Centrosomal amplification and spindle multipolarity in cancer cells

8
Seminars in Cancer Biology 15 (2005) 25–32 Review Centrosomal amplification and spindle multipolarity in cancer cells William Saunders Department of Biological Sciences, 258 Crawford Hall, University of Pittsburgh, Pittsburgh, PA 15260, USA Abstract Recent developments have highlighted the important role centrosomal defects play in the cellular changes associated with tumorigenesis. This article reviews recent developments addressing the impact of numerical centrosomal amplification on chromosomal segregational defects in the cancer cell. Probably, the most significant is the change to the structure of the spindle that leads to increased numbers of spindle poles and abnormal partitioning of the chromosomes in mitosis. I address how centrosomal changes are initiated and how they may lead to spindle multipolarity. © 2004 Elsevier Ltd. All rights reserved. Keywords: Centrosome; Spindle; Cytokinesis Contents 1. Introduction .......................................................................................................... 25 2. Runaway centrosomal duplication and tumor suppressors ................................................................. 27 3. Centrosome amplification and DNA damage ............................................................................. 27 4. Centrosome replication defects versus failure of cytokinesis ............................................................... 28 5. Centrosomal amplification and spindle multipolarity ...................................................................... 28 6. Virally induced centrosomal amplification ............................................................................... 29 7. Future directions ...................................................................................................... 29 References ................................................................................................................ 30 1. Introduction Chromosomal instability, defined as a continuous change in the transcriptional capacity of the cell caused by al- terations in the structure or number of chromosomes, is Tel.: +1 412 624 4320; fax: +1 412 624 4759. E-mail address: [email protected]. URL: http://www.pitt.edu/biohome/Dept/Frame/Faculty/Saunders.htm. receiving growing appreciation as central to our understand- ing of the cellular mechanisms behind tumorigenesis. The impact of chromosomal instability is a varied and ongoing change in the expression of genes controlling cell division, mobility, survival, and the many other cellular and tissue alterations needed for tumor formation. One of the major cause of chromosomal instability in cancer cells appears to be segregational errors during the mitotic division of the cell. 1044-579X/$ – see front matter © 2004 Elsevier Ltd. All rights reserved. doi:10.1016/j.semcancer.2004.09.003

Transcript of Centrosomal amplification and spindle multipolarity in cancer cells

Page 1: Centrosomal amplification and spindle multipolarity in cancer cells

Seminars in Cancer Biology 15 (2005) 25–32

Review

Centrosomal amplification and spindle multipolarity in cancer cells

William Saunders∗

Department of Biological Sciences, 258 Crawford Hall, University of Pittsburgh, Pittsburgh, PA 15260, USA

Abstract

Recent developments have highlighted the important role centrosomal defects play in the cellular changes associated with tumorigenesis.This article reviews recent developments addressing the impact of numerical centrosomal amplification on chromosomal segregational defectsin the cancer cell. Probably, the most significant is the change to the structure of the spindle that leads to increased numbers of spindle polesand abnormal partitioning of the chromosomes in mitosis. I address how centrosomal changes are initiated and how they may lead to spindlemultipolarity.© 2004 Elsevier Ltd. All rights reserved.

Keywords:Centrosome; Spindle; Cytokinesis

C

1

it

U

and-Theing

1d

ontents

1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25

2. Runaway centrosomal duplication and tumor suppressors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27

3. Centrosome amplification and DNA damage. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27

4. Centrosome replication defects versus failure of cytokinesis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28

5. Centrosomal amplification and spindle multipolarity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28

6. Virally induced centrosomal amplification. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29

7. Future directions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29

References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30

. Introduction

Chromosomal instability, defined as a continuous change

receiving growing appreciation as central to our understing of the cellular mechanisms behind tumorigenesis.impact of chromosomal instability is a varied and ongo

n the transcriptional capacity of the cell caused by al-erations in the structure or number of chromosomes, is

∗ Tel.: +1 412 624 4320; fax: +1 412 624 4759.E-mail address:[email protected].

RL: http://www.pitt.edu/∼biohome/Dept/Frame/Faculty/Saunders.htm.

change in the expression of genes controlling cell division,mobility, survival, and the many other cellular and tissuealterations needed for tumor formation. One of the majorcause of chromosomal instability in cancer cells appears tobe segregational errors during the mitotic division of thecell.

