You are on page 1of 15

Chromosome Res (2023) 31:18

https://doi.org/10.1007/s10577-023-09727-7

REVIEW

Twenty years of merotelic kinetochore attachments:


a historical perspective
Daniela Cimini

Received: 1 May 2023 / Revised: 20 June 2023 / Accepted: 8 July 2023 / Published online: 19 July 2023
© The Author(s), under exclusive licence to Springer Nature B.V. 2023

Abstract Micronuclei, small DNA-containing accurately to ensure that each daughter inherits a
structures separate from the main nucleus, were complete copy of the genome. A variety of mitotic
used for decades as an indicator of genotoxic dam- defects can lead to chromosome missegregation and
age. Micronuclei containing whole chromosomes result in genetic imbalances in the daughter cells
were considered a biomarker of aneuploidy and were [reviewed in (Baudoin and Cimini 2018b)]. Many of
believed to form, upon mitotic exit, from chromo- these errors are prevented by a biochemical signaling
somes that lagged behind in anaphase as all other pathway, known as the spindle assembly checkpoint
chromosomes segregated to the poles of the mitotic (or SAC), that monitors chromosome attachment to
spindle. However, the mechanism responsible for the mitotic spindle (Lara-Gonzalez et al. 2021). Ana-
inducing anaphase lagging chromosomes remained phase lagging chromosomes, however, are caused
unknown until just over twenty years ago. Here, I by a defect that can escape SAC detection and there-
summarize what preceded and what followed this dis- fore represent a major threat to mitotic fidelity. Upon
covery, highlighting some of the open questions and mitotic exit, anaphase lagging chromosomes often
opportunities for future investigation. form micronuclei in the nascent daughter cell (Cimini
et al. 2002; He et al. 2019) and micronuclei were
Keywords lagging chromosome · merotelic · shown to cause a variety of adverse effects (Krupina
micronuclei · chromosome segregation · mitosis et al. 2021). Although other chromosome segregation
defects can be observed in cancer cells and tumor tis-
sues (Gisselsson et al. 2004; Stewenius et al. 2007;
Introduction Tucker et al. 2023), anaphase lagging chromosomes
were shown to be prevalent in chromosomally unsta-
During mitosis, the sister chromatids of the repli- ble cancer cells, mouse tumor models, and certain
cated chromosomes must separate and segregate to human tumors (Bakhoum et al. 2011; Bakhoum et al.
opposite poles, and this process must be executed 2015; Bakhoum et al. 2014; Thompson and Comp-
ton 2008; Venkatesan et al. 2021; Zaki et al. 2014).
Responsible Editor: Stefano Santaguida
Similarly, micronuclei are often found in cancer cells
and tumor tissues (Gisselsson et al. 2001; Streffer
D. Cimini (*) et al. 1985), and are acknowledged to be an impor-
Department of Biological Sciences and Fralin Life tant biomarker associated with increased risk of dis-
Sciences Institute, Virginia Tech, Blacksburg, VA 24061,
ease, including cancer (Bonassi et al. 2011a, b). The
USA
e-mail: cimini@vt.edu mechanism responsible for the formation of lagging

Vol.: (0123456789)
13
18 Page 2 of 15 Chromosome Res (2023) 31:18

chromosomes was discovered just over twenty years all other chromosomes segregate to the spindle poles,
ago (Cimini et al. 2001). That discovery became the and it can form a micronucleus upon mitotic exit
seed for a series of new research questions, ranging (Fig. 1B). A micronucleus, as the name suggests, is
from the correction mechanisms, to the mechanisms a small structure consisting of DNA encapsulated by
that increase these errors in cancer cells, to the biol- a nuclear envelope, which is separated from the main
ogy of micronuclei, and more. In this review, I sum- nucleus. By the early 1990s, the cellular toxicology
marize questions, knowledge, and ideas that preceded community viewed anaphase lagging chromosomes
the identification of the mechanism underlying ana- as precursors of certain micronuclei, whose occur-
phase lagging chromosomes. Then, I briefly summa- rence was considered to be a biomarker of aneuploidy
rize all the findings that followed that discovery and (Antoccia et al. 1991; Bonatti et al. 1992; Degrassi
are still a fertile ground of investigation for research- and Tanzarella 1988; Thomson and Perry 1988).
ers in the chromosome segregation, aneuploidy, and This was partly based on detailed cytology studies
cancer biology communities. indicating that missegregation of chromosomes or
chromatids and subsequent micronucleus formation
could lead to aneuploidy in the cell progeny (Gus-
Anaphase lagging chromosomes tavino et al. 1994; Rizzoni et al. 1989). Detecting and
and kinetochore‑positive micronuclei quantifying micronuclei is simple and fast and there-
as biomarkers of aneuploidy fore they have been used as a measure of genotoxic
damage for decades (Pincu et al. 1985) in a stand-
An anaphase lagging chromosome is defined as a ardized assay named the in vitro micronucleus assay
chromosome that lags behind during anaphase, when (Fenech 2000). However, micronuclei can form as

Fig. 1  Diagram showing


formation of micronuclei
upon mitotic exit. Chromo-
somes/DNA are depicted
in blue, kinetochores in
orange. A. Formation of a
kinetochore-negative micro-
nucleus from a lagging
chromosome fragment. B.
Formation of a kinetochore-
positive micronucleus
from an anaphase lagging
chromosome. C. Formation
of a kinetochore-positive
micronucleus in a binucle-
ate cell in the cytokinesis-
block assay

Vol:. (1234567890)
13
Chromosome Res (2023) 31:18 Page 3 of 15 18

a result of either missegregation of whole chromo- lagging chromosomes or counting chromosomes in


somes (Fig. 1B) or acentric chromosome fragments chromosome spreads, this approach could be used to
(Fig. 1A) that cannot interact with the mitotic spin- quantify micronuclei in thousands of interphase cells
dle, and the simple visualization of micronuclei does in tens of minutes (Fenech 2000). A variation of this
not provide information on their origin. In the late approach was subsequently developed to selectively
1980s, the genetic toxicology community recognized identify micronuclei in cells that were proliferative
the need to identify genotoxic agents that affected and underwent cell division during or after exposure.
cells and organisms in ways other than by damaging This method was based on the idea that aneuploidy
their DNA or inducing gene mutations. Specifically, arising in proliferative cells would affect the cell pop-
it had become apparent that certain chemical agents ulation, and hence organismal health; moreover, this
do not cause DNA damage (i.e., are not clastogenic), approach would prevent micronuclei already present
but can cause aneuploidy (i.e., are aneugenic), which in the cell population from confounding the data. In
can severely impair cell and organismal physiology. this approach, named the cytokinesis-block assay,
At that time, the method of choice for assessing ane- cells were treated, during or after exposure, with
uploidy was counting chromosomes in chromosome cytochalasin (an inhibitor of actin polymerization) for
spreads. However, such approach is time consuming a time window corresponding approximately to cell
and has limited statistical power, given that typically cycle duration prior to fixation and analysis. All the
only ~ 50 chromosome spreads are analyzed per sam- cells undergoing cell division during the cytochalasin
ple. Therefore, there was a need to develop an assay treatment would be unable to complete cytokinesis
that allowed to quantify aneuploidy quickly and eas- and would therefore appear as binucleate cells; analy-
ily. The in vitro micronucleus assay appeared to sis of micronuclei could then focus specifically on the
have the right requisites, but only if strategies could binucleate cell population (Fig. 1C). The combination
be devised to distinguish micronuclei that contained of the cytochalasin treatment with kinetochore stain-
entire chromosomes (from aneugenic effects) from ing was viewed as a powerful method in the genetic
those containing chromosome fragments (from clas- toxicology community (Fenech 2000), although com-
togenic effects). To this end, attempts were made to parison of rates of anaphase lagging chromosomes
discriminate micronuclei based on their size and/or vs. CREST-positive micronuclei in binucleate cells
their DNA content, assuming that micronuclei that revealed that this method underestimated the num-
were larger and/or contained more DNA, were more ber of anaphase lagging chromosomes (Cimini et al.
likely to contain entire chromosomes compared to 1999).
smaller micronuclei (Hogstedt and Karlsson 1985; Although the genetic toxicology community
Pincu et al. 1985; Yamamoto and Kikuchi 1980). This broadly used centromere/kinetochore-positive micro-
idea was justified, given that micronuclei containing nuclei as a readout of aneuploidy and overall agreed
larger chromosomes were recently reported to have that such micronuclei originated from anaphase lag-
a larger area (Mammel et al. 2022). However, these ging chromosomes, the mechanism causing lagging
approaches are laborious and difficult to standardize chromosomes remained unknown.
and therefore were never adopted as routine methods
in the genetic toxicology field. The discovery of anti-
bodies from scleroderma patients (CREST) that local- Merotelic kinetochore attachment: the underlying
ized to the kinetochore (Moroi et al. 1980) enabled cause of anaphase lagging chromosomes
the discrimination between micronuclei containing
entire chromosomes (CREST-positive micronuclei) Because the genetic toxicology community was not
and micronuclei containing chromosome fragments accustomed to thinking about cellular mechanisms
(CREST-negative micronuclei) (Fig. 1A-B). A num- and the cell biology community was not focused on
ber of studies focused on assessing the ability of aneuploidy and its consequences, the cellular mech-
such approach to predict the aneugenic potential of anism causing chromosomes to lag behind during
physical and chemical agents (Antoccia et al. 1991; anaphase was unknown until just over 20 years ago.
Fenech and Morley 1989; Hennig et al. 1988; Schuler Nevertheless, some ideas were shared in informal dis-
et al. 1997). As opposed to the detection of anaphase cussions and included overriding of the SAC in the

