood, Increase, DMBA (lung)  181 skin) Pttg1 À / À No  Rb þ / À Decrease (pituitary) 200,201 SA1 þ / À Yes (liver, pancreas, Decrease, 3-methyl- p53 À / À Normal 102 blood) colanthrene (fibrous PTEN þ / À Normal tissue) Decrease, diethyl-nitrosamine (liver) Incenp þ / À No  202 Survivin þ / À No  202 difficult to determine how CIN could be exactly contributing to in turn could allow the induction of other transforming changes, tumorigenesis. which ultimately lead to tumorigenesis. One, rather simple, hypothesis is that CIN is able to promote Alternatively, CIN and aneuploidy could promote tumorigen- tumorigenesis by inducing loss of certain tumor-suppressor genes esis through the creation of a mutator phenotype in cells.42,187 or gain of oncogenes. In line with this, crossing a CIN mouse Aneuploidy could affect levels of DNA repair genes, and thereby model carrying a hypomorphic Bub1 allele with tumor-prone promote genomic instability. In line with this hypothesis, mouse models heterozygous for p53 or APCmin resulted in loss of aneuploidy was initially found to correlate with genomic heterozygosity of these tumor-suppressor genes and enhanced instability in cancer cells158 and has recently been shown to tumor formation.182 CIN could indeed be quite effective in induce genomic instability in budding yeast.156 Moreover, CIN inducing loss of heterozygosity, as it will immediately induce could itself be a mutator by inducing DNA damage and loss of multiple genes. structural chromosomal changes through micronuclei However, since aneuploidy can lead to loss or gain of multiple formation154 or cleavage furrow ingression in the presence of essential genes, it is very likely to be detrimental to cells.52,54,160,161 lagging chromosomes.155 Interestingly, crossing a CIN mouse It is therefore thought that coinciding mutations would have to model with a mouse model deficient for ATM resulted in arise in other pathways to render the cell insensitive to changes in accelerated tumor formation,188 consistent with the notion that chromosome number.46 Examples of such changes are loss of CIN-induced tumor formation involves the induction of DNA function of p53185 or mutations that promote proteasomal damage. degradation in order to circumvent the increases in protein In summary, CIN and aneuploidy could together create a state production obtained by gain of chromosomes.186 These mutations in which cancer cells not only continuously lose and gain whole

& 2013 Macmillan Publishers Limited Oncogene (2013) 4459 – 4470 Genetic instability in tumors A Janssen and RH Medema 4466 chromosomes, but also acquire new mutations and translocations, 2 Mitelman F, Johansson B, Mertens F. The impact of translocations and gene which interfere with the function of genes involved in DNA repair, fusions on cancer causation. Nat Rev Cancer 2007; 7: 233–245. mitotic fidelity and cell cycle progression, thereby promoting 3 Nowell PC. The minute chromosome (Phl) in chronic granulocytic leukemia. Blut transformation and drug resistance. 1962; 8: 65–66. 4 Look AT. Oncogenic transcription factors in the human acute leukemias. Science 1997; 278: 1059–1064. Tumor-suppressive role of numerical CIN 5 Nambiar M, Kari V, Raghavan SC. Chromosomal translocations in cancer. Biochim Although CIN correlates in most models with increased tumor Biophys Acta 2008; 1786: 139–1352. formation, several cases of CIN-associated tumor suppression have 6 Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J et al. The also been described (Table 1). In particular, animals heterozygous landscape of somatic copy-number alteration across human cancers. Nature for the motor protein CENP-E showed a significant decrease in 2010; 463: 899–905. spontaneous liver tumor formation, decreased tumor incidence 7 Castro MA, Onsten TG, Moreira JC, de Almeida RM. Chromosome aberrations in solid tumors have a stochastic nature. Mutat Res 2006; 600: 150–164. in a p19 heterozygous background and a clear reduction in 171 8 Mitelman F, Johansson B, Mertens F. Mitelman Database of Chromosome carcinogen-induced tumor formation. Similarly, BubR1 Aberrations and Gene Fusions in Cancer. http://cgap.nci.nih.gov/Chromosomes/ heterozygosity decreased intestinal tumor incidence in the Mitelman 2012. min/ þ 183,189 tumor-prone APC model and Bub1 hypomorphy 9 Roschke AV, Tonon G, Gehlhaus KS, McTyre N, Bussey KJ, Lababidi S et al. reduced the number of prostate lesions when crossed with Karyotypic complexity of the NCI-60 drug-screening panel. Cancer Res 2003; 63: heterozygous PTEN animals.182 In line with this, inverse 8634–8647. correlations between CIN and patient prognosis in various tumor 10 Abdel-Rahman WM, Katsura K, Rens W, Gorman PA, Sheer D, Bicknell D et al. types have also been described.190,191 This suggests that an Spectral karyotyping suggests additional subsets of colorectal cancers char- optimal level of CIN exists, which, dependent on the tissue, results acterized by pattern of chromosome rearrangement. Proc Natl Acad Sci USA 2001; 98: 2538–2543. in either tumorigenesis or tumor suppression. Excessive CIN could 11 Davidson JM, Gorringe KL, Chin SF, Orsetti B, Besret C, Courtay-Cahen C et al. compromise genome stability in such a way that it is incompatible Molecular cytogenetic analysis