You are on page 1of 10

Review TRENDS in Cell Biology Vol.15 No.

5 May 2005

Chromosome Segregation and Aneuploidy series

Aurora kinases, aneuploidy and cancer,


a coincidence or a real link?
Régis Giet, Clotilde Petretti and Claude Prigent
CNRS UMR6061 Université de Rennes 1, Groupe Cycle Cellulaire, Equipe Labellisée LNCC, Université de Rennes 1, IFR140 GFAS,
Faculté de Médecine, 2 Avenue du Pr Léon Bernard, CS 3417, Rennes cedex, France

As Aurora kinases are overexpressed in a large number of kinetochores and tension. When these two conditions are
cancers, and ectopic expression of Aurora generates satisfied, the checkpoint signals are switched off, the
polyploid cells containing multiple centrosomes, it has chromatids separate and anaphase proceeds. In telophase,
been tempting to suggest that Aurora overexpression nuclear division occurs and the cell undergoes cytokinesis.
provokes genetic instability underlying the tumori- Finally, each daughter cell receives one set of chromo-
genesis. However, examination of the evidence suggests somes and one centrosome (Box 1).
a more complex relationship. Overexpression of Aurora-A Considering the complexity of mitosis, it is not surprising
readily transforms rat-1 and NIH3T3 cells, but not primary that there are many mitotic defects that can lead to the
cells, whereas overexpression of Aurora-B induces meta- formation of aneuploid daughter cells. An aneuploid cell
stasis after implantation of tumors in nude mice. Why do possesses an altered content of DNA (abnormal number of
polyploid cells containing abnormal centrosome numbers chromosomes). To give some examples, during mitosis, one
induced by Aurora not get eliminated at cell-cycle can easily imagine at least four ways to obtain an aneuploid
checkpoints? Does this phenotype determine the origin cell: (1) incorrect centrosome duplication during interphase
of cancer or does it only promote tumor progression? that can lead to the formation of multipolar spindles,
Would drugs against aurora family members be of any (2) improper centrosome separation, (3) defective cytokin-
help for cancer treatment? These and related questions esis and (4) chromosome mis-orientation (Figure 1).
are addressed in this review (which is part of the Progression through mitosis depends on three main
Chromosome Segregation and Aneuploidy series). regulatory mechanisms: protein localization, proteolysis
and phosphorylation. Cdk1 (cyclin-dependent kinase 1 or
p34cdc2), Plk1 (Polo-like kinase), Nek2 (NIMA-related
Introduction kinase 2) and Aurora kinases are the main mitotic protein
The main goal of a mitotic cell is to equally segregate its kinases known to date [3]. Aurora kinases are involved in
chromosomes and centrosomes between two daughter centrosome separation and maturation, spindle assembly
cells (Figure 1). As early as 1902, Theodore Boveri and stability, chromosome condensation, congression and
predicted that mitotic abnormalities could lead to genetic segregation, and cytokinesis [4]. In mammalian cells,
instability and cancer [1]. Indeed, during mitosis, a three aurora kinases have been identified, named Aurora-A,
spectacular reorganization of the cytoskeleton occurs -B and -C [3]. They possess a very conserved catalytic
that builds a bipolar microtubule spindle that assures domain and an N-terminal domain that varies in sequence
proper segregation of chromosomes. Mitosis is progres- and in length [5,6]. It is thought that the different Aurora
sively readied throughout cell-cycle progression: a series kinases localize differentially and might perform different
of precisely coordinated events must have occurred before functions during mitosis. However, all three human
the G2–M transition occurs. By the end of S-phase, the cell kinases are overexpressed in many types of cancers, in
must have duplicated its centrosome and replicated its which polyploid cells containing multiple centrosomes are
DNA. At the end of prophase, the duplicated and matured observed. This review discusses how overexpression of
centrosomes must have become separated. During pro- aurora kinases provokes the development of such a
metaphase, the two centrosomes and the chromosomes phenotype, and whether this phenotype is key to onco-
nucleate highly dynamic mitotic microtubules that genesis. Although many studies of Aurora kinases in
assemble a bipolar spindle [2]. During progression from organisms such as yeasts, Drosophila, Caenorhabditis
prometaphase to metaphase, the chromosomes must elegans and Xenopus have greatly contributed to our
become bi-orientated and aligned at the metaphase understanding of the functions of the kinases, we will only
plate. Bi-orientation is achieved by microtubule-organized use here the nomenclature Aurora-A, -B and -C and the
attachment of kinetochore pairs to opposite centrosomes. names of mammalian proteins whenever possible.
During this process, the mitotic checkpoint is continu-
ously activated; it controls microtubule attachment to the Aurora-A: a non residential centrosome kinase
Corresponding author: Prigent, C. (claude.prigent@univ-rennes1.fr). Aurora-A kinase localizes on duplicated centrosomes from
Available online 2 April 2005 the end of S phase to the beginning of the following G1
www.sciencedirect.com 0962-8924/$ - see front matter Q 2005 Elsevier Ltd. All rights reserved. doi:10.1016/j.tcb.2005.03.004
242 Review TRENDS in Cell Biology Vol.15 No.5 May 2005

2N
G1

2<N<4 S

4N 4N

G2

4N 4N

2N
G1
Three One One Two
aneuploid polyploid 2N polyploid aneuploid
cells cell cell cells
1 2 3 4
2 DIPLOID
CELLS

TRENDS in Cell Biology

Figure 1. Depiction of four examples of events that can lead to loss of normal ploidy while passing through mitosis (red frames). (1) Centrosome amplification giving rise to
more than two centrosomes induces multipolar spindles, leading to aneuploid cells; (2) a defect in separation of centrosomes triggers formation of a monopolar spindle and
abortive mitosis, leading to a tetraploid cell, just like a cytokinesis defect (3); and, finally, (4) improper kinetochore–microtubule attachment gives rise to aneuploid cells.