Vol.: (0123456789)
13
18 Page 4 of 15 Chromosome Res (2023) 31:18

presence of unattached chromosomes/kinetochores, kinetochore binds microtubules from both spindle


accidental detachment of microtubules from kineto- poles instead of just one (Cimini et al. 2001). This
chores during anaphase, and failure of sister chroma- type of mis-attachment had previously been reported
tids to separate (which would lead to the formation in cold- or drug-treated crane-fly spermatocytes,
of micronuclei with two kinetochores) as the poten- where the term merotelic was first used (Janicke and
tial causes of anaphase lagging chromosomes. The LaFountain 1984; Ladrach and LaFountain 1986),
idea that anaphase lagging chromosomes may result and in cells undergoing cell division with individual
from a “leaky” SAC was perhaps influenced by the chromatids or chromosome fragments containing
term used for chromosomes that, in prometaphase single kinetochores (Khodjakov et al. 1997; Wise
cells, were positioned close the spindle poles. These and Brinkley 1997; Yu and Dawe 2000). The obser-
chromosomes were often referred to as “lagging vation that merotelic kinetochore attachment could
chromosomes” and perhaps this terminology led to explain the origin of anaphase lagging chromosomes
the idea that these unaligned prometaphase chromo- in mitotic cells provided a possible mechanism for the
somes failed to align at the metaphase plate prior to aneuploidy and chromosomal instability (CIN) fre-
anaphase onset and were the precursors of anaphase quently observed in cancer cells (Bolhaqueiro et al.
lagging chromosomes. We now prefer to refer to such 2019; Lengauer et al. 1997; Nicholson and Cimini
prometaphase chromosomes as “unaligned” chromo- 2013; Rajagopalan and Lengauer 2004; Weaver and
somes to differentiate them from the anaphase lagging Cleveland 2006). In support of this idea, subsequent
chromosomes (Baudoin and Cimini 2018b). It was studies showed that most cancer cells display high
also shown that unaligned chromosomes are not seen rates of anaphase lagging chromosomes (Bakhoum
at anaphase onset in cells with anaphase lagging chro- et al. 2014, Ganem et al. 2009, Silkworth et al. 2009,
mosomes and that cells with anaphase lagging chro- Thompson and Compton 2008) and lagging chromo-
mosomes do not experience a pre-anaphase mitotic somes were also observed in tumor tissue samples
delay (Cimini et al. 2002), which typically occurs (Bakhoum et al. 2011; Zaki et al. 2014), although
when unaligned chromosomes are present (Rieder this may not be the case for all tumor types (Gissels-
et al. 1994, 1995). These observations definitely dis- son et al. 2004; Stewenius et al. 2007; Tucker et al.
missed the idea of anaphase onset in the presence 2023). Merotelic kinetochore attachment was fur-
of unattached kinetochores as the underlying cause ther shown to be a key mechanism of aneuploidy in
of anaphase lagging chromosomes. The accidental human pluripotent stem cells (Deng et al. 2023) and
detachment of microtubules from the kinetochore was in mammalian oocytes, where it can promote chromo-
also not plausible, given that the stability of micro- some missegregation in both meiosis I and II (Cheng
tubules had been shown to dramatically increase in et al. 2017, Holubcova et al. 2015; Kouznetsova et al.
anaphase compared to metaphase (Zhai et al. 1995) 2014; Shomper et al. 2014, Zielinska et al. 2015),
and we now know that microtubule pulling forces sta- although other mechanisms of chromosome misseg-
bilize kinetochore-microtubule attachments (Akiyoshi regation have also been described in meiotic cells.
et al. 2010), thus making detachment in anaphase For instance, one study identified lack of kinetochore-
highly unlikely. Finally, failure of sister chromatids microtubule attachment as a major cause of lag-
to separate was also very unlikely, given that staining ging chromosome formation and aneuploidy in aged
with individual centromeric probes showed micronu- mouse oocytes (Mihajlovic et al. 2021).
clei contained individual centromeres and anaphase Overall, there is currently strong agreement that
lagging chromosomes consisted of individual daugh- merotelically attached lagging chromosomes repre-
ter chromosomes and not paired chromatids (Cata- sent a major source of aneuploidy and chromosomal
lan et al. 2000; Cimini et al. 1997, 1999; Falck et al. instability in normal and cancer cells.
2002).
Detailed analysis of chromosomes/kinetochores
and microtubules revealed that the underlying cause How do merotelic attachments arise?
of anaphase lagging chromosomes was merotelic
kinetochore attachment (Fig. 2), a type of kine- Establishment of kinetochore-microtubule attach-
tochore-microtubule attachment in which a single ments is a stochastic process and therefore different

Vol:. (1234567890)
13
Chromosome Res (2023) 31:18 Page 5 of 15 18

Fig. 2  Fate of anaphase lagging chromosomes depends on the cally attached kinetochores can be seen. However, because of
relative size of the microtubule bundles connecting the mero- the different size of the two microtubule bundles connecting
telic kinetochore to the two spindle poles. A. Example of live the merotelic kinetochores to the two spindle poles, the chro-
cell (top) and diagram (middle) of a “balanced” merotelically mosomes’ position is shifted close to the group of normally
attached anaphase lagging chromosome. In the images (taken segregating chromosomes on the right. As a result of this, the
two minutes apart), the lagging kinetochore can be seen per- chromosome(s) will be included in the main nucleus in one
sisting at the cell equator as all the other chromosomes move of the daughter cells (bottom). In the images, kinetochores
poleward. As a result of this, a whole chromosome-contain- are in green and microtubules are red. In the diagrams, chro-
ing micronucleus is formed in one of the daughter cells (bot- mosomes/DNA are depicted in blue, kinetochores in orange.
tom). B. Example of live cell (top) and diagram (middle) of an Live cell images are modified from Cimini et al. 2004 (A) and
“unbalanced” merotelically attached anaphase lagging chromo- Cimini et al. 2006 (B)
some. In the images (taken three minutes apart), two meroteli-

types of attachments can be established in early mito- certain fission yeast mutants are believed to form high
sis [reviewed in (Gregan et al. 2011; Sarangapani and rates of merotelic attachments because of disorgan-
Asbury 2014)]. Merotelic kinetochore attachments ized kinetochore structure (Gregan et al. 2011, 2007;
form often during the early stages of mitosis in nor- Pidoux et al. 2000; Rumpf et al. 2010). Similarly, a
mal bipolar cells, but they decrease as mitosis pro- C. elegans mutant in which the kinetochore is twisted
gresses (Cimini et al. 2003). Ultimately, chromosome around the chromatin forms high rates of merotelic
mis-segregation due to merotelic kinetochore attach- attachments (Stear and Roth 2002). Finally, in Indian
ment will depend on the overall balance between muntjac cells, whose chromosomes possess kineto-
rates of formation of such mis-attachments and the chores of varying sizes, the probability of merotelic
rates at which they are corrected. The rates of mero- attachment increases with kinetochore size (Baudoin
telic attachment formation were shown to increase in and Cimini 2018a, Drpic et al. 2018).
several contexts, mostly having to do with either kine- Spindle multipolarity is one spindle geom-
tochore structure or spindle geometry. For instance, etry defect that was shown to increase the rates of

Vol.: (0123456789)
13
18 Page 6 of 15 Chromosome Res (2023) 31:18

merotelic kinetochore attachments. This happens missegregation from merotelic attachment. The next
because within a multipolar spindle it is easier for a section will focus on such mechanisms.
single kinetochore to face more than one spindle pole
than it would be within a bipolar spindle (Ganem
et al. 2009; Paul et al. 2009; Silkworth and Cimini Correction of merotelic kinetochore attachments
2012, Silkworth et al. 2009) and indeed it was shown throughout mitosis
that increasing numbers of spindle poles correlate
with increasing numbers of merotelic attachments Merotelic kinetochore attachments are very common
(Paul et al. 2009). This can explain earlier observa- in early mitosis, but their numbers decrease as mitosis
tions that lagging chromosomes are very commonly progresses (Cimini et al. 2003, 2004). The Aurora B
observed in multipolar anaphases (Sluder et al. kinase, which was already known to be essential for
1997) and in cells recovering from a nocodazole- chromosome bi-orientation in budding yeast (Biggins
induced mitotic arrest (Cimini et al. 2001, 2003, et al. 1999; Tanaka et al. 2002), has been described
1999; Ladrach and LaFountain 1986). Indeed, in as a major player in the merotelic error correction
cells recovering from a nocodazole arrest, microtu- that occurs in prometaphase and metaphase (Cimini
bules assemble from the two centrosomes as well as et al. 2006) thanks to its ability to phosphorylate
from multiple ectopic microtubule organizing cent- kinetochore substrates, specifically the Hec1 subu-
ers (MTOCs) (Cimini et al. 2003). However, these nit of the Ndc80 complex, and cause microtubule
ectopic MTOCs eventually cluster with the two cen- detachment (Cheeseman et al. 2006; Ciferri et al.
trosomes to form a bipolar spindle (Cimini et al. 2008; DeLuca et al. 2006). Detachment of microtu-
2003). The transient multipolarity state is sufficient bules from the kinetochore provides the opportunity
to enhance the rates of merotelic kinetochore attach- for loss of attachments from the incorrect pole and
ment and hence the rates of anaphase lagging chro- formation of new attachments to microtubules from
mosomes (Ganem et al. 2009; Silkworth and Cimini the correct pole. Importantly, measurements of kine-
2012, Silkworth et al. 2009). Notably, this transient tochore-microtubule turnover showed that cells with
multipolarity followed by centrosome clustering is high rates of anaphase lagging chromosomes have
believed to play an important role in promoting chro- more stable kinetochore-microtubule attachments
mosome missegregation in cancer cells with super- in either prometaphase, metaphase, or both (Bak-
numerary centrosomes (Brinkley 2001; Ganem et al. houm et al. 2009a). The exact mechanism by which
2009, Silkworth et al. 2009). Increased rates of mero- Aurora B kinase destabilizes kinetochore-microtubule
telic attachment formation are also observed when the attachments is still debated, with large part of the
centrosomes/spindle poles in mitotic cells do not sep- debate revolving around whether the pool of Aurora
arate prior to nuclear envelope breakdown (Kaseda B localized at the centromere or a pool localized at
et al. 2012; Nam and van Deursen 2014; Silkworth the kinetochore is responsible for the phosphorylation
and Cimini 2012; Silkworth et al. 2012). This hap- of Hec1 required to detach microtubules [for a review
pens because, if the spindle poles are in close prox- on this topic see (Broad and DeLuca 2020)]. The cen-
imity while kinetochore-microtubule attachments are tromere-localized kinesin-13s MCAK and Kif2b were
established, the chance of a single kinetochore to bind also shown to play a key role in the correction of kine-
microtubules from both spindle poles is higher than tochore-microtubule mis-attachments, as their sup-
it would be if the two spindle poles were at diamet- pression results in reduced turnover of kinetochore-
rically opposed positions around the chromosomes microtubules and increased rates of anaphase lagging
(Silkworth and Cimini 2012; Silkworth et al. 2012). chromosomes (Bakhoum et al. 2009b; Kline-Smith
Because merotelic kinetochores are attached to et al. 2004; Maney et al. 1998). Interestingly, Auora
microtubules, the SAC is silenced and therefore B-dependent phosphorylation was also shown to
higher frequencies of merotelic attachments invari- modulate MCAK activity (Andrews et al. 2004; Lan
ably translate into higher frequencies of anaphase et al. 2004; Zhang et al. 2007). Importantly, potenti-
lagging chromosomes. Nevertheless, correction ating MCAK using a small molecule was shown to
mechanisms exist in both prometaphase/metaphase reduce the rates of anaphase lagging chromosomes
and anaphase to reduce the rates of chromosome in cancer cells, suggesting that this missegregation