of breast cancer cell lines. Br J Cancer 2000; 83: with cell viability. Mild CIN could promote growth by providing 1309–1317. the tumor cell with a level of genetic instability that is moderate 12 Thompson SL, Compton DA. Chromosome missegregation in human cells arises enough to maintain a relatively stable genome, while at the same through specific types of kinetochore-microtubule attachment errors. Proc Natl time providing enough genetic variation in the population to Acad Sci USA 2011; 108: 17974–17978. allow for rapid adaptation to changes in the tumor environment. If 13 Nishizaki T, Chew K, Chu L, Isola J, Kallioniemi A, Weidner N et al. Genetic a maximal tolerated level of CIN does exist for tumors, this feature alterations in lobular breast cancer by comparative genomic hybridization. Int J could be exploited in future anticancer strategies.192 This Cancer 1997; 74: 513–517. 14 Nishizaki T, DeVries S, Chew K, Goodson 3rd WH, Ljung BM, Thor A et al. Genetic paradoxical relationship between CIN, tumorigenesis and alterations in primary breast cancers and their metastases: direct comparison cell viability is a highly debated topic and future research will using modified comparative genomic hybridization. Genes Chromosomes Cancer hopefully lead to more insight on the role of CIN in tumorigenesis, 1997; 19: 267–272. but also allow for the successful development of new anticancer 15 Natarajan AT, Palitti F. DNA repair and chromosomal alterations. Mutat Res 2008; strategies specifically targeting CIN or aneuploid tumor cells. 657: 3–7. 16 Obe G, Durante M. DNA double strand breaks and chromosomal aberrations. Cytogenet Genome Res 2010; 128: 8–16. CONCLUDING REMARKS 17 Obe G, Pfeiffer P, Savage JR, Johannes C, Goedecke W, Jeppesen P et al. Chro- mosomal aberrations: formation, identification and distribution. Mutat Res 2002; The question remains whether CIN is an initiator of tumorigenesis, 504: 17–36. whether it mainly provides an enhancing effect to promote 18 Duker NJ. Chromosome breakage syndromes and cancer. Am J Med Genet. transformation or if it is a mere consequence of tumor formation. [Review] 2002; 115: 125–129. The fact that CIN is a common trait of 470% of all tumors makes it 19 Bender MA, Griggs HG, Bedford JS. Mechanisms of chromosomal aberration unlikely to simply be a bystander to tumorigenesis. Whole production. 3. Chemicals and ionizing radiation. Mutat Res. [Review] 1974; 23: genome sequencing of early neoplasia in either mouse models 197–212. or human patients in combination with assessment of CIN status 20 McClintock B. The production of homozygous deficient tissues with mutant could provide more insight on the role of CIN in human tumor characteristics by means of the aberrant mitotic behavior of ring-shaped chro- mosomes. Genetics 1938; 23: 315–376. formation. Generation of CIN mouse models in which CIN can be 21 McClintock B. The stability of broken ends of chromosomes in Zea mays. Genetics induced during later stages of development, will hopefully also 1941; 26: 234–282. provide more knowledge on the effect of CIN in tumorigenesis 22 Hastie ND, Allshire RC. Human telomeres: fusion and interstitial sites. Trends and tumor suppression. In most of the current CIN mouse models, Genet. [Review] 1989; 5: 326–331. CIN is induced already during embryonic development, which 23 Gisselsson D, Jonson T, Petersen A, Strombeck B, Dal Cin P, Hoglund M et al. makes it difficult to draw conclusions about tumor development Telomere dysfunction triggers extensive DNA fragmentation and evolution of in the adult life. In addition, improving diagnostic tools to visualize complex chromosome abnormalities in human malignant tumors. Proc Natl Acad CIN in tissues of mouse models, but also human patients, should Sci USA 2001; 98: 12683–12688. help in evaluating the various levels of CIN and the impact thereof 24 Gisselsson D, Bjork J, Hoglund M, Mertens F, Dal Cin P, Akerman M et al. on tumorigenesis.174 These tools should help to solve the Abnormal nuclear shape in solid tumors reflects mitotic instability. Am J Pathol 2001; 158: 199–206. discrepancies currently found between different mouse models 25 Gisselsson D, Pettersson L, Hoglund M, Heidenblad M, Gorunova L, Wiegant J et of CIN and improve our understanding of the role of CIN in human al. Chromosomal breakage-fusion-bridge events cause genetic intratumor het- tumorigenesis. erogeneity. Proc Natl Acad Sci USA 2000; 97: 5357–5362. 26 Heim S, Mitelman F. Numerical chromosome aberrations in human neoplasia. Cancer Genet Cytogenet 1986; 22: 99–108. CONFLICT OF INTEREST 27 Baudis M. Genomic imbalances in 5918 malignant epithelial tumors: an explorative meta-analysis of chromosomal CGH data. BMC cancer 2007; 7: 226. The authors declare no conflict of interest. 28 Rehen SK, Yung YC, McCreight MP, Kaushal D, Yang AH, Almeida BS et al. Constitutional aneuploidy in the normal human brain. J Neurosci 2005; 25: 2176–2180. REFERENCES 29 Duncan AW, Taylor MH, Hickey RD, Hanlon Newell AE, Lenzi ML, Olson SB et al. 1 Mitelman F, Johansson B, Mandahl N, Mertens F. Clinical significance of cyto- The ploidy conveyor of mature hepatocytes as a source of genetic variation. genetic findings in solid tumors. Cancer Genet Cytogenet 1997; 95: 1–8. Nature 2010; 467: 707–710.