phase [6,7]. The kinase is then degraded by the protea- Association with the substrate not only triggers autophos-
some in a Cdh1-dependent manner [8,9]. During mitosis, phorylation but also protects the kinase from dephos-
the protein also localizes to the poles of the mitotic spindle phorylation by a type 1 phosphatase that associates with
(Box 1). Although these localizations are most commonly the kinase [17] (Figure 2). To date, TPX2 is the best-
observed, it seems that the centrosomal Aurora-A kinase understood protein that activates Aurora-A. TPX2 trans-
exchanges rapidly with an apparently large cytoplasmic ports Aurora-A and activates it at the spindle poles
pool of Aurora-A [10]. [15,16,18]. It should be noted that TPX2 is also an Aurora-A
The catalytic activity of Aurora-A kinase is closely substrate. A comparable mechanism has been described
related to the activity of the mitotic centrosomes, which in G2 phase, where the protein activating Aurora-A is
participate in assembly of the bipolar spindle. The main Ajuba [19]. Among the Aurora-A substrates are Eg5
function of Aurora-A is to assist in the maturation of (a kinesin-like protein involved centrosome separation
duplicated centrosomes by recruiting proteins such as and spindle stability), CDC25B (a phosphatase activating
D-TACC (Drosophila), g-tubulin, SPD-2 (C. elegans), Cdk1–cyclinB), the tumor-suppressor p53 and BRCA-1
centrosomin and chTOG (MAP215-Msps-Zyg9) [10–14] [20] [21–24]. Aurora-A functioning as a centrosome kinase
and consequently to participate in spindle assembly and might also be involved in the asymmetric protein
stability. Remarkably for a member of the protein kinase localization during cell division that contributes to the
family, Aurora-A needs to associate with some of its determination of cell fate [10]. However, the most unusual
substrate to be activated by autophosphorylation [15,16]. function of the kinase was described by a laboratory
www.sciencedirect.com
Review TRENDS in Cell Biology Vol.15 No.5 May 2005 243

Box 1. The cell cycle and aurora kinases


The eukaryotic cell cycle is divided into four phases (Figure Ia): G1, S, the beginning of the following G1 phase. The kinase spreads on the
G2 and M (mitosis) that end with cytokinesis, during which two spindle poles during metaphase until the end of anaphase and
daughter cells containing one centrosome and the original number of remains at the centrosome in the beginning of G1, becoming
chromosomes separate. Five sub-phases divide mitosis: prophase, undetectable in mid-G1.
prometaphase, metaphase, anaphase and telophase. A crucial mitotic Both Aurora-B and overexpressed Aurora-C show the same
event occurs in metaphase when the bipolar spindle assembles. localization (on the bottom panel). Both kinases are chromosome
Faithful chromosome segregation indeed depends on chromosome passenger proteins; they appear in the nucleus in prophase, localize to
bi-orientation and alignment within this spindle. (b) Aurora kinases (in kinetochores during prometaphase and metaphase and relocalize to
red) have different localizations during mitosis. Aurora-A (on the top the equatorial spindle in anaphase and telophase. Finally, they localize
panel) localizes on duplicated centrosomes from the end of S phase to on the contractile ring on the midbody during cytokinesis.

(a) (b)
Metaphase Anaphase

se
ha

is
se

e
ap

se

es
se

as
ha

ha
et

in
Prometaphase

ha

ph
ap
om

ok
ap
op

lo
et

yt
2

An
Pr

Te
Pr

M
G

C
Telophase

Prophase
M Cytokinesis
Aurora-A

G2
Aurora-B or overexpressed Aurora-C

G1
S
TRENDS in Cell Biology

Figure I.

Type 1
phosphatase

Activator
Active Aurora
ATP P ATP
kinase

Inactive Aurora
kinase

Substrates
P
Phosphorylated
substrates
P
P
P ATP P

P
P

TRENDS in Cell Biology

Figure 2. Ways of regulating Aurora kinase. The Aurora kinase (blue) seems to continuously associate with a type 1 phosphatase (green) that keeps the activating threonine in
the T-loop in a dephosphorylated form. In the presence of an activator (pink) that could be a substrate, the phosphatase is pushed away and Aurora autophosphorylates (‘P’)
the activating threonine (or alternatively the activator). As long as the kinase remains bound to its activator, it can phosphorylate many other substrates (purple). Once the
activator leaves the kinase, the phosphatase dephosphorylates the activating threonine, and the kinase becomes inactive.

www.sciencedirect.com
244 Review TRENDS in Cell Biology Vol.15 No.5 May 2005

Intermediate complexes?

INCENP INCENP INCENP


P
P P P
Survivin Survivin Survivin Borealin

Aurora-B Aurora-B Aurora-B Aurora-B

P P P P

Complex # 1 Complex # 2

P
P
Survivin Borealin

Aurora-B

Speculative complex
TRENDS in Cell Biology

Figure 3. Aurora-B complexes. Aurora-B (red) forms different complexes in vivo. The kinase can be activated by the inner centromere protein INCENP that is also a substrate
(blue); this heterodimer has been found in vivo and could be responsible for the phosphorylation of histone H3 (complex #1). Survivin (brown) can also activate the kinase
in vitro, but it is not a substrate. Also Survivin–INCENP–Aurora-B complexes can form in vitro, but these might only be intermediate complexes. Indeed, a fourth protein
participates in the complexes in vivo – the protein Borealin – which is also an Aurora-B substrate but not an activator. This large complex could be involved in the spindle
checkpoint (complex #2). Finally, it seems that the large complex might contain another, as yet unidentified, protein (‘speculative complex’).

working on RNA translation controls: Aurora-A can phos- kinase is required for cytokinesis [37–39]. But Aurora-B
phorylate CPEB (cytoplasmic polyadenylation element fulfils at least three distinct related functions. Aurora-B is
binding protein), required for RNA polyadenylation and a histone kinase, it is a spindle checkpoint kinase and a
translation during Xenopus oocyte maturation [25,26]. cytokinesis kinase.
Aurora-B is indeed responsible for the mitotic phos-
Aurora-B: the chromosome passenger kinase phorylation of not only the celebrated Ser10 residue in the
The Aurora-B kinase belongs to the chromosome passenger tail of histone H3 [39–42] but also of Ser28 in H3 [43] and
protein family (Box 1). Such proteins localize to the of Ser7 in CENP-A [44]. These phosphorylation events are
kinetochores from prophase to metaphase and to the thought to be necessary for chromosome condensation,
central spindle and the midbody in cytokinesis [6]. although the only direct evidence comes from Tetrahymena
Association of Aurora-B with the inner centromere protein [45]. It is quite likely, however, that these phosphorylations
INCENP led to the conclusion that Aurora-B participates might only initiate the recruitment of condensation and
in a ‘chromosome passenger complex’ [27,28]. In human segregation proteins on chromosomes [39,46].
cells, INCENP is required to localize Aurora-B at the Most interestingly, Aurora-B phosphorylates the micro-
kinetochores [27]. And, just like Aurora-A, Aurora-B tubule depolymerase MCAK (mitotic centromere-associ-
activation is triggered by autophosphorylation after asso- ated kinesin) [47]. This phosphorylation by aurora-B has
ciation with its substrate INCENP (Figure 2) [29–31]. In two effects: it inactivates the microtubule depolymerase
mammalian cells, Aurora-B is a component of a large catalytic activity of MCAK and it targets MCAK to the
complex containing INCENP, survivin and Borealin that kinetochores [47–49]. The microtubule depolymerase
forms a chromosome passenger complex [32,33]. Although activity of MCAK can then be locally activated by a
survivin is not an Aurora-B substrate, it associates with phosphatase, presumably the human homolog of cdc14
the kinase and stimulates its activity [34–36]. Borealin, [50] or some other MCAK activator, to correct any
however, is an Aurora-B substrate but not a kinase incorrect kinetochore attachments to the spindle [47]. In
activator. To summarize, Aurora-B seems to participate the absence of Aurora-B, MCAK does not localize to
in at least two different complexes: an Aurora-B– kinetochores and incorrect attachments persist (Box 2).
INCENP–Survivin–Borealin complex and an Aurora-B– During anaphase and telophase, Aurora-B phosphoryl-
INCENP complex (Figure 3). ates Ser72 in vimentin, which is an intermediate filament
So, what is the function of Aurora-B? Elimination of the protein participating in formation of the cleavage furrow.
kinase by RNA interference clearly revealed that the In the absence of Ser72 phosphorylation, the two daughter
www.sciencedirect.com
Review TRENDS in Cell Biology Vol.15 No.5 May 2005 245