Vol:. (1234567890)
13
Chromosome Res (2023) 31:18 Page 7 of 15 18

errors could be a druggable target, although the can- (Cimini et al. 2004; Thompson and Compton 2011)
cer cells quickly adapted and reestablished their (Fig. 2). Specifically, if the lagging chromosome is
pre-treatment rates of lagging chromosomes (Orr bound to microtubule bundles of similar thickness
et al. 2016). A recent study has also suggested that (i.e., containing similar numbers of microtubules;
an interplay between Aurora B kinase activity and Fig. 2A), then the chromosome will remain close to
mechanical tension specifically contributes to correc- the cell equator (Cimini et al. 2004), whereas if the
tion of merotelic kinetochore attachments as opposed two microtubule bundles are of different thickness
to kinetochore-microtubule mis-attachments lacking (as measured by fluorescence intensity; Fig. 2B),
tension (Chen et al. 2021). This model is somewhat then the chromosome will be shifted to one side
inconsistent with in vitro data showing that tension (Cimini et al. 2004), typically sufficiently close to
can suppress Aurora B-mediated detachment of kine- one of the two groups of segregating chromosomes
tochore-bound microtubules (de Regt et al. 2022), but to avoid missegregation. Initially, this behavior
this may be because the interplay between tension was attributed to the effect tension can have on
and Aurora B influences amphitelic and merotelic kinetochore-bound microtubules, with high tension
attachments differently. Indeed, the Aurora B-medi- promoting polymerization (Maddox et al. 2003).
ated correction of merotelic attachments may specifi- In the case of differentially sized microtubule bun-
cally depend on the deformation of merotelic kineto- dles attached to the kinetochore, the smaller bundle
chores in metaphase and on the enrichment of Aurora will experience higher tension and therefore will
B at those sites (Cimini et al. 2004, 2006; Knowlton lengthen during anaphase, resulting in a shift of the
et al. 2006; Trivedi et al. 2019). Finally, there is evi- lagging chromosome away from the equator and
dence that, although a defective SAC may not directly close to the pole connected to the thicker bundle
cause anaphase lagging chromosomes, premature (Cimini et al. 2004) (Fig. 2B). In recent years, two
SAC silencing reduces the time window for the pre- groups have highlighted a contribution of Aurora B
anaphase correction of merotelic attachments, leading kinase, which localizes at the spindle midzone in
to increased rates of anaphase lagging chromosomes anaphase and can directly phosphorylate substrates
(Cimini et al. 2003). at the kinetochore of lagging chromosomes (Papini
Merotelic kinetochore attachments do not sus- et al. 2021), in minimizing the number of anaphase
tain SAC signaling the way unattached kinetochores lagging chromosomes resulting in chromosome
do [for a review on the SAC see (Lara-Gonzalez missegregation (Orr et al. 2021, Sen et al. 2021),
et al. 2021)]. Therefore, although many merotelic although there is still some debate on the exact
attachments can be corrected via the mechanisms mechanism by which this happens [reviewed in
mentioned above, some can persist into anaphase. (Maiato and Silva 2023)]. An “anaphase correction
Early studies showed that merotelic kinetochore mechanism” was also invoked in an earlier study in
attachments that persist into anaphase can lead to mouse oocytes showing a ten-fold ratio between the
different outcomes. In some cases, merotelically number of lagging chromosomes in anaphase II and
attached lagging chromosomes remain positioned at the number of chromosomes that eventually misseg-
the spindle equator, whereas in other cases they are regated (Kouznetsova et al. 2019). Similar observa-
shifted toward one of the two groups of segregating tions were also reported for anaphase II in insect
chromosomes (Cimini et al. 2004; Thompson and spermatocytes (Janicke et al. 2007). In at least one
Compton 2011). Anaphase lagging chromosomes cell line, it was shown that in cases in which a lag-
that remain close to the spindle equator, invariably ging chromosome is incorporated into the main
form micronuclei (Cimini et al. 2002), whereas nucleus, this is often the correct nucleus (Thomp-
those that are shifted towards one of the two spin- son and Compton 2011).
dle poles can be incorporated into the main nucleus Despite all the safety mechanisms described in
(Thompson and Compton 2011). The differential this section, some anaphase lagging chromosomes
positioning of lagging chromosomes during ana- will remain in proximity of the cell equator through
phase was shown to depend on the relative number anaphase and beyond, and this can have dire conse-
of microtubules emanating from the kinetochore of quences [reviewed in (Krupina et al. 2021)], as dis-
the lagging chromosome to the two opposite poles cussed in the next section.

Vol.: (0123456789)
13
18 Page 8 of 15 Chromosome Res (2023) 31:18

Micronucleus formation and downstream effects proteins, including nuclear pore complexes, during
nuclear envelope reassembly upon mitotic exit (Liu
When an anaphase lagging chromosome persists in et al. 2018). Consequently, micronuclei fail to properly
proximity of the spindle equator through the end of import key proteins that are necessary for the integrity
cell division, it invariably forms a micronucleus upon of their nuclear envelope and DNA.
mitotic exit (Cimini et al. 2002; Thompson and Comp- Micronuclei were also shown to frequently expe-
ton 2011). It should be noted that anaphase lagging rience nuclear envelope rupture (Hatch et al. 2013;
chromosomes are not the only source of micronuclei, Maciejowski and Hatch 2020). A recent study
which can also form from “polar chromosomes” that do showed that micronuclei containing larger and more
not align at the metaphase plate prior to anaphase onset gene-dense chromosomes are less likely to rupture
(Ferrandiz et al. 2022, Gomes et al. 2022) or fragments (Mammel et al. 2022), but the chance of membrane
of DNA resulting from pre-mitotic DNA damage or rupture is never zero. When the micronucleus enve-
chromosome bridges that break during cell division lope ruptures, the micronuclear DNA can become
(Hennig et al. 1988, Hoffelder et al. 2004, Pampalona exposed to cytoplasmic content, including nucle-
et al. 2016, Schuler et al. 1997, Umbreit et al. 2020). ases that can digest and fragment the DNA, simi-
Over the past decade, many studies have focused on larly to what was observed for persistent chromatin
the biology of micronuclei and on the fate of cells that bridges (Maciejowski et al. 2015). Defective DNA
enter mitosis with a micronucleus (Crasta et al. 2012, metabolism (e.g., defective DNA repair and repli-
Hatch et al. 2013, He et al. 2019, Liu et al. 2018, Soto cation) within micronuclei and micronucleus mem-
et al. 2018, Vazquez-Diez et al. 2016). Most of these brane rupture have been proposed as the underlying
studies used experimental protocols that resulted in causes of the “chromosome pulverization” reported
increased rates of anaphase lagging chromosomes as a in micronucleated cells that reenter mitosis (Crasta
way to increase the number of analyzable micronuclei et al. 2012; Kato and Sandberg 1968). Importantly,
(Crasta et al. 2012, Hatch et al. 2013, He et al. 2019, this chromosome pulverization is believed to be a
Liu et al. 2018, Soto et al. 2018). Thus, a wealth of precursor of chromothripsis, a catastrophic event in
knowledge has been accumulated on the biology, fate, which a single chromosome or chromosome region
and impact of lagging chromosome-derived micronu- undergoes shattering and re-stitching to gener-
clei. An early study showed that micronuclei accumu- ate a highly rearranged genomic region and that is
lated DNA damage due to defective transport of DNA detected in a substantial number of tumors (Cortes-
repair enzymes and other proteins inside the micronu- Ciriano et al. 2020, Stephens et al. 2011). Several
cleus (Crasta et al. 2012). Similar findings were also studies have shown clear evidence of chromothripsis
reported in mouse embryos, where micronuclei dis- being triggered by encapsulation of chromosomes
played nuclear envelope defects, accumulation of DNA into micronuclei (Ly et al. 2019, 2017; Zhang et al.
damage, and impaired kinetochore assembly, which 2015). Ruptured micronuclei were also shown to
in turn led to persistent missegregation of the micro- activate the cGAS-STING pathway responsible for
nucleated chromosome (Vazquez-Diez et al. 2016). cellular immune response to cytosolic DNA (Hard-
These findings are consistent with earlier studies that ing et al. 2017; Maciejowski and Hatch 2020; Mac-
analyzed micronuclei containing chromosome frag- kenzie et al. 2017), and this was in turn shown to
ments instead of whole chromosomes. For instance, promote cancer progression by driving metastasis
micronuclei emerging from chromosome bridge break- (Bakhoum et al. 2018; Krupina et al. 2021; Macie-
age were shown to display defective nuclear pore jowski and Hatch 2020).
complex assembly, nuclear import defects, reduced Finally, even when they remain intact and trans-
transcriptional activity, and chromatin fragmentation port of DNA repair factors is not impaired, micronu-
(Hoffelder et al. 2004). Similarly, radiation-induced clei were shown to display abnormal mitotic behavior
micronuclei (which likely contain DNA fragments) when the micronucleated cell reentered mitosis (He
displayed defective recruiting of DNA repair fac- et al. 2019; Soto et al. 2018). This was due to a num-
tors (Terradas et al. 2012, 2009), likely due to nuclear ber of mechanisms, including defective or delayed
envelope assembly defects arising from the inability chromosome condensation and defective recruitment
of lagging DNA to recruit non-core nuclear envelope of kinetochore proteins (He et al. 2019; Soto et al.