Oncogene (2013) 4459 – 4470 & 2013 Macmillan Publishers Limited Genetic instability in tumors A Janssen and RH Medema 4467 30 Lengauer C, Kinzler KW, Vogelstein B. Genetic instability in colorectal cancers. 57 Baker DJ, Jeganathan KB, Cameron JD, Thompson M, Juneja S, Kopecka A et al. Nature 1997; 386: 623–627. BubR1 insufficiency causes early onset of aging-associated phenotypes and 31 Lengauer C, Kinzler KW, Vogelstein B. Genetic instabilities in human cancers. infertility in mice. Nat Genet 2004; 36: 744–749. Nature 1998; 396: 643–649. 58 Iwanaga Y, Chi YH, Miyazato A, Sheleg S, Haller K, Peloponese Jr. JM et al. 32 Choi CM, Seo KW, Jang SJ, Oh YM, Shim TS, Kim WS et al. Chromosomal Heterozygous deletion of mitotic arrest-deficient protein 1 (MAD1) increases the instability is a risk factor for poor prognosis of adenocarcinoma of the lung: incidence of tumors in mice. Cancer Res 2007; 67: 160–166. Fluorescence in situ hybridization analysis of paraffin-embedded tissue from 59 Janssen A, Kops GJ, Medema RH. Elevating the frequency of chromosome mis- Korean patients. Lung Cancer 2009; 64: 66–70. segregation as a strategy to kill tumor cells. Proc Natl Acad Sci USA. 2009; 106: 33 Carter SL, Eklund AC, Kohane IS, Harris LN, Szallasi Z. A signature of chromosomal 19108–19113. instability inferred from gene expression profiles predicts clinical outcome in 60 Kalitsis P, Fowler KJ, Griffiths B, Earle E, Chow CW, Jamsen K et al. Increased multiple human cancers. Nat Genet 2006; 38: 1043–1048. chromosome instability but not cancer predisposition in haploinsufficient Bub3 34 Walther A, Houlston R, Tomlinson I. Association between chromosomal mice. Genes Chromosomes Cancer 2005; 44: 29–36. instability and prognosis in colorectal cancer: a meta-analysis. Gut 2008; 57: 61 Cahill DP, Lengauer C, Yu J, Riggins GJ, Willson JK, Markowitz SD et al. Mutations 941–950. of mitotic checkpoint genes in human cancers. Nature 1998; 392: 300–303. 35 Bakhoum SF, Danilova OV, Kaur P, Levy NB, Compton DA. Chromosomal 62 Tighe A, Johnson VL, Albertella M, Taylor SS. Aneuploid colon cancer cells have a instability substantiates poor prognosis in patients with diffuse large B-cell robust spindle checkpoint. EMBO Rep 2001; 2: 609–614. lymphoma. Clin Cancer Res 2011; 17: 7704–7711. 63 Hanks S, Coleman K, Reid S, Plaja A, Firth H, Fitzpatrick D et al. Constitutional 36 Kuukasjarvi T, Karhu R, Tanner M, Kahkonen M, Schaffer A, Nupponen N et al. aneuploidy and cancer predisposition caused by biallelic mutations in BUB1B. Genetic heterogeneity and clonal evolution underlying development of Nat Genet 2004; 36: 1159–1161. asynchronous metastasis in human breast cancer. Cancer Res 1997; 57: 64 Matsuura S, Matsumoto Y, Morishima K, Izumi H, Matsumoto H, Ito E et al. 1597–1604. Monoallelic BUB1B mutations and defective mitotic-spindle checkpoint in seven 37 Gao C, Furge K, Koeman J, Dykema K, Su Y, Cutler ML et al. Chromosome families with premature chromatid separation (PCS) syndrome. Am J Med Genet instability, chromosome transcriptome, and clonal evolution of tumor cell A 2006; 140: 358–367. populations. Proc Natl Acad Sci USA 2007; 104: 8995–9000. 65 Wang X, Jin DY, Ng RW, Feng H, Wong YC, Cheung AL et al. Significance of MAD2 38 Sheffer M, Bacolod MD, Zuk O, Giardina SF, Pincas H, Barany F et al. Association expression to mitotic checkpoint control in ovarian cancer cells. Cancer Res 2002; of survival and disease progression with chromosomal instability: a genomic 62: 1662–1668. exploration of colorectal cancer. Proc Natl Acad Sci USA 2009; 106: 7131–7136. 66 Wang X, Jin DY, Wong YC, Cheung AL, Chun AC, Lo AK et al. 39 McClelland SE, Burrell RA, Swanton C. Chromosomal instability: a composite Correlation of defective mitotic checkpoint with aberrantly reduced expression phenotype that influences sensitivity to chemotherapy. Cell Cycle 2009; 8: of MAD2 protein in nasopharyngeal carcinoma cells. Carcinogenesis 2000; 21: 3262–3266. 2293–2297. 40 Lee AJ, Endesfelder D, Rowan AJ, Walther A, Birkbak NJ, Futreal PA et al. Chro- 67 Kasai T, Iwanaga Y, Iha H, Jeang KT. Prevalent loss of mitotic spindle checkpoint mosomal instability confers intrinsic multidrug resistance. Cancer Res 2011; 71: in adult T-cell leukemia confers resistance to microtubule inhibitors. J Biol Chem 1858–1870. 2002; 277: 5187–5193. 41 Swanton C, Nicke B, Schuett M, Eklund AC, Ng C, Li Q et al. Chromosomal 68 Yoon DS, Wersto RP, Zhou W, Chrest FJ, Garrett ES, Kwon TK et al. Variable levels instability determines taxane response. Proc Natl Acad Sci USA 2009; 106: of chromosomal instability and mitotic spindle checkpoint defects in breast 8671–8676. cancer. Am J Pathol 2002; 161: 391–397. 42 Kolodner RD, Cleveland DW, Putnam CD. Cancer. aneuploidy drives a mutator 69 Li Y, Benezra R. Identification of a human mitotic checkpoint gene: hsMAD2. phenotype in cancer. Science 2011; 333: 942–943. Science 1996; 274: 246–248. 