044-579X/$ – see front matter © 2004 Elsevier Ltd. All rights reserved.oi:10.1016/j.semcancer.2004.09.003

Page 2: Centrosomal amplification and spindle multipolarity in cancer cells

26 W. Saunders / Seminars in Cancer Biology 15 (2005) 25–32

Chromosomes are segregated by attachment to the highlyorganized microtubule spindle, which forms uniquely duringdivision, replacing the long monoastral array of the interphasecell. The spindle in a normal cell has mirror symmetry withthe now duplicated microtubule asters connected by cross-linked polar microtubules from each spindle pole. To thishighly symmetrical structure, the duplicated chromatids at-tach and are diametrically separated in anaphase to give equalproportions to each daughter cell – or at least that is how itis supposed to work. In cancer cells, spindles can often havemore than two poles causing the chromosomes to segregateasymmetrically, and often chaotically, leaving the daughtercells with abnormal numbers of chromosomes.

The microtubule organizing center in mammalian cells istypically associated with the centrosome. The centrosomecontains a highly organized centriolar core composed of apair of barrel-shaped orthogonally arranged cylinders of mi-crotubules. The surrounding amorphous pericentriolar regionis where microtubules are nucleated and organized into theastral array critical for directional movement in the cell. Cen-trosomes are not essential for spindle formation[1,2] but areimportant for normal chromosome segregation. Centrosomesalso play other key roles controlling cell division includinginitiation of cytokinesis and the entry into S-phase of thecell cycle [1,3,4]. Centrosomes also influence cell motility[ ort iv-a rolei ngt of thecd weenc les inc

eachd cen-t ruc-t repli-c andi p-a atesb Du-p l1,PC tro-s witht s-sr pa-rDc lin-d egge c-tD du-

plication[29]. Some these kinases may be hyperactive in tu-mor tissue as centrosomes are hyperphosphorylated in breasttumor compared to normal breast tissue[30].

During G2, a variety of additional proteins are recruitedto centrosomes and presumably play roles in maturation, in-cluding members of the Polo-like, Aurora family, and Cdk1kinases[31–33]. In mitosis, nucleophosmin is again observedat the spindle poles and associates with the centrosomal or-ganizing protein NuMA[34]. Also during mitosis, the tumorsuppressor proteins p53, TACC, and BRCA1 are found atthe centrosomes[35,36]. Centrosomes are segregated duringcell division by positioning the cell cleavage furrow near theequator of the spindle, allowing the contractile ring to sepa-rate them to the daughter cells.

Centrosomal defects were originally proposed to lead toaneuploidy and cancer in 1914 by Boveri[37,38]. He saw thatcancer cells commonly have centrosomal defects includingincreased centrosome number and postulated that changes incentrosome functionality may be key to cancer formation.Centrosome defects are indeed seen in many types of carci-nomas including breast, gall bladder, lung, bone, pancreas,colorectal, prostate, head, and neck cancers[10,30,39–45].For example, nearly 80% of invasive breast tumor cells haveamplified centrosome numbers[46]. Centrosomal changesare strongly linked to aneuploidy and chromosomal instabil-i

ells?O s ac-c stingt pro-g mald stageo riort la ans-g ) E6o ci-no tiona rly int rapido

pro-g n-t sedfi 0%i forc somald tabil-i otis morshg ors,a

5], polarity, and shape[6], which may also be important fumorigenesis[7]. In yeast, the centrosomal functional equlent, which is called the spindle pole body, plays a key

nitiating exit from mitosis[8]. Thus, scientists are beginnio appreciate the centrosome as a central componentell regulatory machinery[9–13]. This review will primarilyiscuss recent literature dissecting the relationship bethanges in centrosomes number and multipolar spindancer cells.

For chromosome segregation to proceed accurately,aughter must receive only a single centrosome, and this

rosome must only replicate once per cell cycle. Ultrastural studies have shown that centriole and centrosomalation begins near the G1/S boundary of the cell cycles completed by G2[14–16]. Replication begins by the seration of the centriolar pair. Each centriole now duplicy growth of a procentriole at a right angle to the parent.lication is controlled by members of the Cdk, Aurora/Ipolo-like, and NIMA families of cell cycle kinases[17–22].dk2 bound to cyclins A or E acts to stabilize the cenomal kinase hMps1 allowing its continued associationhe centrosome in mice[23] while phosphorylating and diociating nucleophosmin/B23[24]. Knockdown of Aurora-Aeduces�-tubulin accumulation and prevents centriolar seation during replication inCaenorhabditis elegans[25] androsophila melanogaster[26]. Work inXenopushas impli-ated a role for calcium, calmodulin and the calmoduependent protein kinase II in centrosomal replication inxtracts[27]. In budding yeast, mitotic cyclins also funion to inhibit re-replication of the spindle pole body[28]. Inrosophila, the Zyg-1 kinase is required for centrosomal