Vol:. (1234567890)
13
Chromosome Res (2023) 31:18 Page 9 of 15 18

2018). Due to defective chromosome condensation, prolific. A specific question that remains unanswered
micronuclear DNA could also become trapped within is whether a correlation exists between chromosome
the cleavage furrow and cause it to regress (He et al. identity and efficiency of mis-attachment correction.
2019), resulting in the formation of a binucleate, Such a bias could explain recent reports that certain
tetraploid cell. This is relevant because tetraploidy human chromosomes are more prone to missegregate
has been shown to promote tumorigenesis (Fujiwara than others (Bochtler et al. 2019; Klaasen et al. 2022;
et al. 2005; Nguyen et al. 2009) and whole genome Worrall et al. 2018). It will be interesting to investi-
doubling is a common event during tumor evolution gate whether this bias in chromosome missegregation
(Carter et al. 2012; Zack et al. 2013). depends on a bias in formation and/or correction of
In conclusion, micronuclei arising from anaphase merotelic attachments.
lagging chromosomes represent a serious threat to It will also be important to gain a deeper under-
genomic stability. This is true even if the lagging standing of how merotelic kinetochore attachment
chromosome segregates to and forms a micronucleus contributes to cancer. It is clear that lagging chromo-
in the correct daughter cell. somes are only one type of chromosome segregation
errors found in cancer cells. It will be important to
focus on examining tumor patient tissues and other
Concluding remarks experimental systems to better understand the contri-
bution of different types of mitotic defects and their
The study that identified merotelic kinetochore inter-connections. For instance, chromosome bridges,
attachment as the underlying cause of anaphase lag- which are often seen in tumor cells (Bakhoum et al.
ging chromosomes represented an important juncture. 2011; Bolhaqueiro et al. 2019; Zaki et al. 2014), may
The mitosis community recognized the importance of arise as a result of DNA damage in a micronucleus
aneuploidy and chromosomal instability (CIN) and that originally formed from a lagging chromosome.
embarked on investigations aimed at determining the This type of temporal information cannot be obtained
contribution of anaphase lagging chromosomes to from analyzing fixed tissue samples and new strate-
CIN in cancer cells (Bakhoum et al. 2014; Thompson gies may be needed to study the evolution of cell pop-
and Compton 2008), examining what mechanisms ulations in order to understand these interdependen-
may enhance the formation of merotelic attachments cies. Such studies would untangle the many persistent
(Ganem et al. 2009; Kline-Smith et al. 2004; Silk- questions regarding the role of aneuploidy in cancer.
worth et al. 2012; Silkworth et al. 2009) or promote Indeed, although it is generally acknowledged that
their correction (Bakhoum et al. 2009b; Cimini et al. aneuploidy is a cancer hallmark, the exact role of ane-
2006; DeLuca et al. 2006; Knowlton et al. 2006), and uploidy has not been fully elucidated and how ane-
explore the long-term consequences of lagging chro- uploidy accumulates in cancer cells is still unclear.
mosomes (Crasta et al. 2012; He et al. 2019; Ly et al. Notably, the recently described link between
2019; Ly et al. 2017; Soto et al. 2018; Vazquez-Diez micronuclei and activation of the cGAS-STING path-
et al. 2016; Zhang et al. 2015). way and the evidence that activation of this pathway
What we have learned in recent years about the fate can promote tumorigenic phenotypes (Bakhoum
of lagging chromosomes and the micronuclei derived et al. 2018; Harding et al. 2017; Krupina et al. 2021;
from them reinforces the traditional view that these Maciejowski and Hatch 2020; Mackenzie et al. 2017)
structures are good biomarkers of genotoxic dam- opens up new areas of investigation and opportunities
age (Fenech et al. 2020), highlighting that this story to exploit the phenotypic traits of CIN cancers and
has come full circle. We have learned a tremendous their immune signature for therapeutic purposes.
amount of detail along the way and this will serve the Finally, there have been, over the years, reports
scientific community well as we try to fill the remain- on a potential interplay between canonical tumor
ing gaps and answer new questions. suppressor genes or oncogenes, including P53, P21,
As described earlier, there is still substantial debate MYC, K-RAS, RAF/MEK/ERK, and fidelity of mitotic
on the exact mechanisms that contribute to correction chromosome segregation, tolerance to aneuploidy,
of kinetochore mis-attachments, including merotely, and CIN (Annibali et al. 2014; Giaretti 1997; Gron-
and this area of investigation will likely continue to be roos and Lopez-Garcia 2018; Kreis et al. 2014; Littler

Vol.: (0123456789)
13
18 Page 10 of 15 Chromosome Res (2023) 31:18

et al. 2019; Perera and Venkitaraman 2016; Rizzotto Sodir NM, Masso-Valles D, Beaulieu ME, Swigart LB,
et al. 2021; Zerbib et al. 2023). However, while there Mc Gee MM, Somma MP, Nasi S, Seoane J, Evan GI,
Soucek L (2014) Myc inhibition is effective against gli-
is substantial evidence that P53 protects cells from oma and reveals a role for Myc in proficient mitosis. Nat
aneuploidy, tetraploidy, and CIN (Rizzotto et al. Commun 5:4632. https://​doi.​org/​10.​1038/​ncomm​s5632
2021), the relation between other tumor suppressor Antoccia A, Degrassi F, Battistoni A, Ciliutti P, Tanzarella C
genes or oncogenes and chromosome missegregation, (1991) In vitro micronucleus test with kinetochore stain-
ing: evaluation of test performance. Mutagenesis 6:319–
ploidy alterations, and CIN remains largely unex- 324. https://​doi.​org/​10.​1093/​mutage/​6.4.​319
plored. Future studies in this area promise to uncover Bakhoum SF, Danilova OV, Kaur P, Levy NB, Compton DA
new molecular details on the mechanisms of accurate (2011) Chromosomal instability substantiates poor prog-
vs. defective chromosome segregation, bring to light nosis in patients with diffuse large B-cell lymphoma.
Clin Cancer Res 17:7704–7711. https://​doi.​org/​10.​1158/​
pathways to CIN and CIN tolerance, and possibly 1078-​0432.​CCR-​11-​2049
inform new therapeutic strategies for cancer and other Bakhoum SF, Genovese G, Compton DA (2009a) Deviant
diseases. kinetochore microtubule dynamics underlie chromo-
somal instability. Curr Biol 19:1937–1942. https://​doi.​
Acknowledgements I would like to acknowledge all mem- org/​10.​1016/j.​cub.​2009.​09.​055
bers of the Cimini lab for helpful discussions and careful read- Bakhoum SF, Kabeche L, Wood MD, Laucius CD, Qu D,
ing of the manuscript. Laughney AM, Reynolds GE, Louie RJ, Phillips J, Chan
DA, Zaki BI, Murnane JP, Petritsch C, Compton DA
(2015) Numerical chromosomal instability mediates sus-
Authors’ contributions DC conceived the idea, performed ceptibility to radiation treatment. Nat Commun 6:5990.
the literature search, wrote the manuscript, and prepared the https://​doi.​org/​10.​1038/​ncomm​s6990
figures. Bakhoum SF, Ngo B, Laughney AM, Cavallo JA, Murphy CJ,
Ly P, Shah P, Sriram RK, Watkins TBK, Taunk NK,
Funding Work in the Cimini lab is supported by grant Duran M, Pauli C, Shaw C, Chadalavada K, Rajasekhar
#R01GM140042 from the U.S. National Institutes of Health. VK, Genovese G, Venkatesan S, Birkbak NJ, McGrana-
Additional funding is provided by a Dean’s Discovery Fund han N, Lundquist M, LaPlant Q, Healey JH, Elemento O,
award to DC from the Virginia Tech College of Science. Chung CH, Lee NY, Imielenski M, Nanjangud G, Pe’er
D, Cleveland DW, Powell SN, Lammerding J, Swanton
Data availability No original data or material are reported in C, Cantley LC (2018) Chromosomal instability drives
this review article. metastasis through a cytosolic DNA response. Nature
553:467–472. https://​doi.​org/​10.​1038/​natur​e25432
Declarations Bakhoum SF, Silkworth WT, Nardi IK, Nicholson JM, Comp-
ton DA, Cimini D (2014) The mitotic origin of chromo-
somal instability. Curr Biol 24:R148-149. https://​doi.​org/​
Ethical approval/consents This is a review article. As such, 10.​1016/j.​cub.​2014.​01.​019
no original research involving humans and/or animals is pre- Bakhoum SF, Thompson SL, Manning AL, Compton DA
sented. (2009b) Genome stability is ensured by temporal con-
trol of kinetochore-microtubule dynamics. Nat Cell Biol
Competing interests The author has no competing interests 11:27–35. https://​doi.​org/​10.​1038/​ncb18​09
or other interests that might be perceived to influence the dis- Baudoin NC, Cimini D (2018a) Chromosome Segregation:
cussion reported in this paper. The Bigger They Come, the Harder They Fall. Curr Biol
28:R665–R667. https://​doi.​org/​10.​1016/j.​cub.​2018.​04.​036
Baudoin NC, Cimini D (2018b) A guide to classifying mitotic
stages and mitotic defects in fixed cells. Chromosoma
127:215–227. https://​doi.​org/​10.​1007/​s00412-​018-​0660-2
References Biggins S, Severin FF, Bhalla N, Sassoon I, Hyman AA, Mur-
ray AW (1999) The conserved protein kinase Ipl1 regu-
Akiyoshi B, Sarangapani KK, Powers AF, Nelson CR, Reichow lates microtubule binding to kinetochores in budding
SL, Arellano-Santoyo H, Gonen T, Ranish JA, Asbury yeast. Genes Dev 13:532–544. https://​doi.​org/​10.​1101/​
CL, Biggins S (2010) Tension directly stabilizes recon- gad.​13.5.​532
stituted kinetochore-microtubule attachments. Nature Bochtler T, Kartal-Kaess M, Granzow M, Hielscher T, Cosenza
468:576–579. https://​doi.​org/​10.​1038/​natur​e09594 MR, Herold-Mende C, Jauch A, Kramer A (2019) Micro-
Andrews PD, Ovechkina Y, Morrice N, Wagenbach M, Duncan nucleus formation in human cancer cells is biased by
K, Wordeman L, Swedlow JR (2004) Aurora B regulates chromosome size. Genes Chromosomes Cancer 58:392–
MCAK at the mitotic centromere. Dev Cell 6:253–268. 395. https://​doi.​org/​10.​1002/​gcc.​22707
https://​doi.​org/​10.​1016/​s1534-​5807(04)​00025-5 Bolhaqueiro ACF, Ponsioen B, Bakker B, Klaasen SJ, Kucuk-
Annibali D, Whitfield JR, Favuzzi E, Jauset T, Serrano E, Cuar- kose E, van Jaarsveld RH, Vivie J, Verlaan-Klink I, Hami
tas I, Redondo-Campos S, Folch G, Gonzalez-Junca A, N, Spierings DCJ, Sasaki N, Dutta D, Boj SF, Vries RGJ,