43 Weaver BA, Cleveland DW. Does aneuploidy cause cancer? Curr Opin Cell Biol 70 Minhas KM, Singh B, Jiang WW, Sidransky D, Califano JA. Spindle assembly 2006; 18: 658–667. checkpoint defects and chromosomal instability in head and neck squamous cell 44 Duesberg P, Stindl R, Hehlmann R. Explaining the high mutation rates of cancer carcinoma. Int J Cancer 2003; 107: 46–52. cells to drug and multidrug resistance by chromosome reassortments that are 71 Saeki A, Tamura S, Ito N, Kiso S, Matsuda Y, Yabuuchi I et al. Frequent impairment catalyzed by aneuploidy. Proc Natl Acad Sci USA 2000; 97: 14295–14300. of the spindle assembly checkpoint in hepatocellular carcinoma. Cancer 2002; 45 Duesberg P, Li R, Sachs R, Fabarius A, Upender MB, Hehlmann R. Cancer drug 94: 2047–2054. resistance: the central role of the karyotype. Drug Resist Updat 2007; 10: 51–58. 72 Sze KM, Ching YP, Jin DY, Ng IO. Association of MAD2 expression with 46 Pfau SJ, Amon A. Chromosomal instability and aneuploidy in cancer: from yeast mitotic checkpoint competence in hepatoma cells. J Biomed Sci 2004; 11: to man. EMBO Rep 2012; 13: 515–527. 920–927. 47 Nowak MA, Komarova NL, Sengupta A, Jallepalli PV, Shih IeM, Vogelstein B et al. 73 Takahashi T, Haruki N, Nomoto S, Masuda A, Saji S, Osada H et al. Identification of The role of chromosomal instability in tumor initiation. Proc Natl Acad Sci USA frequent impairment of the mitotic checkpoint and molecular analysis of the 2002; 99: 16226–16231. mitotic checkpoint genes, hsMAD2 and p55CDC, in human lung cancers. 48 Rajagopalan H, Nowak MA, Vogelstein B, Lengauer C. The significance of Oncogene 1999; 18: 4295–4300. unstable chromosomes in colorectal cancer. Nat Rev Cancer 2003; 3: 695–701. 74 Weitzel DH, Vandre DD. Differential spindle assembly checkpoint response in 49 Musacchio A, Salmon ED. The spindle-assembly checkpoint in space and time. human lung adenocarcinoma cells. Cell Tissue Res 2000; 300: 57–65. Nat Rev Mol Cell Biol 2007; 8: 379–393. 75 Hempen PM, Kurpad H, Calhoun ES, Abraham S, Kern SE. A double missense 50 Kops GJ, Weaver BA, Cleveland DW. On the road to cancer: aneuploidy and the variation of the BUB1 gene and a defective mitotic spindle checkpoint in the mitotic checkpoint. Nat Rev Cancer 2005; 5: 773–785. pancreatic cancer cell line Hs766T. Hum Mutat 2003; 21:445. 51 Michel LS, Liberal V, Chatterjee A, Kirchwegger R, Pasche B, Gerald W et al. MAD2 76 Ouyang B, Knauf JA, Ain K, Nacev B, Fagin JA. Mechanisms of aneuploidy in haplo-insufficiency causes premature anaphase and chromosome instability in thyroid cancer cell lines and tissues: evidence for mitotic checkpoint mammalian cells. Nature 2001; 409: 355–359. dysfunction without mutations in BUB1 and BUBR1. Clin Endocrinol 2002; 56: 52 Kops GJ, Foltz DR, Cleveland DW. Lethality to human cancer cells through 341–350. massive chromosome loss by inhibition of the mitotic checkpoint. Proc Natl Acad 77 Suijkerbuijk SJ, van Osch MH, Bos FL, Hanks S, Rahman N, Kops GJ. Molecular Sci USA 2004; 101: 8699–8704. causes for BUBR1 dysfunction in the human cancer predisposition syndrome 53 Kalitsis P, Earle E, Fowler KJ, Choo KH. Bub3 gene disruption in mice reveals mosaic variegated aneuploidy. Cancer Res 2010; 70: 4891–4900. essential mitotic spindle checkpoint function during early embryogenesis. Genes 78 Matsuura S, Ito E, Tauchi H, Komatsu K, Ikeuchi T, Kajii T. Chromosomal instability Dev 2000; 14: 2277–2282. syndrome of total premature chromatid separation with mosaic variegated 54 Michel L, Diaz-Rodriguez E, Narayan G, Hernando E, Murty VV, Benezra R. aneuploidy is defective in mitotic-spindle checkpoint. Am J Hum Genet 2000; 67: Complete loss of the tumor suppressor MAD2 causes premature cyclin B 483–486. degradation and mitotic failure in human somatic cells. Proc Natl Acad Sci USA. 79 Wang Q, Liu T, Fang Y, Xie S, Huang X, Mahmood R et al. BUBR1 deficiency 2004; 101: 4459–4464. results in abnormal megakaryopoiesis. Blood 2004; 103: 1278–1285. 55 Babu JR, Jeganathan KB, Baker DJ, Wu X, Kang-Decker N, van Deursen JM. Rae1 is 80 Cahill DP, da Costa LT, Carson-Walter EB, Kinzler KW, Vogelstein B, Lengauer C. an essential mitotic checkpoint regulator that cooperates with Bub3 to prevent Characterization of MAD2B and other mitotic spindle checkpoint genes. Geno- chromosome missegregation. J Cell Biol 2003; 160: 341–353. mics 1999; 58: 181–187. 56 Dai W, Wang Q, Liu T, Swamy M, Fang Y, Xie S et al. Slippage of mitotic arrest and 81 Myrie KA, Percy MJ, Azim JN, Neeley CK, Petty EM. Mutation and expression enhanced tumor development in mice with BubR1 haploinsufficiency. Cancer analysis of human BUB1 and BUB1B in aneuploid breast cancer cell lines. Cancer Res 2004; 64: 440–445. Lett 2000; 152: 193–199.

& 2013 Macmillan Publishers Limited Oncogene (2013) 4459 – 4470 Genetic instability in tumors A Janssen and RH Medema 4468 82 Haruki N, Saito H, Harano T, Nomoto S, Takahashi T, Osada H et al. Molecular correction is achieved by two different mechanisms. J Cell Sci 2003; 116(Pt 20): analysis of the mitotic checkpoint genes BUB1, BUBR1 and BUB3 in human lung 4213–4225. cancers. Cancer Lett 2001; 162: 201–205. 108 Cimini D, Cameron LA, Salmon ED. Anaphase spindle mechanics prevent 83 Yuan B, Xu Y, Woo JH, Wang Y, Bae YK, Yoon DS et al. Increased expression of mis-segregation of merotelically oriented chromosomes. Curr Biol 2004; 14: mitotic checkpoint genes in breast cancer cells with chromosomal instability. 2149–2155. Clin Cancer Res 2006; 12: 405–410. 