ty in a variety of studies[30,41,43,47].When do centrosomal changes first occur in tumor c

ne study showed that centrosomal defects were alwayompanied by complex chromosomal changes suggehat centrosomal changes occur relatively late in tumorression[48]. Other studies have indicated that centrosoefects were found to occur at a very early premalignantf tumor formation in a mouse mammary tumor model, p

o the appearance of detectable lesions[49]. Centrosomamplification was also seen in premalignant lesions in trenic mice expressing the human papilloma virus (HPVr E7 oncoprotein[50] and in 30–72% of preinvasive caroma in situ of cervix, breast, and prostate[46,47]and rarelybserved in normal tissue. Thus, centrosomal amplificand other centrosomal changes appear to occur very ea

umorigenesis, but are apparently associated with thenset of complex chromosomal changes.

Centrosomal defects increase in severity during tumorression[10,41,47,48,51]. For example, the frequency of ce

rosomal amplification in cervical carcinoma cells increarom nearly zero in normal epithelium, to∼20% of the cellsn grade 1 tumors,∼50% in grade 2 tumors, and nearly 7n grade 3 tumors[47]. Similar results were observedhanges in the shape and size of centrosomes. Centroefects are also correlated with increased genetic ins

ty and aneuploidy[41,43,48,52], although apparently nn HPV virally induced centrosomal amplification[53]. In atudy of breast tumors, genetically unstable aneuploid tuad∼6.8 centrosomes per cell, compared to∼3.6 per cell inenetically stable aneuploid tumors, 2.8 in diploid tumnd 1.5 in normal tissues[46].

Page 3: Centrosomal amplification and spindle multipolarity in cancer cells

W. Saunders / Seminars in Cancer Biology 15 (2005) 25–32 27

2. Runaway centrosomal duplication and tumorsuppressors

Under some conditions, centrosome replication canbecome uncoupled from the cell cycle allowing multiplecentrosomes to form in a single cell. In tumor cell lines,over-replication is correlated with reduced activity of thep53 tumor suppressor pathway, commonly by mutationalinactivation of theTP53gene[42,54–56]. For example, p53mutant mice had a marked increase in sensitivity to papillomaformation following exposure to chemical carcinogens, with75% of the tumor cells containing three or more centrosomes[56]. A reduction in p53 activity in tumors can also beachieved by overexpression of the p53 inactivating proteinMdm2 [42] or reduced activity of the downstream targetsof p53, p21Cip1/waf1 [55,57] and Gadd45[58,59] and all ofthese are correlated with centrosomal amplification.

Why is loss of p53 important? As I discuss further below,centrosomal amplification is often correlated with an increasein ploidy from cytokinesis defects. It has been suggested thatloss of p53 activity allows cells with polyploidy to proliferateinstead of apoptose[60–63]. Treatment of cells with actininhibitors like cytochalasin induces polyploidy by blockingcytokinesis[64]. When primary rat embryo fibroblasts cellswere treated with cytochalasin, they arrested indefinitely with4 thes sint NArH avear enceo s toc ulatec crip-t mesi smi ectso

fica-t ationa withp

ors lossoo toc no cen-t theu bi s inH orE y actt ssiono rip-

tion regulators[68]. Taken together, these results suggestthat loss of Rb alone is insufficient to induce centrosomalamplification, but that loss of Rb may play an important rolein allowing centrosomal amplification by other mechanisms.Thus, it is likely loss of Rb and/or p53 may promote centro-somal amplification by setting up a permissive condition forcells with amplified centrosomes to proliferate.

3. Centrosome amplification and DNA damage

There are many observations linking DNA damage tochanges in centrosome number. For example, incompletelyreplicated or damaged DNA leads to centrosomal splitting inDrosophilaand mammalian cells[69] and�-irradiation canlead to centrosome amplification in tumor cells[70]. Also,overexpression of the DNA damage sensors ATM and ATRcan lead to centrosomal amplification[71], as can mutationsin BRCA1 or BRCA2[72,73], Xrcc2 and Xrcc3[72], andin the presence of a dominant negative mutant of the Rad51DNA repair protein[74]. However, some of these proteinshave pleiotropic effects on the cell cycle, apoptosis, chro-matin and other cellular processes that may indirectly influ-ence centrosome number.