Vol:. (1234567890)
13
Chromosome Res (2023) 31:18 Page 11 of 15 18

Lansdorp PM, van de Wetering M, van Oudenaarden Ciferri C, Pasqualato S, Screpanti E, Varetti G, Santaguida
A, Clevers H, Kranenburg O, Foijer F, Snippert HJG, S, Dos Reis G, Maiolica A, Polka J, De Luca JG, De
Kops G (2019) Ongoing chromosomal instability and Wulf P, Salek M, Rappsilber J, Moores CA, Salmon
karyotype evolution in human colorectal cancer orga- ED, Musacchio A (2008) Implications for kinetochore-
noids. Nat Genet 51:824–834. https://​doi.​org/​10.​1038/​ microtubule attachment from the structure of an engi-
s41588-​019-​0399-6 neered Ndc80 complex. Cell 133:427–439. https://​doi.​
Bonassi S, Coskun E, Ceppi M, Lando C, Bolognesi C, Burgaz org/​10.​1016/j.​cell.​2008.​03.​020
S, Holland N, Kirsh-Volders M, Knasmueller S, Zeiger Cimini D, Antoccia A, Tanzarella C, Degrassi F (1997)
E, Carnesoltas D, Cavallo D, da Silva J, de Andrade VM, Topoisomerase II inhibition in mitosis produces
Demircigil GC, Dominguez Odio A, Donmez-Altuntas H, numerical and structural chromosomal aberrations in
Gattas G, Giri A, Giri S, Gomez-Meda B, Gomez-Arroyo human fibroblasts. Cytogenet Cell Genet 76:61–67.
S, Hadjidekova V, Haveric A, Kamboj M, Kurteshi K, Mar- https://​doi.​org/​10.​1159/​00013​4517
tino-Roth MG, Montero Montoya R, Nersesyan A, Pastor- Cimini D, Cameron LA, Salmon ED (2004) Anaphase spin-
Benito S, Favero Salvadori DM, Shaposhnikova A, Stopper dle mechanics prevent mis-segregation of meroteli-
H, Thomas P, Torres-Bugarin O, Yadav AS, Zuniga Gonza- cally oriented chromosomes. Curr Biol 14:2149–2155.
lez G, Fenech M (2011a) The HUman MicroNucleus pro- https://​doi.​org/​10.​1016/j.​cub.​2004.​11.​029
ject on eXfoLiated buccal cells (HUMN(XL)): the role of Cimini D, Fioravanti D, Salmon ED, Degrassi F (2002)
life-style, host factors, occupational exposures, health status, Merotelic kinetochore orientation versus chromosome
and assay protocol. Mutat Res 728:88–97. https://​doi.​org/​ mono-orientation in the origin of lagging chromo-
10.​1016/j.​mrrev.​2011.​06.​005 somes in human primary cells. J Cell Sci 115:507–515.
Bonassi S, El-Zein R, Bolognesi C, Fenech M (2011b) Micro- https://​doi.​org/​10.​1242/​jcs.​115.3.​507
nuclei frequency in peripheral blood lymphocytes and Cimini D, Howell B, Maddox P, Khodjakov A, Degrassi F,
cancer risk: evidence from human studies. Mutagenesis Salmon ED (2001) Merotelic kinetochore orientation is
26:93–100. https://​doi.​org/​10.​1093/​mutage/​geq075 a major mechanism of aneuploidy in mitotic mamma-
Bonatti S, Cavalieri Z, Viaggi S, Abbondandolo A (1992) The lian tissue cells. J Cell Biol 153:517–527. https://​doi.​
analysis of 10 potential spindle poisons for their ability org/​10.​1083/​jcb.​153.3.​517
to induce CREST-positive micronuclei in human diploid Cimini D, Moree B, Canman JC, Salmon ED (2003) Mero-
fibroblasts. Mutagenesis 7:111–114. https://​doi.​org/​10.​ telic kinetochore orientation occurs frequently dur-
1093/​mutage/​7.2.​111 ing early mitosis in mammalian tissue cells and error
Brinkley BR (2001) Managing the centrosome numbers game: correction is achieved by two different mechanisms. J
from chaos to stability in cancer cell division. Trends Cell Sci 116:4213–4225. https://​doi.​org/​10.​1242/​jcs.​
Cell Biol 11:18–21. https://​doi.​org/​10.​1016/​s0962-​ 00716
8924(00)​01872-9 Cimini D, Tanzarella C, Degrassi F (1999) Differences in
Broad AJ, DeLuca JG (2020) The right place at the right time: malsegregation rates obtained by scoring ana-telo-
Aurora B kinase localization to centromeres and kineto- phases or binucleate cells. Mutagenesis 14:563–568.
chores. Essays Biochem 64:299–311. https://​doi.​org/​10.​ https://​doi.​org/​10.​1093/​mutage/​14.6.​563
1042/​EBC20​190081 Cimini D, Wan X, Hirel CB, Salmon ED (2006) Aurora kinase
Carter SL, Cibulskis K, Helman E, McKenna A, Shen H, Zack promotes turnover of kinetochore microtubules to reduce
T, Laird PW, Onofrio RC, Winckler W, Weir BA, Ber- chromosome segregation errors. Curr Biol 16:1711–
oukhim R, Pellman D, Levine DA, Lander ES, Meyer- 1718. https://​doi.​org/​10.​1016/j.​cub.​2006.​07.​022
son M, Getz G (2012) Absolute quantification of somatic Cortes-Ciriano I, Lee JJ, Xi R, Jain D, Jung YL, Yang L, Gor-
DNA alterations in human cancer. Nat Biotechnol denin D, Klimczak LJ, Zhang CZ, Pellman DS, Group
30:413–421. https://​doi.​org/​10.​1038/​nbt.​2203 PSVW, Park PJ, Consortium P (2020) Comprehensive
Catalan J, Falck GC, Norppa H (2000) The X chromosome analysis of chromothripsis in 2,658 human cancers using
frequently lags behind in female lymphocyte anaphase. whole-genome sequencing. Nat Genet 52:331–341.
Am J Hum Genet 66:687–691. https://​doi.​org/​10.​1086/​ https://​doi.​org/​10.​1038/​s41588-​019-​0576-7
302769 Crasta K, Ganem NJ, Dagher R, Lantermann AB, Ivanova
Cheeseman IM, Chappie JS, Wilson-Kubalek EM, Desai A EV, Pan Y, Nezi L, Protopopov A, Chowdhury D, Pell-
(2006) The conserved KMN network constitutes the man D (2012) DNA breaks and chromosome pulveriza-
core microtubule-binding site of the kinetochore. Cell tion from errors in mitosis. Nature 482:53–58. https://​
127:983–997. https://​doi.​org/​10.​1016/j.​cell.​2006.​09.​039 doi.​org/​10.​1038/​natur​e10802
Chen GY, Renda F, Zhang H, Gokden A, Wu DZ, Chenoweth de Regt AK, Clark CJ, Asbury CL, Biggins S (2022) Tension
DM, Khodjakov A, Lampson MA (2021) Tension pro- can directly suppress Aurora B kinase-triggered release
motes kinetochore-microtubule release by Aurora B of kinetochore-microtubule attachments. Nat Commun
kinase. J Cell Biol 22010.1083/jcb.202007030 13:2152. https://​doi.​org/​10.​1038/​s41467-​022-​29542-8
Cheng JM, Li J, Tang JX, Hao XX, Wang ZP, Sun TC, Wang Degrassi F, Tanzarella C (1988) Immunofluorescent staining of
XX, Zhang Y, Chen SR, Liu YX (2017) Merotelic kinetochores in micronuclei: a new assay for the detec-
kinetochore attachment in oocyte meiosis II causes tion of aneuploidy. Mutat Res 203:339–345. https://​doi.​
sister chromatids segregation errors in aged mice. Cell org/​10.​1016/​0165-​1161(88)​90030-1
Cycle 16:1404–1413. https://​doi.​org/​10.​1080/​15384​ DeLuca JG, Gall WE, Ciferri C, Cimini D, Musacchio A,
101.​2017.​13274​88 Salmon ED (2006) Kinetochore microtubule dynamics