Box 2. The metaphase–anaphase transition, checkpoint and spindle defects


During spindle assembly, sister chromatids remain linked by cohesins. spindle checkpoint and a normal anaphase (7). In the presence of
The sister chromatids of each individual duplicated chromosome monotelic attachment (only one kinetochore linked to one pole) (2) or
must then be attached by microtubules to centrosomes on opposite syntelic attachment (both sister kinetochores to one pole) (1), the
sides of the spindle through their kinetochores. When this is achieved, spindle checkpoint remains on and separation of sister chromatids
the spindle checkpoint is relieved, allowing destruction of the cohesins cannot proceed. However, in the absence of Aurora-B, abnormal
and separation of the sister chromatids. The spindle checkpoint merotelic attachments (one kinetochore to both centrosomes) (4) are
inhibits the metaphase–anaphase transition until all kinetochores not detected by the checkpoint, anaphase proceeds (5) and can give
have been attached to microtubules, establishing a correct level of rise to aneuploidy (6). (b) Aurora-A is involved in centrosome
tension within the spindle. maturation and spindle assembly; in the absence of Aurora-A, many
The function of Aurora-B during bipolar spindle assembly is to spindle defects are observed: monopolar spindles with centrosomes
correct abnormal microtubule attachments to kinetochore pairs at the pole (1), multipolar spindles with centrosomes at each pole (2),
(Figure Ia). In a normal situation, amphitelic attachment (3) of sister monoastral bipolar spindles with one pole lacking a centrosome (3),
kinetochores to opposite centrosomes leads to termination of the and bipolar spindles with several centrosomes at one pole (4).

(a) (b) (1) Monopolar (2) Multipolar


Aurora-B Aurora-B

syntelic monotelic amphitelic merotelic


1 2 3 4
Metaphase

Spindle checkpoint ON Spindle checkpoint OFF

7 6
Anaphase

(3) Monoastral (4) Bipolar with several


bipolar centrosomes per pole

Figure I.

cells remain associated through long bridges of cyto- analysis performed by another group revealed that
plasm, and cytokinesis fails [51]. Aurora-B also phos- Aurora-C was highly expressed in cancer cell lines such
phorylates MgcRacGAP, a GTPase-activating protein as HepG2, HuH7, MDA-MB-453 and HeLa cells [55]. The
(GAP) required for cytokinesis [52]. Phosphorylation of same study also reported the first Aurora-C localization
Ser387 in MgcRacGAP occurs in the midbody and that was restricted to centrosomes from anaphase through
activates its GAP activity towards RhoA. MgcRacGAP to cytokinesis [55]. However, the centrosome localization
and RhoA seem to regulate cortical movement during of Aurora-C is not definitive, and others have never
cytokinesis [53]. The midbody localization of the kinesin- observed this localization. Instead, Aurora-C has been
like protein MKLP-1, whose function is required late in reported by Li et al. to be a chromosome passenger protein
cytokinesis, is also dependent on Aurora-B. Together, strictly localizing in a manner similar to, or even with,
these data clearly point to a central function for Aurora-B Aurora-B [58]. In their study, Aurora-C was actually found
during cytokinesis. to co-immunoprecipitate with Aurora-B [58]. It must be
noted, however, that the fidelity of the anti-pan-Aurora
Aurora-C: only a male meiotic kinase or another antibodies used to reveal Aurora-B and -C that are
chromosome passenger protein? thought to recognize all three Aurora kinases might not
The gene encoding Aurora-C was first isolated from a be trustworthy. However, interestingly enough, just as for
testis cDNA library and the gene localized to Chr19q13 Aurora-B, Aurora-C is activated by INCENP, and over-
[54]. Northern blot analyses of Aurora-C then confirmed expression of INCENP together with Aurora-C leads to an
that the kinase was indeed only detectable in testis increased level of phosphorylation in vivo of histone H3, a
[55,56]. During spermatogenesis, high expression of true Aurora-B substrate [58,59]. So can Aurora-C fulfil the
Aurora-C mRNA was detected mainly in pachytene function of Aurora-B? A recent study has proved that
spermatocytes, concomitant with meiotic spindle for- Aurora-C can rescue Aurora-B-depleted cells [59]. Never-
mation [57]. Although the protein level was not measured, theless, one must bear in mind that, to date, Aurora-C has
the data suggested that Aurora-C might be involved in not been found expressed in somatic cells, and its
meiotic spindle formation [56]. Strikingly, western blot expression is restricted to testis.
www.sciencedirect.com
246 Review TRENDS in Cell Biology Vol.15 No.5 May 2005