109 van der Waal MS, Hengeveld RC, van der Horst A, Lens SM. Cell 84 Grabsch H, Takeno S, Parsons WJ, Pomjanski N, Boecking A, Gabbert HE et al. division control by the chromosomal passenger complex. Exp Cell Res 2012; 318: Overexpression of the mitotic checkpoint genes BUB1, BUBR1, and BUB3 in 1407–1420. gastric cancer—association with tumour cell proliferation. J Pathol 2003; 200: 110 Heneen WK. Kinetochores and microtubules in multipolar mitosis and chro- 16–22. mosome orientation. Exp Cell Res 1975; 91: 57–62. 85 Daniel J, Coulter J, Woo JH, Wilsbach K, Gabrielson E. High levels of the Mps1 111 Sluder G, Thompson EA, Miller FJ, Hayes J, Rieder CL. The checkpoint control for checkpoint protein are protective of aneuploidy in breast cancer cells. Proc Natl anaphase onset does not monitor excess numbers of spindle poles or bipolar Acad Sci USA 2011; 108: 5384–5389. spindle symmetry. J Cell Sci 1997; 110(Pt 4): 421–429. 86 Tanaka K, Nishioka J, Kato K, Nakamura A, Mouri T, Miki C et al. Mitotic check- 112 Lingle WL, Barrett SL, Negron VC, D’Assoro AB, Boeneman K, Liu W et al. Cen- point protein hsMAD2 as a marker predicting liver metastasis of human gastric trosome amplification drives chromosomal instability in breast tumor develop- cancers. Jpn J Cancer Res 2001; 92: 952–958. ment. Proc Natl Acad Sci USA. 2002; 99: 1978–1983. 87 Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A et al. Distinct 113 Sato N, Mizumoto K, Nakamura M, Maehara N, Minamishima YA, Nishio S et al. types of diffuse large B-cell lymphoma identified by gene expression profiling. Correlation between centrosome abnormalities and chromosomal Nature 2000; 403: 503–511. instability in human pancreatic cancer cells. Cancer Genet Cytogenet 2001; 126: 88 Garber ME, Troyanskaya OG, Schluens K, Petersen S, Thaesler Z, Pacyna-Gen- 13–19. gelbach M et al. Diversity of gene expression in adenocarcinoma of the lung. 114 Pihan GA, Purohit A, Wallace J, Malhotra R, Liotta L, Doxsey SJ. Centrosome Proc Natl Acad Sci USA 2001; 98: 13784–13789. defects can account for cellular and genetic changes that characterize prostate 89 Ryan SD, Britigan EM, Zasadil LM, Witte K, Audhya A, Roopra A et al. Up-reg- cancer progression. Cancer Res 2001; 61: 2212–2219. ulation of the mitotic checkpoint component Mad1 causes chromosomal 115 Ghadimi BM, Sackett DL, Difilippantonio MJ, Schrock E, Neumann T, instability and resistance to microtubule poisons. Proc Natl Acad Sci USA. 2012; Jauho A et al. Centrosome amplification and instability occurs exclusively in 109: E2205–E2214. aneuploid, but not in diploid colorectal cancer cell lines, and correlates with 90 Hernando E, Nahle Z, Juan G, Diaz-Rodriguez E, Alaminos M, Hemann M et al. Rb numerical chromosomal aberrations. Genes Chromosomes Cancer 2000; 27: inactivation promotes genomic instability by uncoupling cell cycle progression 183–190. from mitotic control. Nature 2004; 430: 797–802. 116 Nigg EA. Centrosome aberrations: cause or consequence of cancer progression? 91 Manning AL, Dyson NJ. pRB a tumor suppressor with a stabilizing presence. Nat Rev Cancer 2002; 2: 815–825. Trends Cell Biol 2011; 21: 433–441. 117 Shi Q, King RW. Chromosome nondisjunction yields tetraploid rather than 92 Manning AL, Longworth MS, Dyson NJ. Loss of pRB causes centromere dys- aneuploid cells in human cell lines. Nature 2005; 437: 1038–1042. function and chromosomal instability. Genes Dev 2010; 24: 1364–1376. 118 Silkworth WT, Nardi IK, Scholl LM, Cimini D. Multipolar spindle pole coalescence 93 Sotillo R, Hernando E, Diaz-Rodriguez E, Teruya-Feldstein J, Cordon-Cardo C, is a major source of kinetochore mis-attachment and chromosome mis-segre- Lowe SW et al. Mad2 overexpression promotes aneuploidy and tumorigenesis in gation in cancer cells. PLoS One 2009; 4: e6564. mice. Cancer Cell 2007; 11: 9–23. 119 Ganem NJ, Godinho SA, Pellman D. A mechanism linking extra centrosomes to 94 Schvartzman JM, Duijf PH, Sotillo R, Coker C, Benezra R. Mad2 is a critical chromosomal instability. Nature 2009; 460: 278–282. mediator of the chromosome instability observed upon Rb and p53 pathway 120 Fujiwara T, Bandi M, Nitta M, Ivanova EV, Bronson RT, Pellman D. Cytokinesis inhibition. Cancer Cell 2011; 19: 701–714. failure generating tetraploids promotes tumorigenesis in p53-null cells. Nature 95 Iwaizumi M, Shinmura K, Mori H, Yamada H, Suzuki M, Kitayama Y et al. Human 2005; 437: 1043–1047. Sgo1 downregulation leads to chromosomal instability in colorectal cancer. Gut 121 Davoli T, de Lange T. Telomere-driven tetraploidization occurs in human cells 2009; 58: 249–260. undergoing crisis and promotes transformation of mouse cells. Cancer Cell 2012; 96 Barber TD, McManus K, Yuen KW, Reis M, Parmigiani G, Shen D et al. Chromatid 21: 765–776. cohesion defects may underlie chromosome instability in human colorectal 122 Overholtzer M, Mailleux AA, Mouneimne G, Normand G, Schnitt SJ, King RW et al. cancers. Proc Natl Acad Sci USA. 2008; 105: 3443–3448. A nonapoptotic cell death process, entosis, that occurs by cell-in-cell invasion. 97JallepalliPV,WaizeneggerIC,BunzF,LangerS,SpeicherMR,PetersJMet al. Securin is Cell 2007; 131: 966–979. required for chromosomal stability in human cells. Cell 2001; 105: 445–457. 123 Overholtzer M, Brugge JS. The cell biology of cell-in-cell structures. Nat Rev Mol 98 Wang Z, Cummins JM, Shen D, Cahill DP, Jallepalli PV, Wang TL et al. Three Cell Biol 2008; 9: 796–809. classes of genes mutated in colorectal cancers with chromosomal instability. 