Vol.: (0123456789)
13
18 Page 12 of 15 Chromosome Res (2023) 31:18

and attachment stability are regulated by Hec1. Cell Nasmyth K (2007) The kinetochore proteins Pcs1 and
127:969–982. https://​doi.​org/​10.​1016/j.​cell.​2006.​09.​047 Mde4 and heterochromatin are required to prevent mero-
Deng C, Ya A, Compton DA, Godek KM (2023) A pluripo- telic orientation. Curr Biol 17:1190–1200. https://​doi.​
tent developmental state confers a low fidelity of chro- org/​10.​1016/j.​cub.​2007.​06.​044
mosome segregation. Stem Cell Reports 18:475–488. Gronroos E, Lopez-Garcia C (2018) Tolerance of Chromo-
https://​doi.​org/​10.​1016/j.​stemcr.​2022.​12.​008 somal Instability in Cancer: Mechanisms and Therapeu-
Drpic D, Almeida AC, Aguiar P, Renda F, Damas J, Lewin tic Opportunities. Cancer Res 78:6529–6535. https://​doi.​
HA, Larkin DM, Khodjakov A, Maiato H (2018) Chro- org/​10.​1158/​0008-​5472.​CAN-​18-​1958
mosome Segregation Is Biased by Kinetochore Size. Gustavino B, Degrassi F, Filipponi R, Modesti D, Tanzarella C,
Curr Biol 28:1344-1356 e1345. https://​doi.​org/​10.​1016/j.​ Rizzoni M (1994) Mitotic indirect non-disjunction in phyto-
cub.​2018.​03.​023 hemagglutinin stimulated human lymphocytes. Mutagenesis
Falck GC, Catalan J, Norppa H (2002) Nature of anaphase 9:17–21. https://​doi.​org/​10.​1093/​mutage/​9.1.​17
laggards and micronuclei in female cytokinesis-blocked Harding SM, Benci JL, Irianto J, Discher DE, Minn AJ, Green-
lymphocytes. Mutagenesis 17:111–117. https://​doi.​org/​ berg RA (2017) Mitotic progression following DNA dam-
10.​1093/​mutage/​17.2.​111 age enables pattern recognition within micronuclei. Nature
Fenech M (2000) The in vitro micronucleus technique. Mutat Res 548:466–470. https://​doi.​org/​10.​1038/​natur​e23470
455:81–95. https://​doi.​org/​10.​1016/​s0027-​5107(00)​00065-8 Hatch EM, Fischer AH, Deerinck TJ, Hetzer MW (2013) Cata-
Fenech M, Knasmueller S, Bolognesi C, Holland N, Bonassi strophic nuclear envelope collapse in cancer cell micronuclei.
S, Kirsch-Volders M (2020) Micronuclei as biomarkers Cell 154:47–60. https://​doi.​org/​10.​1016/j.​cell.​2013.​06.​007
of DNA damage, aneuploidy, inducers of chromosomal He B, Gnawali N, Hinman AW, Mattingly AJ, Osimani A,
hypermutation and as sources of pro-inflammatory Cimini D (2019) Chromosomes missegregated into
DNA in humans. Mutat Res Rev Mutat Res 786:108342. micronuclei contribute to chromosomal instability by
https://​doi.​org/​10.​1016/j.​mrrev.​2020.​108342 missegregating at the next division. Oncotarget 10:2660–
Fenech M, Morley AA (1989) Kinetochore detection in micro- 2674. https://​doi.​org/​10.​18632/​oncot​arget.​26853
nuclei: an alternative method for measuring chromosome Hennig UG, Rudd NL, Hoar DI (1988) Kinetochore immu-
loss. Mutagenesis 4:98–104. https://​doi.​org/​10.​1093/​ nofluorescence in micronuclei: a rapid method for the
mutage/​4.2.​98 in situ detection of aneuploidy and chromosome break-
Ferrandiz N, Downie L, Starling GP, Royle SJ (2022) Endo- age in human fibroblasts. Mutat Res 203:405–414.
membranes promote chromosome missegregation by https://​doi.​org/​10.​1016/​0165-​1161(88)​90013-1
ensheathing misaligned chromosomes. J Cell Biol Hoffelder DR, Luo L, Burke NA, Watkins SC, Gollin SM,
22110.1083/jcb.202203021 Saunders WS (2004) Resolution of anaphase bridges in
Fujiwara T, Bandi M, Nitta M, Ivanova EV, Bronson RT, cancer cells. Chromosoma 112:389–397. https://​doi.​org/​
Pellman D (2005) Cytokinesis failure generating tetra- 10.​1007/​s00412-​004-​0284-6
ploids promotes tumorigenesis in p53-null cells. Nature Hogstedt B, Karlsson A (1985) The size of micronuclei in
437:1043–1047 human lymphocytes varies according to inducing agent
Ganem NJ, Godinho SA, Pellman D (2009) A mechanism link- used. Mutat Res 156:229–232. https://​doi.​org/​10.​1016/​
ing extra centrosomes to chromosomal instability. Nature 0165-​1218(85)​90067-9
460:278–282. https://​doi.​org/​10.​1038/​natur​e08136 Holubcova Z, Blayney M, Elder K, Schuh M (2015) Human
Giaretti W (1997) Aneuploidy mechanisms in human colorec- oocytes. Error-prone chromosome-mediated spindle
tal preneoplastic lesions and Barrett’s esophagus. Is there assembly favors chromosome segregation defects in
a role for K-ras and p53 mutations? Anal Cell Pathol human oocytes. Science 348:1143–1147. https://​doi.​org/​
15:99–117. https://​doi.​org/​10.​1155/​1997/​264135 10.​1126/​scien​ce.​aaa95​29
Gisselsson D, Bjork J, Hoglund M, Mertens F, Dal Cin P, Aker- Janicke MA, LaFountain JR Jr (1984) Malorientation in half-
man M, Mandahl N (2001) Abnormal nuclear shape in solid bivalents at anaphase: analysis of autosomal laggards
tumors reflects mitotic instability. Am J Pathol 158:199– in untreated, cold-treated, and cold-recovering crane fly
206. https://​doi.​org/​10.​1016/​S0002-​9440(10)​63958-2 spermatocytes. J Cell Biol 98:859–869. https://​doi.​org/​
Gisselsson D, Palsson E, Yu C, Mertens F, Mandahl N (2004) 10.​1083/​jcb.​98.3.​859
Mitotic instability associated with late genomic changes Janicke MA, Lasko L, Oldenbourg R, LaFountain JR Jr (2007)
in bone and soft tissue tumours. Cancer Lett 206:69–76. Chromosome malorientations after meiosis II arrest
https://​doi.​org/​10.​1016/j.​canlet.​2003.​10.​022 cause nondisjunction. Mol Biol Cell 18:1645–1656.
Gomes AM, Orr B, Novais-Cruz M, De Sousa F, Macario-Mon- https://​doi.​org/​10.​1091/​mbc.​e06-​10-​0963
teiro J, Lemos C, Ferras C, Maiato H (2022) Micronuclei Kaseda K, McAinsh AD, Cross RA (2012) Dual pathway spindle
from misaligned chromosomes that satisfy the spindle assembly increases both the speed and the fidelity of mitosis.
assembly checkpoint in cancer cells. Curr Biol 32:4240- Biol Open 1:12–18. https://​doi.​org/​10.​1242/​bio.​20110​12
4254 e4245. https://​doi.​org/​10.​1016/j.​cub.​2022.​08.​026 Kato H, Sandberg AA (1968) Chromosome pulverization
Gregan J, Polakova S, Zhang L, Tolic-Norrelykke IM, Cimini in human cells with micronuclei. J Natl Cancer Inst
D (2011) Merotelic kinetochore attachment: causes and 40:165–179
effects. Trends Cell Biol 21:374–381. https://​doi.​org/​10.​ Khodjakov A, Cole RW, McEwen BF, Buttle KF, Rieder CL
1016/j.​tcb.​2011.​01.​003 (1997) Chromosome fragments possessing only one
Gregan J, Riedel CG, Pidoux AL, Katou Y, Rumpf C, kinetochore can congress to the spindle equator. J Cell
Schleiffer A, Kearsey SE, Shirahige K, Allshire RC, Biol 136:229–240. https://​doi.​org/​10.​1083/​jcb.​136.2.​229