Aurora kinases control cell ploidy and centrosome


number Aurora-A Aurora-B
Surviving without Aurora kinase defect defect
Cells usually do not tolerate a lack of Aurora kinase. In the
absence of Aurora-A, centrosomes fail to assemble bipolar G2
spindles, inducing the formation of several abnormal
mitotic spindles such as spindles with short astral Prolonged mitosis: Normal mitosis
several hours Length: one hour
microtubules, monopolar spindles (one pole), monoastral
MITOSIS
bipolar spindles (bipolar, but with one pole lacking a
centrosome), bipolar spindles with multiple centrosomes
at the poles, and multipolar spindles (Box 2) [11,60]. A
decrease in Aurora-A protein level induced by RNA
interference leads to G2–M arrest, many spindle defects,
the appearance of tetraploid cells and apoptosis [61].
Tetraploid cells are formed because mitosis aborts and
cells enter the following G1 with not one but two nuclei p53–/– p53+/+ p53+/+
and two centrosomes. It is indeed well known that cells
escape mitosis when it persists too long; for example this
has been observed in the presence of spindle poisons [62].
In the following G1 phase, cell-cycle progression is G1
arrest
arrested by a so-called ‘post-mitotic checkpoint’ [63].
Whether this is a ‘tetraploid checkpoint’ is debatable
[64–66]. This G1 arrest following abortive mitosis owing to S phase S phase Cell death S phase
a spindle defect in the previous mitosis is a real curiosity.
1 2 3 4
Although the G1-activated checkpoint seems to be a
classic one, depending on p53 and the retinoblastoma
protein pRb [62], it is unknown what is detected in G1 that
Mitosis with multipolar spindles
triggers the cell-cycle arrest (Figure 4). However, the G1
checkpoint is required for the cell to trigger apoptosis.
The cells better tolerate the absence of Aurora-B than 5 6 7
Aurora-A. Inhibition of Aurora-B provokes two main events: S phase Cell death S phase
first, it eliminates the mitotic checkpoint because cells
without Aurora-B activity do not arrest in mitosis in the TRENDS in Cell Biology
presence of nocodazole and, second, it efficiently inhibits
Figure 4. Consequences of defects in Aurora kinases. In the presence of defective
cytokinesis [67]. The cells do not divide and instead become Aurora-A (top left), mitosis is prolonged because the presence of an abnormal
tetraploid, with two centrosomes. In this case, apoptosis is spindle maintains the mitotic checkpoint on. But the cell will eventually escape from
not observed (Figure 4). Elimination of Aurora-C through mitosis and become tetraploid. In cells containing inactive p53, tetraploid cells will
readily enter S phase (1). In cells containing wild-type p53, tetraploid cells arrest in
RNA interference in immortalized cells such as HeLa also the subsequent G1 phase and, depending on the severity of the defects detected by
leads to multinucleated cells, just as for Aurora-B. Further- the post-mitotic (G1) checkpoint, they will either enter S phase (2) or die (3). In the
more, double knockdown of Aurora-B and Aurora-C shows presence of defective Aurora-B (top right), the length of mitosis is not changed
because the spindle checkpoint is now deficient. However, cytokinesis does not
additive effects, indicating that both kinases might share occur and the cell becomes tetraploid. The G1 post-mitotic checkpoint is not
the same functions during mitosis [59]. activated because mitosis has not been prolonged, and the cell enters S phase (4).
There is a major difference between tetraploid cells The tetraploid cells in the above cases are not equivalent: without Aurora-A, they
will survive in p53-defective cells (5) and die in cells possessing wild-type p53 (6),
obtained after Aurora-A elimination and after Aurora-B or whereas, without Aurora-B, the cells will keep dividing (7).
Aurora-C elimination. First, without Aurora-A, the mito-
tic checkpoint detects the spindle defects, arrests the cells
in mitosis, from which they eventually escape a few hours they might be comparable to tetraploid cells that survive
later. But these tetraploid cells then face a second after limited Aurora-A depletion.
checkpoint in G1, which depends on p53. These cells can Clearly, the act of entering mitosis for a tetraploid cell is
follow two routes: they arrest in G1 and die or they enter S another challenge, it is not the amount of DNA that
phase and progress through the cell cycle. This choice matters – rather, it is the number of centrosomes. A
might depend on how badly the previous mitosis has been tetraploid G1 cell contains two centrosomes that will
affected. After serious defects and a long mitotic arrest, duplicate during the following S phase, driving a cell
tetraploid cells die, whereas, after mild defects and a short containing four centrosomes into mitosis. Usually, such a
period of arrest, tetraploid cells survive and go through cell will give rise to multipolar spindles when entering
another cycle. But again, what triggers this G1 checkpoint mitosis. This is true for tetraploid cells obtained after
remains a central question. Second, tetraploid cells inhibition of any Aurora kinase. However, again it will
generated by a lack of Aurora-B (and possibly the same then take more time for the checkpoint to be switched off
for Aurora-C) do not have to face this G1 checkpoint and and for the cell to exit mitosis in cells lacking aurora-A. By
do not die; presumably, because the mitotic checkpoint has contrast, cells lacking Aurora-B will exit from mitosis on
not been activated, mitosis is not delayed. In some ways, schedule because of the lack of checkpoint activation.
www.sciencedirect.com
Review TRENDS in Cell Biology Vol.15 No.5 May 2005 247