124 Kracjovic M, Johnson NB, Sun Q, Normand G, Hoover N, Yao E et al. A non- Cancer Res 2004; 64: 2998–3001. genetic route to aneuploidy in human cancers. Nature Cell Biol 2010. 99 Solomon DA, Kim T, Diaz-Martinez LA, Fair J, Elkahloun AG, Harris BT et al. 125 Janssen A, Medema RH. Entosis: aneuploidy by invasion. Nat Cell Biol 2011; 13: Mutational inactivation of STAG2 causes aneuploidy in human cancer. Science 199–201. 2011; 333: 1039–1043. 126 Stolz A, Ertych N, Kienitz A, Vogel C, Schneider V, Fritz B et al. The CHK2-BRCA1 100 Zhang N, Ge G, Meyer R, Sethi S, Basu D, Pradhan S et al. Overexpression of tumour suppressor pathway ensures chromosomal stability in human somatic Separase induces aneuploidy and mammary tumorigenesis. Proc Natl Acad Sci cells. Nat Cell Biol 2010; 12: 492–499. USA 2008; 105: 13033–13038. 127 Bartek J, J Lukas. Chk1and Chk2 kinases in checkpoint control and cancer. Cancer 101 Nasmyth K, Haering CH. Cohesin: its roles and mechanisms. Annu Rev Genet. Cell 2003; 3: 421–429. [Review] 2009; 43: 525–558. 128 Bakhoum SF, Genovese G, Compton DA. Deviant kinetochore microtubule 102 Remeseiro S, Cuadrado A, Carretero M, Martinez P, Drosopoulos WC, Canamero dynamics underlie chromosomal instability. Curr Biol 2009; 19: 1937–1942. M et al. Cohesin-SA1 deficiency drives aneuploidy and tumourigenesis in mice 129 Bakhoum SF, Thompson SL, Manning AL, Compton DA. Genome stability is due to impaired replication of telomeres. EMBO J 2012; 31: 2076–2089. ensured by temporal control of kinetochore-microtubule dynamics. Nat Cell Biol 103 Thompson SL, Compton DA. Examining the link between chromosomal 2009; 11: 27–35. instability and aneuploidy in human cells. J Cell Biol 2008; 180: 665–672. 130 Kabeche L, Compton DA. Checkpoint-independent stabilization of kinetochore- 104 Gascoigne KE, Taylor SS. Cancer cells display profound intra- and interline microtubule attachments by Mad2 in human cells. Curr Biol 2012; 22: variation following prolonged exposure to antimitotic drugs. Cancer Cell 2008; 638–644. 14: 111–122. 131 Salimian KJ, Ballister ER, Smoak EM, Wood S, Panchenko T, Lampson MA et al. 105 Cimini D, Fioravanti D, Salmon ED, Degrassi F. Merotelic kinetochore orientation Feedback control in sensing chromosome biorientation by the Aurora B kinase. versus chromosome mono-orientation in the origin of lagging chromosomes in Curr Biol 2011; 21: 1158–1165. human primary cells. J Cell Sci 2002; 115(Pt 3): 507–515. 132 Kinzler KW, Nilbert MC, Su LK, Vogelstein B, Bryan TM, Levy DB et al. Identifi- 106 Cimini D, Howell B, Maddox P, Khodjakov A, Degrassi F, Salmon ED. Merotelic cation of FAP locus genes from chromosome 5q21. Science 1991; 253: 661–665. kinetochore orientation is a major mechanism of aneuploidy in mitotic mam- 133 Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. [Review] malian tissue cells. J Cell Biol 2001; 153: 517–527. 2006; 127: 469–480. 107 Cimini D, Moree B, Canman JC, Salmon ED. Merotelic kinetochore orientation 134 Rusan NM, Peifer M. Original CIN: reviewing roles for APC in chromosome occurs frequently during early mitosis in mammalian tissue cells and error instability. J Cell Biol 2008; 181: 719–726.

Oncogene (2013) 4459 – 4470 & 2013 Macmillan Publishers Limited Genetic instability in tumors A Janssen and RH Medema 4469 135 Kaplan KB, Burds AA, Swedlow JR, Bekir SS, Sorger PK, Nathke IS. A role for the 161 Williams BR, Prabhu VR, Hunter KE, Glazier CM, Whittaker CA, Housman DE et al. Adenomatous Polyposis Coli protein in chromosome segregation. Nat Cell Biol Aneuploidy affects proliferation and spontaneous immortalization in mamma- 2001; 3: 429–432. lian cells. Science 2008; 322: 703–709. 136 Fodde R, Kuipers J, Rosenberg C, Smits R, Kielman M, Gaspar C et al. Mutations in 162 Harrigan JA, Belotserkovskaya R, Coates J, Dimitrova DS, Polo SE, Bradshaw CR et the APC tumour suppressor gene cause chromosomal instability. Nat Cell Biol al. Replication stress induces 53BP1-containing OPT domains in G1 cells. JCell 2001; 3: 433–438. Biol 2011; 193: 97–108. 137 Dikovskaya D, Schiffmann D, Newton IP, Oakley A, Kroboth K, Sansom O et al. 163 Lukas C, Savic V, Bekker-Jensen S, Doil C, Neumann B, Pedersen RS et al. 53BP1 Loss of APC induces polyploidy as a result of a combination of defects in mitosis nuclear bodies form around DNA lesions generated by mitotic transmission of and apoptosis. J Cell Biol 2007; 176: 183–195. chromosomes under replication stress. Nat Cell Biol 2011; 13: 243–253. 138 Draviam VM, Shapiro I, Aldridge B, Sorger PK. Misorientation and reduced 164 Guerrero AA, Gamero MC, Trachana V, Futterer A, Pacios-Bras C, Diaz-Concha NP stretching of aligned sister kinetochores promote chromosome missegregation et al. Centromere-localized breaks indicate the generation of DNA damage by in EB1- or APC-depleted cells. EMBO J 2006; 25: 2814–2827. the mitotic spindle. Proc Natl Acad Sci USA. 2009; 107: 4159–4164. 139 Zhang J, Ahmad S, Mao Y. BubR1 and APC/EB1 cooperate to maintain meta- 165 Boveri T. Zur Frage der Entstehung maligner Tumoren, vol. 1. Jena, Gustav Fischer, phase chromosome alignment. J Cell Biol 2007; 178: 773–784. 1914. 140 Aoki K, Aoki M, Sugai M, Harada N, Miyoshi H, Tsukamoto T et al. Chromosomal 166 Hansemann DPv. Ueber patologische Mitosen. Archiv fu¨r pathologische Anatomie instability by beta-catenin/TCF transcription in APC or beta-catenin mutant cells. and Physiologie und fu¨r klinische Medicin 1891; 119: 299–326. Oncogene 2007; 26: 3511–3520. 