Vol:. (1234567890)
13
Chromosome Res (2023) 31:18 Page 13 of 15 18

Klaasen SJ, Truong MA, van Jaarsveld RH, Koprivec I, Sti- Ly P, Teitz LS, Kim DH, Shoshani O, Skaletsky H, Fachi-
mac V, de Vries SG, Risteski P, Kodba S, Vukusic K, netti D, Page DC, Cleveland DW (2017) Selective Y
de Luca KL, Marques JF, Gerrits EM, Bakker B, Foijer centromere inactivation triggers chromosome shatter-
F, Kind J, Tolic IM, Lens SMA, Kops G (2022) Nuclear ing in micronuclei and repair by non-homologous end
chromosome locations dictate segregation error fre- joining. Nat Cell Biol 19:68–75. https://​doi.​org/​10.​
quencies. Nature 607:604–609. https://​doi.​org/​10.​1038/​ 1038/​ncb34​50
s41586-​022-​04938-0 Maciejowski J, Hatch EM (2020) Nuclear Membrane Rup-
Kline-Smith SL, Khodjakov A, Hergert P, Walczak CE (2004) ture and Its Consequences. Annu Rev Cell Dev Biol
Depletion of centromeric MCAK leads to chromosome 36:85–114. https://​doi.​org/​10.​1146/​annur​ev-​cellb​
congression and segregation defects due to improper io-​020520-​120627
kinetochore attachments. Mol Biol Cell 15:1146–1159. Maciejowski J, Li Y, Bosco N, Campbell PJ, de Lange T (2015)
https://​doi.​org/​10.​1091/​mbc.​e03-​08-​0581 Chromothripsis and Kataegis Induced by Telomere Cri-
Knowlton AL, Lan W, Stukenberg PT (2006) Aurora B is sis. Cell 163:1641–1654. https://​doi.​org/​10.​1016/j.​cell.​
enriched at merotelic attachment sites, where it regulates 2015.​11.​054
MCAK. Curr Biol 16:1705–1710. https://​doi.​org/​10.​ Mackenzie KJ, Carroll P, Martin CA, Murina O, Fluteau A,
1016/j.​cub.​2006.​07.​057 Simpson DJ, Olova N, Sutcliffe H, Rainger JK, Leitch A,
Kouznetsova A, Hernandez-Hernandez A, Hoog C (2014) Osborn RT, Wheeler AP, Nowotny M, Gilbert N, Chan-
Merotelic attachments allow alignment and stabiliza- dra T, Reijns MAM, Jackson AP (2017) cGAS surveil-
tion of chromatids in meiosis II oocytes. Nat Commun lance of micronuclei links genome instability to innate
5:4409. https://​doi.​org/​10.​1038/​ncomm​s5409 immunity. Nature 548:461–465. https://​doi.​org/​10.​1038/​
Kouznetsova A, Kitajima TS, Brismar H, Hoog C (2019) Post- natur​e23449
metaphase correction of aberrant kinetochore-micro- Maddox P, Straight A, Coughlin P, Mitchison TJ, Salmon ED
tubule attachments in mammalian eggs. EMBO Rep (2003) Direct observation of microtubule dynamics at
20:e47905. https://​doi.​org/​10.​15252/​embr.​20194​7905 kinetochores in Xenopus extract spindles: implications
Kreis NN, Sanhaji M, Rieger MA, Louwen F, Yuan J (2014) for spindle mechanics. J Cell Biol 162:377–382. https://​
p21Waf1/Cip1 deficiency causes multiple mitotic defects doi.​org/​10.​1083/​jcb.​20030​1088
in tumor cells. Oncogene 33:5716–5728. https://​doi.​org/​ Maiato H, Silva S (2023) Double-checking chromosome segre-
10.​1038/​onc.​2013.​518 gation. J Cell Biol 22210.1083/jcb.202301106
Krupina K, Goginashvili A, Cleveland DW (2021) Causes Mammel AE, Huang HZ, Gunn AL, Choo E, Hatch EM
and consequences of micronuclei. Curr Opin Cell Biol (2022) Chromosome length and gene density contribute
70:91–99. https://​doi.​org/​10.​1016/j.​ceb.​2021.​01.​004 to micronuclear membrane stability. Life Sci Alliance
Ladrach KS, LaFountain JR Jr (1986) Malorientation and 510.26508/lsa.202101210
abnormal segregation of chromosomes during recovery Maney T, Hunter AW, Wagenbach M, Wordeman L (1998)
from colcemid and nocodazole. Cell Motil Cytoskeleton Mitotic centromere-associated kinesin is important for
6:419–427. https://​doi.​org/​10.​1002/​cm.​97006​0407 anaphase chromosome segregation. J Cell Biol 142:787–
Lan W, Zhang X, Kline-Smith SL, Rosasco SE, Barrett-Wilt 801. https://​doi.​org/​10.​1083/​jcb.​142.3.​787
GA, Shabanowitz J, Hunt DF, Walczak CE, Stukenberg Mihajlovic AI, Haverfield J, FitzHarris G (2021) Distinct
PT (2004) Aurora B phosphorylates centromeric MCAK classes of lagging chromosome underpin age-related
and regulates its localization and microtubule depolym- oocyte aneuploidy in mouse. Dev Cell 56:2273-2283
erization activity. Curr Biol 14:273–286. https://​doi.​org/​ e2273. https://​doi.​org/​10.​1016/j.​devcel.​2021.​07.​022
10.​1016/j.​cub.​2004.​01.​055 Moroi Y, Peebles C, Fritzler MJ, Steigerwald J, Tan EM (1980)
Lara-Gonzalez P, Pines J, Desai A (2021) Spindle assem- Autoantibody to centromere (kinetochore) in sclero-
bly checkpoint activation and silencing at kinetochores. derma sera. Proc Natl Acad Sci U S A 77:1627–1631.
Semin Cell Dev Biol 117:86–98. https://​doi.​org/​10.​ https://​doi.​org/​10.​1073/​pnas.​77.3.​1627
1016/j.​semcdb.​2021.​06.​009 Nam HJ, van Deursen JM (2014) Cyclin B2 and p53 control
Lengauer C, Kinzler KW, Vogelstein B (1997) Genetic insta- proper timing of centrosome separation. Nat Cell Biol
bility in colorectal cancers. Nature 386:623–627. https://​ 16:538–549. https://​doi.​org/​10.​1038/​ncb29​52
doi.​org/​10.​1038/​38662​3a0 Nguyen HG, Makitalo M, Yang D, Chinnappan D, St Hilaire
Littler S, Sloss O, Geary B, Pierce A, Whetton AD, Taylor SS C, Ravid K (2009) Deregulated Aurora-B induced tetra-
(2019) Oncogenic MYC amplifies mitotic perturbations. ploidy promotes tumorigenesis. FASEB J 23:2741–2748.
Open Biol 9:190136. https://​doi.​org/​10.​1098/​rsob.​190136 https://​doi.​org/​10.​1096/​fj.​09-​130963
Liu S, Kwon M, Mannino M, Yang N, Renda F, Khodjakov A, Nicholson JM, Cimini D (2013) Cancer karyotypes: survival
Pellman D (2018) Nuclear envelope assembly defects of the fittest. Front Oncol 3:148. https://​doi.​org/​10.​3389/​
link mitotic errors to chromothripsis. Nature 561:551– fonc.​2013.​00148
555. https://​doi.​org/​10.​1038/​s41586-​018-​0534-z Orr B, De Sousa F, Gomes AM, Afonso O, Ferreira LT,
Ly P, Brunner SF, Shoshani O, Kim DH, Lan W, Pyntik- Figueiredo AC, Maiato H (2021) An anaphase surveil-
ova T, Flanagan AM, Behjati S, Page DC, Campbell lance mechanism prevents micronuclei formation from
PJ, Cleveland DW (2019) Chromosome segregation frequent chromosome segregation errors. Cell Rep
errors generate a diverse spectrum of simple and com- 37:109783. https://​doi.​org/​10.​1016/j.​celrep.​2021.​109783
plex genomic rearrangements. Nat Genet 51:705–715. Orr B, Talje L, Liu Z, Kwok BH, Compton DA (2016)
https://​doi.​org/​10.​1038/​s41588-​019-​0360-8 Adaptive Resistance to an Inhibitor of Chromosomal

Vol.: (0123456789)
13
18 Page 14 of 15 Chromosome Res (2023) 31:18

Instability in Human Cancer Cells. Cell Rep 17:1755– Mutat Res 392:81–95. https://​doi.​org/​10.​1016/​s0165-​
1763. https://​doi.​org/​10.​1016/j.​celrep.​2016.​10.​030 1218(97)​00047-5
Pampalona J, Roscioli E, Silkworth WT, Bowden B, Genesca A, Sen O, Harrison JU, Burroughs NJ, McAinsh AD (2021) Kine-
Tusell L, Cimini D (2016) Chromosome Bridges Main- tochore life histories reveal an Aurora-B-dependent error
tain Kinetochore-Microtubule Attachment throughout correction mechanism in anaphase. Dev Cell 56:3082-
Mitosis and Rarely Break during Anaphase. PLoS One 3099 e3085. https://​doi.​org/​10.​1016/j.​devcel.​2021.​10.​007
11:e0147420. https://​doi.​org/​10.​1371/​journ​al.​pone.​01474​20 Shomper M, Lappa C, FitzHarris G (2014) Kinetochore microtu-
Papini D, Levasseur MD, Higgins JMG (2021) The Aurora B gra- bule establishment is defective in oocytes from aged mice.
dient sustains kinetochore stability in anaphase. Cell Rep Cell Cycle 13:1171–1179. https://​doi.​org/​10.​4161/​cc.​28046
37:109818. https://​doi.​org/​10.​1016/j.​celrep.​2021.​109818 Silkworth WT, Cimini D (2012) Transient defects of mitotic
Paul R, Wollman R, Silkworth WT, Nardi IK, Cimini D, spindle geometry and chromosome segregation errors.
Mogilner A (2009) Computer simulations predict that Cell Div 7:19. https://​doi.​org/​10.​1186/​1747-​1028-7-​19
chromosome movements and rotations accelerate mitotic Silkworth WT, Nardi IK, Paul R, Mogilner A, Cimini D (2012)
spindle assembly without compromising accuracy. Proc Timing of centrosome separation is important for accu-
Natl Acad Sci U S A 106:15708–15713. https://​doi.​org/​ rate chromosome segregation. Mol Biol Cell 23:401–
10.​1073/​pnas.​09082​61106 411. https://​doi.​org/​10.​1091/​mbc.​E11-​02-​0095
Perera D, Venkitaraman AR (2016) Oncogenic KRAS triggers Silkworth WT, Nardi IK, Scholl LM, Cimini D (2009)
MAPK-dependent errors in mitosis and MYC-depend- Multipolar spindle pole coalescence is a major source of
ent sensitivity to anti-mitotic agents. Sci Rep 6:29741. kinetochore mis-attachment and chromosome mis-segre-
https://​doi.​org/​10.​1038/​srep2​9741 gation in cancer cells. PLoS One 4:e6564. https://​doi.​org/​
Pidoux AL, Uzawa S, Perry PE, Cande WZ, Allshire RC 10.​1371/​journ​al.​pone.​00065​64
(2000) Live analysis of lagging chromosomes during Sluder G, Thompson EA, Miller FJ, Hayes J, Rieder CL (1997)
anaphase and their effect on spindle elongation rate in The checkpoint control for anaphase onset does not mon-
fission yeast. J Cell Sci 113(Pt 23):4177–4191. https://​ itor excess numbers of spindle poles or bipolar spindle
doi.​org/​10.​1242/​jcs.​113.​23.​4177 symmetry. J Cell Sci 110(Pt 4):421–429. https://​doi.​org/​
Pincu M, Callisen H, Norman A (1985) DNA content of micro- 10.​1242/​jcs.​110.4.​421
nuclei in human lymphocytes. Int J Radiat Biol Relat Soto M, Garcia-Santisteban I, Krenning L, Medema RH,
Stud Phys Chem Med 47:423–432 Raaijmakers JA (2018) Chromosomes trapped in
Rajagopalan H, Lengauer C (2004) Aneuploidy and cancer. micronuclei are liable to segregation errors. J Cell Sci
Nature 432:338–341. https://​doi.​org/​10.​1038/​natur​e03099 13110.1242/jcs.214742
Rieder CL, Cole RW, Khodjakov A, Sluder G (1995) The Stear JH, Roth MB (2002) Characterization of HCP-6, a C.
checkpoint delaying anaphase in response to chromo- elegans protein required to prevent chromosome twist-
some monoorientation is mediated by an inhibitory sig- ing and merotelic attachment. Genes Dev 16:1498–1508.
nal produced by unattached kinetochores. J Cell Biol https://​doi.​org/​10.​1101/​gad.​989102
130:941–948. https://​doi.​org/​10.​1083/​jcb.​130.4.​941 Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR, Mudie
Rieder CL, Schultz A, Cole R, Sluder G (1994) Anaphase LJ, Pleasance ED, Lau KW, Beare D, Stebbings LA,
onset in vertebrate somatic cells is controlled by a check- McLaren S, Lin ML, McBride DJ, Varela I, Nik-Zainal S,
point that monitors sister kinetochore attachment to the Leroy C, Jia M, Menzies A, Butler AP, Teague JW, Quail
spindle. J Cell Biol 127:1301–1310. https://​doi.​org/​10.​ MA, Burton J, Swerdlow H, Carter NP, Morsberger LA,
1083/​jcb.​127.5.​1301 Iacobuzio-Donahue C, Follows GA, Green AR, Flanagan
Rizzoni M, Tanzarella C, Gustavino B, Degrassi F, Guarino AM, Stratton MR, Futreal PA, Campbell PJ (2011) Mas-
A, Vitagliano E (1989) Indirect mitotic nondisjunction sive genomic rearrangement acquired in a single cata-
in Vicia faba and Chinese hamster cells. Chromosoma strophic event during cancer development. Cell 144:27–
97:339–346. https://​doi.​org/​10.​1007/​BF003​71976 40. https://​doi.​org/​10.​1016/j.​cell.​2010.​11.​055
Rizzotto D, Englmaier L, Villunger A (2021) At a Cross- Stewenius Y, Jin Y, Ora I, de Kraker J, Bras J, Frigyesi A, Alumets
roads to Cancer: How p53-Induced Cell Fate Decisions J, Sandstedt B, Meeker AK, Gisselsson D (2007) Defec-
Secure Genome Integrity. Int J Mol Sci 2210.3390/ tive chromosome segregation and telomere dysfunction in
ijms221910883 aggressive Wilms’ tumors. Clin Cancer Res 13:6593–6602.
Rumpf C, Cipak L, Schleiffer A, Pidoux A, Mechtler K, Tolic- https://​doi.​org/​10.​1158/​1078-​0432.​CCR-​07-​1081
Norrelykke IM, Gregan J (2010) Laser microsurgery Streffer C, van Beuningen D, Rebmann A (1985) DNA meas-
provides evidence for merotelic kinetochore attachments urements and micronuclei in human rectal carcinoma.
in fission yeast cells lacking Pcs1 or Clr4. Cell Cycle Radiat Med 3:147–150
9:3997–4004. https://​doi.​org/​10.​4161/​cc.9.​19.​13233 Tanaka TU, Rachidi N, Janke C, Pereira G, Galova M, Schiebel
Sarangapani KK, Asbury CL (2014) Catch and release: how do E, Stark MJ, Nasmyth K (2002) Evidence that the Ipl1-
kinetochores hook the right microtubules during mitosis? Sli15 (Aurora kinase-INCENP) complex promotes chro-
Trends Genet 30:150–159. https://​doi.​org/​10.​1016/j.​tig.​ mosome bi-orientation by altering kinetochore-spindle
2014.​02.​004 pole connections. Cell 108:317–329. https://​doi.​org/​10.​
Schuler M, Rupa DS, Eastmond DA (1997) A critical evalu- 1016/​s0092-​8674(02)​00633-5
ation of centromeric labeling to distinguish micronuclei Terradas M, Martin M, Hernandez L, Tusell L, Genesca A
induced by chromosomal loss and breakage in vitro. (2012) Nuclear envelope defects impede a proper response