Again, the G1 checkpoint will take care of cells that have carcinogenesis [69]. The same authors also indicated that
had a prolonged mitosis by inducing G1 phase cell-cycle they could not obtain any colony formation when Aurora-A
arrest and cell death. was overexpressed together with a trans-dominant p53
gene that eliminates the DNA damage checkpoint or
Two much aurora causes problems too together with BCL-2 that eliminates an apoptosis path-
Now for the most intriguing part of the story. Not only does way. As immortalized NIH3T3 and rat1 cells carry many
overexpression of any of the aurora kinases lead to genetic abnormalities, it must be assumed that other
polyploid cells containing multiple centrosomes, but also defects cooperate with Aurora-A to induce oncogenicity.
the activity of the kinase itself is dispensable. In the case Identification of the responsible genes is now a priority.
of Aurora-A, both the active and the inactive kinase gave This situation has also been confirmed in vivo. A trans-
rise to multinucleated cells containing multiple centro- genic mouse designed to overexpress Aurora-A in an
somes with equal efficiency in HeLa cells [68]. However, in inducible manner in mammary glands does not develop
primary mouse embryonic fibroblasts, Aurora-A activity any tumors – only formation of tetraploid cells and
seemed essential for induction of polyploidy [69]. This apoptosis are observed in the mammary epithelium [74].
might be explained by inactivation of p53 through The mouse model can now be used to search the genetic
phosphorylation of serines 215 and 315 by Aurora-A background required to get an oncogene effect from
[22,23]. Phosphorylation of Ser215 inactivates p53 trans- overexpression of Aurora-A.
activation activity, whereas phosphorylation of Ser315 Aurora-A was also recently identified as a candidate
triggers p53 degradation. Not only does Aurora-A over- low-penetrance tumor-susceptibility gene. A T-to-A poly-
expression lead to abnormal mitosis but it provides a way morphism resulting in a change in phenylalanine 31 (F31)
to escape the p53-dependent G1 post-mitotic checkpoint. to isoleucine (I31) has been observed in the aurora-A gene
This is in agreement with the fact that only the active and reported to be associated with the degree of aneu-
Aurora-A can induce polyploidy in primary cells [69]. ploidy in human colon tumors [75]. The F31I variant is
Overexpression of inactive Aurora-A induces abnormal also preferentially amplified in esophageal squamous cell
spindles, leading to prolonged mitosis, G1 checkpoint carcinomas, and this correlates with an increase in
arrest and cell death. By contrast, overexpression of active aneuploidy [76]. However, whether the presence of this
Aurora also induces an abnormal spindle that leads to polymorphism correlates with the risk of breast cancer
prolonged mitosis, but the cells survive because active remains debatable. Indeed, a report indicates that homo-
Aurora-A inactivates p53 and promotes its degradation. zygotes for the F31I mutation have increased suscepti-
There are also differences between active Aurora-B bility to breast carcinoma [77], whereas another indicates
or -C and inactive versions of the protein. The inactive no correlation [78].
kinase is more efficient at inducing polyploidy [30,59,70]. In the case of Aurora-B, the situation is unclear. The
The fact that too much active or too much inactive kinase kinase is overexpressed in cancer cells, and an increased
can give rise to the same phenotype might also suggest level of Aurora-B correlates with advanced stages of
that the overexpressed kinase has a dominant-negative colorectal cancer [79–82]. Only one report strongly
effect or that Aurora kinases contribute to protein suggests a direct link between Aurora-B and carcino-
complexes. Increasing the level of the kinase disrupts genesis [83]. Stable clones overexpressing Aurora-B have
the stoichiometry of the protein–protein interactions, been isolated and injected in nude mice. The authors used
inhibiting the formation of active protein complexes. CHE diploid fibroblasts carrying a G245S mutation in
Furthermore, the phenotype obtained after overexpression both alleles of p53 because they could not isolate any
of Aurora-A or Aurora-B is enhanced in p53K/K cells [68]. stable clone using wild-type CHE. The mutation in p53
This again is indicative of a central role of a p53-dependent eliminates transactivation of p21, and consequently
checkpoint in the following G1 phase to establish polyploidy. abrogates the p53-dependent G1 checkpoint. More impor-
tantly, nontransfected cells induce tumor formation when
To be or not to be an oncogene – that is the question injected in nude mice, but these tumors do not induce any
The protein encoded by the Aurora-A gene was first named metastases. However, mice injected with p53(G245S) cells
BTAK for ‘breast tumor activated kinase’ because its gene, overexpressing Aurora-B form more aggressive tumors
located at 20q13, was found amplified and its mRNA that do develop metastases [83]. It was suggested that
overexpressed in breast tumors. The presence of the Aurora-B overexpression might be a secondary event in
amplicon 20q13 in breast tumors is an indicator of poor p53-defective tumor cells that leads to malignancy [83]. In
prognosis [71]. But a breakthrough came from two both Aurora-A- and Aurora-B-overexpressing cells,
different groups that demonstrated that ectopic over- defects in p53 seem to play an essential role in stabilizing
expression of Aurora-A was sufficient to transform polyploidy.
NIH3T3 and rat1 cells and that transformed cells induced
tumors when implanted in nude mice [72,73]. Since 1998, Which one of the aurora kinases is the best anti-cancer
Aurora-A has been designated as an oncogene, and therapeutic target?
approximately 30 published studies have reported over- It has been established that, first, overexpression of
expression of Aurora-A in many types of tumors. However, Aurora-A can transform cells carrying genetic defects
overexpression of Aurora-A in primary MEF cells does and, second, that Aurora-B overexpression induces tumor
not induce colony formation, clearly indicating that metastasis. In both cases, the catalytic activity of the
Aurora-A overexpression alone is insufficient to induce kinase is required. So would it be a good idea to develop
www.sciencedirect.com
248 Review TRENDS in Cell Biology Vol.15 No.5 May 2005

inhibitors of Aurora as chemotherapeutic drugs? It is that overexpression of Aurora kinases leads to polyploidy