167 Ricke RM, van Ree JH, van Deursen JM. Whole chromosome instability and 141 Aoki K, Taketo MM. Adenomatous polyposis coli (APC): a multi-functional tumor cancer: a complex relationship. Trends Genet 2008; 24: 457–466. suppressor gene. J Cell Sci 2007; 120(Pt 19): 3327–3335. 168 Rajagopalan H, Lengauer C. Aneuploidy and cancer. Nature 2004; 432: 338–341. 142 Heun P, Erhardt S, Blower MD, Weiss S, Skora AD, Karpen GH. Mislocalization of 169 Pei L, Melmed S. Isolation and characterization of a pituitary tumor-transforming the Drosophila centromere-specific histone CID promotes formation of func- gene (PTTG). Mol Endocrinol 1997; 11: 433–441. tional ectopic kinetochores. Dev Cell 2006; 10: 303–315. 170 Zhang D, Hirota T, Marumoto T, Shimizu M, Kunitoku N, Sasayama T et al. Cre-loxP- 143 Olszak AM, van Essen D, Pereira AJ, Diehl S, Manke T, Maiato H et al. Hetero- controlled periodic Aurora-A overexpression induces mitotic abnormalities and chromatin boundaries are hotspots for de novo kinetochore formation. Nat Cell hyperplasia in mammary glands of mouse models. Oncogene 2004; 23: 8720–8730. Biol 2011; 13: 799–808. 171 Weaver BA, Silk AD, Montagna C, Verdier-Pinard P, Cleveland DW. Aneuploidy 144 Pampalona J, Soler D, Genesca A, Tusell L. Whole chromosome loss is promoted acts both oncogenically and as a tumor suppressor. Cancer Cell 2007; 11: 25–36. by telomere dysfunction in primary cells. Genes Chromosomes Cancer 2010; 49: 172 Pavelka N, Rancati G, Zhu J, Bradford WD, Saraf A, Florens L et al. Aneuploidy 368–378. confers quantitative proteome changes and phenotypic variation in budding 145 Ichijima Y, Yoshioka K, Yoshioka Y, Shinohe K, Fujimori H, Unno J et al. DNA yeast. Nature 2010; 468: 321–325. lesions induced by replication stress trigger mitotic aberration and tetraploidy 173 Rancati G, Pavelka N, Fleharty B, Noll A, Trimble R, Walton K et al. Aneuploidy development. PLoS One 2010; 5: e8821. underlies rapid adaptive evolution of yeast cells deprived of a conserved cyto- 146 Chan KL, Palmai-Pallag T, Ying S, Hickson ID. Replication stress induces kinesis motor. Cell 2008; 135: 879–893. sister-chromatid bridging at fragile site loci in mitosis. Nat Cell Biol 2009; 11: 174 McGranahan N, Burrell RA, Endesfelder D, Novelli MR, Swanton C. Cancer 753–760. chromosomal instability: therapeutic and diagnostic challenges. EMBO Rep 2012; 147 Stewenius Y, Jin Y, Ora I, de Kraker J, Bras J, Frigyesi A et al. Defective chro- 13: 528–538. mosome segregation and telomere dysfunction in aggressive Wilms’ tumors. 175 Holland AJ, Cleveland DW. Losing balance: the origin and impact of aneuploidy Clin Cancer Res 2007; 13(22 Pt 1): 6593–6602. in cancer. EMBO Rep 2012; 13: 501–514. 148 Chan KL, North PS, Hickson ID. BLM is required for faithful chromosome seg- 176 Dobles M, Liberal V, Scott ML, Benezra R, Sorger PK. Chromosome missegrega- regation and its localization defines a class of ultrafine anaphase bridges. EMBO J tion and apoptosis in mice lacking the mitotic checkpoint protein Mad2. Cell 2007; 26: 3397–3409. 2000; 101: 635–645. 149 Allshire RC, Karpen GH. Epigenetic regulation of centromeric chromatin: old 177 Baker DJ, Jeganathan KB, Malureanu L, Perez-Terzic C, Terzic A, van Deursen JM. dogs, new tricks? Nat Rev Genet 2008; 9: 923–937. Early aging-associated phenotypes in Bub3/Rae1 haploinsufficient mice. JCell 150 Hu Z, Huang G, Sadanandam A, Gu S, Lenburg ME, Pai M et al. The Biol 2006; 172: 529–540. expression level of HJURP has an independent prognostic impact and 178 Jeganathan K, Malureanu L, Baker DJ, Abraham SC, van Deursen JM. Bub1 predicts the sensitivity to radiotherapy in breast cancer. Breast Cancer Res 2010; mediates cell death in response to chromosome missegregation and acts to 12: R18. suppress spontaneous tumorigenesis. J Cell Biol 2007; 179: 255–267. 151 Tomonaga T, Matsushita K, Yamaguchi S, Oohashi T, Shimada H, Ochiai T et al. 179 Weaver BA, Bonday ZQ, Putkey FR, Kops GJ, Silk AD, Cleveland DW. Centromere- Overexpression and mistargeting of centromere protein-A in human primary associated protein-E is essential for the mammalian mitotic checkpoint to pre- colorectal cancer. Cancer Res 2003; 63: 3511–3516. vent aneuploidy due to single chromosome loss. J Cell Biol 2003; 162: 551–563. 152 Van Hooser AA, Ouspenski II, Gregson HC, Starr DA, Yen TJ, Goldberg ML et al. 180 Ricke RM, Jeganathan KB, van Deursen JM. Bub1 overexpression induces Specification of kinetochore-forming chromatin by the histone H3 variant CENP- aneuploidy and tumor formation through Aurora B kinase hyperactivation. JCell A. J Cell Sci 2001; 114(Pt 19): 3529–3542. Biol 2011; 193: 1049–1064. 153 Barnhart MC, Kuich PH, Stellfox ME, Ward JA, Bassett EA, Black BE et al. HJURP is a 181 van Ree JH, Jeganathan KB, Malureanu L, van Deursen JM. Overexpression of the CENP-A chromatin assembly factor sufficient to form a functional de novo E2 ubiquitin-conjugating enzyme UbcH10 causes chromosome missegregation kinetochore. J Cell Biol 2011; 194: 229–243. and tumor formation. J Cell Biol 2010; 188: 83–100. 