Vol:. (1234567890)
13
Chromosome Res (2023) 31:18 Page 15 of 15 18

to micronuclear DNA lesions. Mutat Res 729:35–40. https://​doi.​org/​10.​1002/​(SICI)​1097-​0169(1997)​36:3%​


https://​doi.​org/​10.​1016/j.​mrfmmm.​2011.​09.​003 3c291::​AID-​CM9%​3e3.0.​CO;2-A
Terradas M, Martin M, Tusell L, Genesca A (2009) DNA Worrall JT, Tamura N, Mazzagatti A, Shaikh N, van Lingen
lesions sequestered in micronuclei induce a local defec- T, Bakker B, Spierings DCJ, Vladimirou E, Foijer F,
tive-damage response. DNA Repair (amst) 8:1225–1234. McClelland SE (2018) Non-random Mis-segregation of
https://​doi.​org/​10.​1016/j.​dnarep.​2009.​07.​004 Human Chromosomes. Cell Rep 23:3366–3380. https://​
Thompson SL, Compton DA (2008) Examining the link doi.​org/​10.​1016/j.​celrep.​2018.​05.​047
between chromosomal instability and aneuploidy in Yamamoto KI, Kikuchi Y (1980) A comparison of diameters
human cells. J Cell Biol 180:665–672. https://​doi.​org/​10.​ of micronuclei induced by clastogens and by spindle poi-
1083/​jcb.​20071​2029 sons. Mutat Res 71:127–131. https://​doi.​org/​10.​1016/​
Thompson SL, Compton DA (2011) Chromosome missegrega- 0027-​5107(80)​90012-3
tion in human cells arises through specific types of kine- Yu HG, Dawe RK (2000) Functional redundancy in the maize
tochore-microtubule attachment errors. Proc Natl Acad meiotic kinetochore. J Cell Biol 151:131–142. https://​
Sci U S A 108:17974–17978. https://​doi.​org/​10.​1073/​ doi.​org/​10.​1083/​jcb.​151.1.​131
pnas.​11097​20108 Zack TI, Schumacher SE, Carter SL, Cherniack AD, Saksena G,
Thomson EJ, Perry PE (1988) The identification of micronucleated Tabak B, Lawrence MS, Zhsng CZ, Wala J, Mermel CH,
chromosomes: a possible assay for aneuploidy. Mutagenesis Sougnez C, Gabriel SB, Hernandez B, Shen H, Laird PW,
3:415–418. https://​doi.​org/​10.​1093/​mutage/​3.5.​415 Getz G, Meyerson M, Beroukhim R (2013) Pan-cancer
Trivedi P, Zaytsev AV, Godzi M, Ataullakhanov FI, Grish- patterns of somatic copy number alteration. Nat Genet
chuk EL, Stukenberg PT (2019) The binding of 45:1134–1140. https://​doi.​org/​10.​1038/​ng.​2760
Borealin to microtubules underlies a tension inde- Zaki BI, Suriawinata AA, Eastman AR, Garner KM, Bakhoum
pendent kinetochore-microtubule error correction path- SF (2014) Chromosomal instability portends superior
way. Nat Commun 10:682. https://​doi.​org/​10.​1038/​ response of rectal adenocarcinoma to chemoradiation
s41467-​019-​08418-4 therapy. Cancer 120:1733–1742. https://​doi.​org/​10.​1002/​
Tucker JB, Bonema SC, Garcia-Varela R, Denu RA, Hu Y, cncr.​28656
McGregor SM, Burkard ME, Weaver BA (2023) Mis- Zerbib J, Ippolito MR, Eliezer Y, De Feudis G, Reuveni E,
aligned Chromosomes are a Major Source of Chromo- Kadmon AS, Martin S, Viganò S, Leor G, Berstler J,
somal Instability in Breast Cancer. Cancer Res Commun Laue K, Cohen-Sharir Y, Scorzoni S, Vazquez F, Ben-
3:54–65. https://​doi.​org/​10.​1158/​2767-​9764.​CRC-​22-​0302 David U, Santaguida S (2023) Human aneuploid cells
Umbreit NT, Zhang CZ, Lynch LD, Blaine LJ, Cheng AM, depend on the RAF/MEK/ERK pathway for overcoming
Tourdot R, Sun L, Almubarak HF, Judge K, Mitchell TJ, increased DNA damage. bioRxiv
Spektor A, Pellman D (2020) Mechanisms generating can- Zhai Y, Kronebusch PJ, Borisy GG (1995) Kinetochore microtubule
cer genome complexity from a single cell division error. dynamics and the metaphase-anaphase transition. J Cell Biol
Science 368. https://​doi.​org/​10.​1126/​scien​ce.​aba07​12 131:721–734. https://​doi.​org/​10.​1083/​jcb.​131.3.​721
Vazquez-Diez C, Yamagata K, Trivedi S, Haverfield J, FitzHar- Zhang CZ, Spektor A, Cornils H, Francis JM, Jackson EK,
ris G (2016) Micronucleus formation causes perpetual Liu S, Meyerson M, Pellman D (2015) Chromothripsis
unilateral chromosome inheritance in mouse embryos. from DNA damage in micronuclei. Nature 522:179–184.
Proc Natl Acad Sci U S A 113:626–631. https://​doi.​org/​ https://​doi.​org/​10.​1038/​natur​e14493
10.​1073/​pnas.​15176​28112 Zhang X, Lan W, Ems-McClung SC, Stukenberg PT, Walczak
Venkatesan S, Angelova M, Puttick C, Zhai H, Caswell DR, CE (2007) Aurora B phosphorylates multiple sites on
Lu WT, Dietzen M, Galanos P, Evangelou K, Bellelli R, mitotic centromere-associated kinesin to spatially and
Lim EL, Watkins TBK, Rowan A, Teixeira VH, Zhao Y, temporally regulate its function. Mol Biol Cell 18:3264–
Chen H, Ngo B, Zalmas LP, Al Bakir M, Hobor S, Gron- 3276. https://​doi.​org/​10.​1091/​mbc.​e07-​01-​0086
roos E, Pennycuick A, Nigro E, Campbell BB, Brown Zielinska AP, Holubcova Z, Blayney M, Elder K, Schuh M
WL, Akarca AU, Marafioti T, Wu MY, Howell M, Boul- (2015) Sister kinetochore splitting and precocious disinte-
ton SJ, Bertoli C, Fenton TR, de Bruin RAM, Maya- gration of bivalents could explain the maternal age effect.
Mendoza A, Santoni-Rugiu E, Hynds RE, Gorgoulis Elife 4:e11389. https://​doi.​org/​10.​7554/​eLife.​11389
VG, Jamal-Hanjani M, McGranahan N, Harris RS, Janes
SM, Bartkova J, Bakhoum SF, Bartek J, Kanu N, Swan- Publisher’s note Springer Nature remains neutral with regard
ton C, Consortium TR (2021) Induction of APOBEC3 to jurisdictional claims in published maps and institutional
Exacerbates DNA Replication Stress and Chromosomal affiliations.
Instability in Early Breast and Lung Cancer Evolution.
Cancer Discov 11:2456–2473. https://​doi.​org/​10.​1158/​
Springer Nature or its licensor (e.g. a society or other partner)
2159-​8290.​CD-​20-​0725
holds exclusive rights to this article under a publishing
Weaver BA, Cleveland DW (2006) Does aneuploidy cause can-
agreement with the author(s) or other rightsholder(s); author
cer? Curr Opin Cell Biol 18:658–667. https://​doi.​org/​10.​
self-archiving of the accepted manuscript version of this article
1016/j.​ceb.​2006.​10.​002
is solely governed by the terms of such publishing agreement
Wise DA, Brinkley BR (1997) Mitosis in cells with unreplicated
and applicable law.
genomes (MUGs): spindle assembly and behavior of cen-
tromere fragments. Cell Motil Cytoskeleton 36:291–302.

Vol.: (0123456789)
13

You might also like