always difficult to answer such questions when the and abnormal centrosome number? The answer is yes.
functions of the kinases are not yet fully understood. But Does overexpression of Aurora kinase underlie the origin
usually one takes a chance – and indeed a breakthrough of chromosome instability? The answer is yes too. Is this
was announced recently. A compound named VX-680 that how Aurora overexpression contributes to carcinogenesis?
inhibits all three Aurora kinases is reported to possess an The answer is clearly no. Indeed, overexpression of active
antiproliferative effect and to induce tumor regression in and inactive Aurora gives rise to chromosome instability
animal models, indicating that drugs targeting Aurora through the formation of polyploid cells containing
have an excellent potential for clinical trials [84,85]. But abnormal centrosome numbers [59,68]. However, only
there is an apparent paradox – although only Aurora-A is active kinases, at least for Aurora-A and Aurora-B,
considered an oncoprotein, it seems that it is Aurora-B respectively, transform cells or induce metastasis. This is
inhibition that induces tumor regression. Indeed, inhibi- an extremely important point because, if overexpression of
tors of Aurora kill proliferating cells through apoptosis the active kinase or inactive kinase gives rise to the same
after accumulation of tetraploid cells generated by phenotype, it is clear that the kinase activity participates
inhibition of cytokinesis and blocking mitotic histone H3 in carcinogenesis by phosphorylating proteins not
phosphorylation [84]. This undoubtedly corresponds to the involved in the appearance of the phenotype. This has
phenotype obtained only after RNA interference against recently been confirmed in a report showing that Aurora-A
Aurora-B and not against Aurora-A. This result was overexpression potentiates the oncogene activity of HRAS,
shocking because everybody had been looking for an not by interfering with the ploidy of the cell or the number
inhibitor specific for Aurora-A. However, there might be of centrosomes but by influencing cell growth [89]. Also
an explanation. Inhibition of Aurora-A leads to abnormal Aurora-A phosphorylation of p53, leading to the inhibition
spindles that can be observed only in cells having a of its transactivation activity and to its degradation, might
functional spindle checkpoint. However, in the presence of be a pivotal point. The exact role of Aurora-A in
an anti-Aurora compound that inhibits Aurora-A and carcinogenesis and Aurora-B in metastasis remains a
Aurora-B, the abnormal spindles arising from the lack of mystery; however, some indications point towards control
Aurora-A activity might not be visible because, in the of a G1 phase post-mitotic checkpoint. But even if we
absence of Aurora-B activity, they would not trigger know that a p53 defect plays a central role in allowing
mitotic arrest. Mitosis will progress with formation of an tetraploid cells to enter S phase following an abnormal
abnormal spindle until cytokinesis that itself will abort in mitosis, we still do not know what exactly is triggering this
the absence of Aurora-B activity (no cytokinesis), leading p53-dependent checkpoint. Understanding this mechan-
to a phenotype that looks like the one observed after ism seems crucial. Furthermore, future investigations
Aurora-B RNA interference. So possibly the best anti- must concentrate on the identification of the genetic
tumor drug will have to inhibit all the Aurora kinases. defects that contribute to making Aurora-A an oncogene.
Several other inhibitors have been developed against Identifying other substrates of Aurora kinases will also
Aurora kinases. Some inhibitors are currently in pre- undoubtedly help.
clinical development such as the compounds from Astra- In conclusion, let’s think about how the science might
Zeneca AZD1152 (http://www.astrazeneca.ch/corp/corp- actually progress. We might soon be able to use an
archiv/corp-archiv-read.htm?idZ17180) and compound inhibitor of Aurora kinase to treat cancers, without
677 [86], both described as inhibitors of Aurora-B. knowing exactly the function of Aurora kinases, without
Academic research laboratories are analysing Aurora knowing how Aurora kinases contribute to carcinogenesis
functions using other compounds – for example, Hesper- and without knowing why the Aurora inhibitors kill the
adin, which inhibits Aurora-B and several other kinases tumors.
[67], and ZM447439, which inhibits both Aurora-A and
Aurora-B [87]. It is likely that many pharmaceutical Acknowledgements
companies are now searching for inhibitors of the Aurora I express my apologies to all the authors who have published work related
proteins. to this review but have not been cited here owing to space constraints.
Research in the Claude Prigent laboratory is financed by the Ligue
Nationale Contre le Cancer (équipe labellisée) and by the CNRS. Clotilde
Concluding remarks Petretti is a fellow of the region Bretagne.
The similarities and differences between the three Aurora
kinases seem to fit with the proposed evolutionary origin References
of the kinases [88]. Aurora kinases evolved from a common 1 Boveri, T. (1902) Über mehrpolige Mitosen als Mittel zur Analyse des
chordate ancestor, then two Aurora kinases appeared Zellkerns. Verhandlungen der physicalisch-medizinischen Ges-
in cold-blooded vertebrates: a true Aurora-A kinase and selschaft zu Würzburg. Neu Folge 35, 67–90
2 Vernos, I. and Karsenti, E. (1995) Chromosomes take the lead in
an Aurora-B/C ancestor. In mammals, the Aurora-B/C spindle assembly. Trends Cell Biol. 5, 297–301
ancestor gave rise to two closely related kinases – Aurora-B 3 Nigg, E.A. (2001) Mitotic kinases as regulators of cell division and its
and Aurora-C. It is not surprising that these last two checkpoints. Nat. Rev. Mol. Cell Biol. 2, 21–32
kinases colocalize (to kinetochore and midbody) and share 4 Marumoto, T. et al. (2003) Aurora-A kinase maintains the fidelity of
substrates (histone H3). Overexpression of the three early and late mitotic events in HeLa cells. J. Biol. Chem. 278,
51786–51795
kinases has been observed in a large variety of tumors 5 Giet, R. and Prigent, C. (1999) Aurora/Ipl1p-related kinases, a new
containing polyploid cells with an abnormal number of oncogenic family of mitotic serine-threonine kinases. J. Cell Sci. 112,
centrosomes. So, where is the link? Is there any evidence 3591–3601
www.sciencedirect.com
Review TRENDS in Cell Biology Vol.15 No.5 May 2005 249