154 Crasta K, Ganem NJ, Dagher R, Lantermann AB, Ivanova EV, Pan Y et al. DNA 182 Baker DJ, Jin F, Jeganathan KB, van Deursen JM. Whole chromosome instability breaks and chromosome pulverization from errors in mitosis. Nature 2012; 482: caused by Bub1 insufficiency drives tumorigenesis through tumor suppressor 53–58. gene loss of heterozygosity. Cancer Cell 2009; 16: 475–486. 155 Janssen A, van der Burg M, Szuhai K, Kops GJ, Medema RH. Chromosome seg- 183 Rao CV, Yang YM, Swamy MV, Liu T, Fang Y, Mahmood R et al. Colonic tumor- regation errors as a cause of DNA damage and structural chromosome aberra- igenesis in BubR1 þ /-ApcMin/ þ compound mutant mice is linked to premature tions. Science 2011; 333: 1895–1898. separation of sister chromatids and enhanced genomic instability. Proc Natl Acad 156 Sheltzer JM, Blank HM, Pfau SJ, Tange Y, George BM, Humpton TJ et al. Sci USA. 2005; 102: 4365–4370. Aneuploidy drives genomic instability in yeast. Science 2011; 333: 1026–1030. 184 Baker DJ, Perez-Terzic C, Jin F, Pitel K, Niederlander NJ, Jeganathan K et al. 157 Fabarius A, Hehlmann R, Duesberg PH. Instability of chromosome structure in Opposing roles for p16Ink4a and p19Arf in senescence and ageing caused by cancer cells increases exponentially with degrees of aneuploidy. Cancer Genet BubR1 insufficiency. Nat Cell Biol 2008; 10: 825–836. Cytogenet 2003; 143: 59–72. 185 Thompson SL, Compton DA. Proliferation of aneuploid human cells is limited by 158 Duesberg P, Rausch C, Rasnick D, Hehlmann R. Genetic instability of cancer cells a p53-dependent mechanism. J Cell Biol 2010; 188: 369–381. is proportional to their degree of aneuploidy. Proc Natl Acad Sci USA 1998; 95: 186 Torres EM, Dephoure N, Panneerselvam A, Tucker CM, Whittaker CA, Gygi SP 13692–13697. et al. Identification of aneuploidy-tolerating mutations. Cell 2010; 143: 71–83. 159 Burds AA, Lutum AS, Sorger PK. Generating chromosome instability through the 187 Duesberg P, Fabarius A, Hehlmann R. Aneuploidy the primary cause of the simultaneous deletion of Mad2 and p53. Proc Natl Acad Sci USA 2005; 102: multilateral genomic instability of neoplastic and preneoplastic cells. IUBMB Life 11296–11301. 2004; 56: 65–81. 160 Torres EM, Sokolsky T, Tucker CM, Chan LY, Boselli M, Dunham MJ et al. Effects of 188 Li M, Fang X, Baker DJ, Guo L, Gao X, Wei Z et al. The ATM-p53 pathway aneuploidy on cellular physiology and cell division in haploid yeast. Science suppresses aneuploidy-induced tumorigenesis. Proc Natl Acad Sci USA. 2010; 2007; 317: 916–924. 107: 14188–14193.

& 2013 Macmillan Publishers Limited Oncogene (2013) 4459 – 4470 Genetic instability in tumors A Janssen and RH Medema 4470 189 Luongo C, Moser AR, Gledhill S, Dove WF. Loss of Apc þ in intestinal adenomas 196 Garcia-Higuera I, Manchado E, Dubus P, Canamero M, Mendez J, Moreno S et al. from Min mice. Cancer Res 1994; 54: 5947–5952. Genomic stability and tumour suppression by the APC/C cofactor Cdh1. Nat Cell 190 Birkbak NJ, Eklund AC, Li Q, McClelland SE, Endesfelder D, Tan P et al. Paradoxical Biol 2008; 10: 802–811. relationship between chromosomal instability and survival outcome in cancer. 197 Diaz-Rodriguez E, Sotillo R, Schvartzman JM, Benezra R. Hec1 overexpression Cancer Res 2011; 71: 3447–3452. hyperactivates the mitotic checkpoint and induces tumor formation in vivo. Proc 191 Roylance R, Endesfelder D, Gorman P, Burrell RA, Sander J, Tomlinson I et al. Natl Acad Sci USA 2008; 105: 16719–16724. Relationship of extreme chromosomal instability with long-term survival in a 198 Malureanu L, Jeganathan KB, Jin F, Baker DJ, van Ree JH, Gullon O et al. Cdc20 retrospective analysis of primary breast cancer. Cancer Epidemiol Biomarkers Prev hypomorphic mice fail to counteract de novo synthesis of cyclin B1 in mitosis. 2011; 20: 2183–2194. J Cell Biol 2010; 191: 313–329. 192 Janssen A, Medema RH. Mitosis as an anti-cancer target. Oncogene. [Review] 199LiM,FangX,WeiZ,YorkJP,ZhangP.Lossof spindle assembly checkpoint-mediated 2011; 30: 2799–2809. inhibition of Cdc20 promotes tumorigenesis in mice. JCellBiol2009; 185: 983–994. 193 Chi YH, Ward JM, Cheng LI, Yasunaga J, Jeang KT. Spindle assembly checkpoint 200 Chesnokova V, Kovacs K, Castro AV, Zonis S, Melmed S. Pituitary hypoplasia in and p53 deficiencies cooperate for tumorigenesis in mice. Int J Cancer 2009; 124: Pttg-/- mice is protective for Rb þ /- pituitary tumorigenesis. Mol Endocrinol 2005; 1483–1489. 19: 2371–2379. 194 Schliekelman M, Cowley DO, O’Quinn R, Oliver TG, Lu L, Salmon ED et al. 201 Wang Z, Yu R, Melmed S. Mice lacking pituitary tumor transforming gene show Impaired Bub1 function in vivo compromises tension-dependent testicular and splenic hypoplasia, thymic hyperplasia, thrombocytopenia, checkpoint function leading to aneuploidy and tumorigenesis. Cancer Res 2009; aberrant cell cycle progression, and premature centromere division. Mol Endo- 69: 45–54. crinol. 2001; 15: 1870–1879. 195 Sotillo R, Schvartzman JM, Socci ND, Benezra R. Mad2-induced chromosome 202 Uren AG, Wong L, Pakusch M, Fowler KJ, Burrows FJ, Vaux DL et al. Survivin and the instability leads to lung tumour relapse after oncogene withdrawal. Nature 2010; inner centromere protein INCENP show similar cell-cycle localization and gene 464: 436–440. knockout phenotype. Curr Biol. [Research Support, Non-U.S. Gov’t] 2000; 10: 1319–1328.

Oncogene (2013) 4459 – 4470 & 2013 Macmillan Publishers Limited