6 Carmena, M. and Earnshaw, W.C. (2003) The cellular geography of 33 Sampath, S.C. et al. (2004) The chromosomal passenger complex is
aurora kinases. Nat. Rev. Mol. Cell Biol. 4, 842–854 required for chromatin-induced microtubule stabilization and spindle
7 Arlot-Bonnemains, Y. et al. (2001) Identification of a functional assembly. Cell 118, 187–202
destruction box in the Xenopus laevis aurora-A kinase pEg2. FEBS 34 Speliotes, E.K. et al. (2000) The survivin-like C. elegans BIR-1 protein
Lett. 508, 149–152 acts with the Aurora-like kinase AIR-2 to affect chromosomes and the
8 Castro, A. et al. (2002) APC/Fizzy-Related targets Aurora-A kinase for spindle midzone. Mol. Cell 6, 211–223
proteolysis. EMBO Rep. 3, 457–462 35 Chen, J. et al. (2003) Survivin enhances Aurora-B kinase activity and
9 Taguchi, S. et al. (2002) Degradation of human Aurora-A protein localizes Aurora-B in human cells. J. Biol. Chem. 278, 486–490
kinase is mediated by hCdh1. FEBS Lett. 519, 59–65 36 Wheatley, S.P. et al. (2004) Aurora-B phosphorylation in vitro
10 Berdnik, D. and Knoblich, J.A. (2002) Drosophila Aurora-A is required identifies a residue of survivin that is essential for its localization
for centrosome maturation and actin-dependent asymmetric protein and binding to inner centromere protein (INCENP) in vivo. J. Biol.
localization during mitosis. Curr. Biol. 12, 640–647 Chem. 279, 5655–5660
11 Giet, R. et al. (2002) Drosophila Aurora A kinase is required to localize 37 Schumacher, J.M. et al. (1998) AIR-2: An Aurora/Ipl1-related protein
D-TACC to centrosomes and to regulate astral microtubules. J. Cell kinase associated with chromosomes and midbody microtubules is
Biol. 156, 437–451 required for polar body extrusion and cytokinesis in Caenorhabditis
12 Kemp, C.A. et al. (2004) Centrosome maturation and duplication in C. elegans embryos. J. Cell Biol. 143, 1635–1646
elegans require the coiled-coil protein SPD-2. Dev. Cell 6, 511–523 38 Terada, Y. et al. (1998) AIM-1: a mammalian midbody-associated
13 Hannak, E. et al. (2001) Aurora-A kinase is required for centrosome protein required for cytokinesis. EMBO J. 17, 667–676
maturation in Caenorhabditis elegans. J. Cell Biol. 155, 1109–1116 39 Giet, R. and Glover, D.M. (2001) Drosophila aurora B kinase is
14 Terada, Y. et al. (2003) Interaction of Aurora-A and centrosomin at the required for histone H3 phosphorylation and condensin recruitment
microtubule-nucleating site in Drosophila and mammalian cells. during chromosome condensation and to organize the central spindle
J. Cell Biol. 162, 757–763 during cytokinesis. J. Cell Biol. 152, 669–682
15 Tsai, M.Y. et al. (2003) A Ran signalling pathway mediated by the 40 Hsu, J.Y. et al. (2000) Mitotic phosphorylation of histone H3 is
mitotic kinase Aurora A in spindle assembly. Nat. Cell Biol. 5, 242–248 governed by Ipl1/aurora kinase and Glc7/PP1 phosphatase in budding
16 Eyers, P.A. et al. (2003) A novel mechanism for activation of the yeast and nematodes. Cell 102, 279–291
protein kinase Aurora A. Curr. Biol. 13, 691–697 41 Adams, R.R. et al. (2001) Essential roles of Drosophila inner
17 Katayama, H. et al. (2001) Interaction and feedback regulation centromere protein (INCENP) and aurora B in histone H3 phos-
between STK15/BTAK/Aurora-A kinase and protein phosphatase 1 phorylation, metaphase chromosome alignment, kinetochore disjunc-
through mitotic cell division cycle. J. Biol. Chem. 276, 46219–46224 tion, and chromosome segregation. J. Cell Biol. 153, 865–880
18 Kufer, T.A. et al. (2002) Human TPX2 is required for targeting Aurora- 42 Petersen, J. et al. (2001) The S. pombe aurora-related kinase Ark1
A kinase to the spindle. J. Cell Biol. 158, 617–623 associates with mitotic structures in a stage dependent manner and is
19 Hirota, T. et al. (2003) Aurora-A and an interacting activator, the LIM required for chromosome segregation. J. Cell Sci. 114, 4371–4384
protein Ajuba, are required for mitotic commitment in human cells. 43 Goto, H. et al. (2002) Aurora-B phosphorylates Histone H3 at serine28
Cell 114, 585–598 with regard to the mitotic chromosome condensation. Genes Cells 7,
20 Giet, R. et al. (1999) The Xenopus laevis aurora-related protein kinase 11–17
pEg2 associates with and phosphorylates the kinesin-related protein 44 Zeitlin, S.G. et al. (2001) CENP-A is phosphorylated by Aurora B
XlEg5. J. Biol. Chem. 274, 15005–15013 kinase and plays an unexpected role in completion of cytokinesis.
21 Dutertre, S. et al. (2004) Phosphorylation of CDC25B by Aurora-A at J. Cell Biol. 155, 1147–1157
the centrosome contributes to the G2-M transition. J. Cell Sci. 117, 45 Wei, Y. et al. (1999) Phosphorylation of histone H3 is required for
2523–2531 proper chromosome condensation and segregation. Cell 97, 99–109
22 Liu, Q. et al. (2004) Aurora-A Abrogation of p53 DNA binding and 46 Petersen, J. and Hagan, I.M. (2003) S. pombe aurora kinase/survivin
transactivation activity by phosphorylation of serine 215. J. Biol. is required for chromosome condensation and the spindle checkpoint
Chem. 279, 52175–52182 attachment response. Curr. Biol. 13, 590–597
23 Katayama, H. et al. (2004) Phosphorylation by aurora kinase A 47 Lan, W. et al. (2004) Aurora B phosphorylates centromeric MCAK and
induces Mdm2-mediated destabilization and inhibition of p53. Nat. regulates its localization and microtubule depolymerization activity.
Genet. 36, 55–62 Curr. Biol. 14, 273–286
24 Ouchi, M. et al. (2004) BRCA1 phosphorylation by Aurora-A in the 48 Ohi, R. et al. (2004) Differentiation of cytoplasmic and meiotic spindle
regulation of G2 to M transition. J. Biol. Chem. 279, 19643–19648 assembly MCAK functions by Aurora B-dependent phosphorylation.
25 Mendez, R. et al. (2000) Phosphorylation of CPE binding factor by Eg2 Mol. Biol. Cell 15, 2895–2906
regulates translation of c-mos mRNA. Nature 404, 302–307 49 Andrews, P.D. et al. (2004) Aurora B regulates MCAK at the mitotic
26 Hodgman, R. et al. (2001) CPEB phosphorylation and cytoplasmic centromere. Dev. Cell 6, 253–268
polyadenylation are catalyzed by the kinase IAK1/Eg2 in maturing 50 Trautmann, S. et al. (2004) The S. pombe Cdc14-like phosphatase
mouse oocytes. Development 128, 2815–2822 Clp1p regulates chromosome biorientation and interacts with Aurora
27 Adams, R.R. et al. (2000) INCENP binds the Aurora-related kinase kinase. Dev. Cell 7, 755–762
AIRK2 and is required to target it to chromosomes, the central spindle 51 Goto, H. et al. (2003) Aurora-B regulates the cleavage furrow-specific
and cleavage furrow. Curr. Biol. 10, 1075–1078 vimentin phosphorylation in the cytokinetic process. J. Biol. Chem.
28 Kaitna, S. et al. (2000) Incenp and an aurora-like kinase form a 278, 8526–8530
complex essential for chromosome segregation and efficient com- 52 Hirose, K. et al. (2001) MgcRacGAP is involved in cytokinesis through
pletion of cytokinesis. Curr. Biol. 10, 1172–1181 associating with mitotic spindle and midbody. J. Biol. Chem. 276,
29 Bishop, J.D. and Schumacher, J.M. (2002) Phosphorylation of the 5821–5828
carboxyl terminus of inner centromere protein (INCENP) by the 53 Minoshima, Y. et al. (2003) Phosphorylation by aurora B converts
Aurora B Kinase stimulates Aurora B kinase activity. J. Biol. Chem. MgcRacGAP to a RhoGAP during cytokinesis. Dev. Cell 4, 549–560
277, 27577–27580 54 Bernard, M. et al. (1998) Cloning of STK13, a third human protein
30 Honda, R. et al. (2003) Exploring the functional interactions between kinase related to Drosophila aurora and budding yeast Ipl1 that maps
Aurora B, INCENP, and survivin in mitosis. Mol. Biol. Cell 14, on chromosome 19q13.3-ter. Genomics 53, 406–409
3325–3341 55 Kimura, M. et al. (1998) Identification and characterization of
31 Yasui, Y. et al. (2004) Autophosphorylation of a newly identified STK12/Aik2: a human gene related to aurora of Drosophila and
site of Aurora-B is indispensable for cytokinesis. J. Biol. Chem. yeast IPL1. Cytogenet. Cell Genet. 82, 147–152
279, 12997–13003 56 Tseng, T.C. et al. (1998) Protein kinase profile of sperm and eggs:
32 Gassmann, R. et al. (2004) Borealin: a novel chromosomal passenger cloning and characterization of two novel testis-specific protein
required for stability of the bipolar mitotic spindle. J. Cell Biol. 166, kinases (AIE1, AIE2) related to yeast and fly chromosome segregation
179–191 regulators. DNA Cell Biol. 17, 823–833
www.sciencedirect.com
250 Review TRENDS in Cell Biology Vol.15 No.5 May 2005

57 Hu, H.M. et al. (2000) Genomic organization, expression, and 73 Zhou, H. et al. (1998) Tumour amplified kinase STK15/BTAK induces
chromosome localization of a third aurora-related kinase gene, Aie1. centrosome amplification, aneuploidy and transformation. Nat. Genet.
DNA Cell Biol. 19, 679–688 20, 189–193
58 Li, X. et al. (2004) Direct association with inner centromere protein 74 Zhang, D. et al. (2004) Cre-loxP-controlled periodic Aurora-A over-
(INCENP) activates the novel chromosomal passenger protein, expression induces mitotic abnormalities and hyperplasia in mam-
Aurora-C. J. Biol. Chem. 279, 47201–47211 mary glands of mouse models. Oncogene 23, 8720–8730
59 Sasai, K. et al. (2004) Aurora-C kinase is a novel chromosomal 75 Ewart-Toland, A. et al. (2003) Identification of Stk6/STK15 as a
passenger protein that can complement Aurora-B kinase function in candidate low-penetrance tumor-susceptibility gene in mouse and
mitotic cells. Cell Motil. Cytoskeleton 59, 249–263 human. Nat. Genet. 34, 403–412
60 Dutertre, S. et al. (2002) On the role of aurora-A in centrosome 76 Miao, X. et al. (2004) Functional STK15 Phe31Ile polymorphism is
function. Oncogene 21, 6175–6183 associated with the occurrence and advanced disease status of
61 Du, J. and Hannon, G.J. (2004) Suppression of p160ROCK bypasses esophageal squamous cell carcinoma. Cancer Res. 64, 2680–2683
cell cycle arrest after Aurora-A/STK15 depletion. Proc. Natl. Acad. 77 Sun, T. et al. (2004) Functional Phe31Ile polymorphism in Aurora A
Sci. U. S. A. 101, 8975–8980 and risk of breast carcinoma. Carcinogenesis 25, 2225–2230
62 Di Leonardo, A. et al. (1997) DNA rereplication in the presence of 78 Egan, K.M. et al. (2004) STK15 polymorphism and breast cancer risk
mitotic spindle inhibitors in human and mouse fibroblasts lacking in a population-based study. Carcinogenesis 25, 2149–2153
either p53 or pRb function. Cancer Res. 57, 1013–1019 79 Katayama, H. et al. (1999) Mitotic kinase expression and colorectal
63 Lanni, J.S. and Jacks, T. (1998) Characterization of the p53-dependent cancer progression. J. Natl. Cancer Inst. 91, 1160–1162
postmitotic checkpoint following spindle disruption. Mol. Cell. Biol. 18, 80 Takahashi, T. et al. (2000) Centrosomal kinases, HsAIRK1 and
1055–1064 HsAIRK3, are overexpressed in primary colorectal cancers. Jpn. J.
64 Andreassen, P.R. et al. (2001) Tetraploid state induces p53-dependent
Cancer Res. 91, 1007–1014
arrest of nontransformed mammalian cells in G1. Mol. Biol. Cell 12,
81 Adams, R.R. et al. (2001) Human INCENP colocalizes with the
1315–1328
Aurora-B/AIRK2 kinase on chromosomes and is overexpressed in
65 Borel, F. et al. (2002) Multiple centrosomes arise from tetraploidy
tumour cells. Chromosoma 110, 65–74
checkpoint failure and mitotic centrosome clusters in p53 and RB
82 Sorrentino, R. et al. (2004) Aurora B overexpression associates with
pocket protein-compromised cells. Proc. Natl. Acad. Sci. U. S. A. 99,
the thyroid carcinoma undifferentiated phenotype and is required for
9819–9824
thyroid carcinoma cell proliferation. J. Clin. Endocrinol. Metab. 90,
66 Uetake, Y. and Sluder, G. (2004) Cell cycle progression after cleavage
928–935
failure: mammalian somatic cells do not possess a “tetraploidy
83 Ota, T. et al. (2002) Increased mitotic phosphorylation of histone H3
checkpoint”. J. Cell Biol. 165, 609–615
attributable to AIM-1/Aurora-B overexpression contributes to
67 Hauf, S. et al. (2003) The small molecule Hesperadin reveals a
role for Aurora B in correcting kinetochore-microtubule attach- chromosome number instability. Cancer Res. 62, 5168–5177
ment and in maintaining the spindle assembly checkpoint. J. Cell 84 Harrington, E.A. et al. (2004) VX-680, a potent and selective small-
Biol. 161, 281–294 molecule inhibitor of the Aurora kinases, suppresses tumor growth in
68 Meraldi, P. et al. (2002) Aurora-A overexpression reveals tetraploidi- vivo. Nat. Med. 10, 262–267
zation as a major route to centrosome amplification in p53K/K cells. 85 Sausville, E.A. (2004) Aurora kinases dawn as cancer drug targets.
EMBO J. 21, 483–492 Nat. Med. 10, 234–235
69 Anand, S. et al. (2003) AURORA-A amplification overrides the mitotic 86 Nair, J.S. et al. (2004) A novel Aurora B kinase inhibitor with potent
spindle assembly checkpoint, inducing resistance to Taxol. Cancer anticancer activity either as a single agent or in combination with
Cell 3, 51–62 chemotherapy. Journal of Clinical Oncology / ASCO Annual meeting
70 Tatsuka, M. et al. (1998) Multinuclearity and increased ploidy caused 22, 9568
by overexpression of the aurora- and Ipl1-like midbody-associated 87 Ditchfield, C. et al. (2003) Aurora B couples chromosome alignment
protein mitotic kinase in human cancer cells. Cancer Res. 58, with anaphase by targeting BubR1, Mad2, and Cenp-E to kineto-
4811–4816 chores. J. Cell Biol. 161, 267–280
71 Sen, S. et al. (1997) A putative serine/threonine kinase encoding gene 88 Brown, J.R. et al. (2004) Evolutionary relationships of Aurora kinases:
BTAK on chromosome 20q13 is amplified and overexpressed in human implications for model organism studies and the development of anti-
breast cancer cell lines. Oncogene 14, 2195–2200 cancer drugs. BMC Evol. Biol. 4, 39
72 Bischoff, J.R. et al. (1998) A homologue of Drosophila aurora kinase is 89 Tatsuka, M. et al. (2005) Overexpression of Aurora-A potentiates
oncogenic and amplified in human colorectal cancers. EMBO J. 17, HRAS-mediated oncogenic transformation and is implicated in oral
3052–3065 carcinogenesis. Oncogene 24, 1122–1127

ScienceDirect collection reaches six million full-text articles


Elsevier recently announced that six million articles are now available on its premier electronic platform, ScienceDirect. This milestone
in electronic scientific, technical and medical publishing means that researchers around the globe will be able to access an unsurpassed
volume of information from the convenience of their desktop.
The rapid growth of the ScienceDirect collection is due to the integration of several prestigious publications as well as ongoing addition
to the Backfiles - heritage collections in a number of disciplines. The latest step in this ambitious project to digitize all of Elsevier’s
journals back to volume one, issue one, is the addition of the highly cited Cell Press journal collection on ScienceDirect. Also available
online for the first time are six Cell titles’ long-awaited Backfiles, containing more than 12,000 articles highlighting important historic
developments in the field of life sciences.

www.sciencedirect.com

www.sciencedirect.com

You might also like