A particularly well-described example of centrosome de-fE n ofD g-i mi-tr ncet ucest rasei tro-s ore,i -i losso n ina witha cen-t rticalm ucleif Ad nasea losso tos Chk1o spin-d agentc cen-t andm ytoki-n olars , butp nd int

n DNA. When the same cells were transformed withimian virus-40 large T antigen to inhibit p53, cytochalareatment did not arrest the cells but allowed further Deplication to 8n cells, followed by severe aneuploidy[60].uman colorectal cancer cell lines with p53 mutations hn increased tendency towards polyploidy[65], which mayesult from increased survival of these cells in the absf p53-mediated genomic surveillance. p53 localizeentrosomes before and after duplication and can regentrosome replication independent of its role as transion regulator[66]. p53 disassociates from the centroson cells with mitotic defects and is stabilized in the cytoplanducing G1 arrest in the next cell cycle. When mitotic defccurred in cells without p53, no arrest occurred[35,36].

Is loss of p53 an essential part of centrosomal ampliion? The answer appears to be no. Centrosomal amplificnd other centrosomal changes can be seen in tumorsersistent p53 activity[46,47,51].

There is still uncertainty about the role of the Rb tumuppressor protein in centrosomal amplification. Thef Rb itself, or reduced expression of p16ink4a or inductionf cdk4, both of which block Rb function, did not leadentrosomal alterations[42,54]. However, while expressiof a mutant HPV E7, unable to bind Rb, decreased

rosomal amplification by E7 by three-fold, similar tontransfected control[67], inhibitory phosphorylation of R

s required to allow over-replication of the centrosomeU treated cells[68]. Overexpression of either cyclin A2F1 can compensate for loss of Rb, suggesting Rb ma

o regulate centrosome number by transcriptional repref the cyclin A gene, a target of the E2F family of transc

ects and DNA damage has been demonstrated inDrosophila.mbryos induced to undergo mitosis prior to completioNA replication, or following treatment with DNA dama

ng X-rays and UV light show centrosomal defects inosis, including loss of the microtubule nucleating�-tubulining complex[75,76]. Takaeda et al. have gathered evidehat DNA double strand breaks are the signal that indhe centrosomal changes. DNA break-forming topoisomenhibitors or the drug bleomycin produced similar cenome changes in 100% of injected embryos. Furthermnjection of restriction-digested�-X174 DNA induced simlar changes as DNA damaging agents, including thef �-tubulin staining from the centrosomes, a reductiostral microtubules and disorganization of the spindle,ccompanying chromosomal segregational defects. The

rosomal changes led to loss of the nucleus from the coonolayer of the embryo and prevented the damaged n

rom being included in the developing embryo. Like DNamaging agents, mutational inactivation of the Chk1 kilso led to spontaneous centrosomal inactivation withf �-tubulin. The Chk2 DNA damage kinase is requiredignal the centrosomal changes associated with loss ofr DNA damage. Chk2 localizes to the centrosome andle, a response that is enhanced by the DNA damagingamptothecin or loss of Chk1. In the absence of Chk2rosomal inactivation, the damaged cells were not lostitosis proceeded, but chromosome segregation and cesis failed, leading to multiple centrosomes and multippindles in the next mitosis. Chk2 acts upstream of p5353 was not required for the centrosomal changes fou

he Chk2 mutant.

Page 4: Centrosomal amplification and spindle multipolarity in cancer cells

28 W. Saunders / Seminars in Cancer Biology 15 (2005) 25–32

Fig. 1. A pathway for multipolarity is shown. A cell with normal centrosome numbers can amplify the centrosomes by over-replication or failure of cytokinesis.Alternatively, centrosomes may fragment into multiple microtubule organizing centers. The multiple centrosomes may initially be clustered in a bipolar spindleand require further steps to overcome this clustering mechanism. Alternatively, the amplified centrosomes may form multipolar spindles without further changeand cluster only in unusual circumstances (not shown).

Spindle multipolarity is strongly correlated with anaphasebridges, which frequently result in DNA breaks, in tumorsof the head and neck (p< 0.05) and bone and soft tissue(p< 0.0006)[48,77]. The authors suggest this linkage couldbe due to bridges blocking cytokinesis and leading to cen-trosome amplification. The previously cited work from fliessuggest that multipolarity may also be triggered in some casesby DNA damage in cells with anaphase bridges via a Chk2-related pathway.

Cells from BRCA1 or BRCA2 deficient mice have cen-trosomal amplification[72,73]and BRCA1 binds to centro-somes in a�-tubulin-dependent manner. Expression of theBRCA1 �-tubulin binding domain alone reduces BRCA1 atthe centrosome and results in multipolar spindles. BRCA1 isphosphorylated in S-phase and in cells with DNA damage,and a reduction in BRCA1 phosphorylation results in reducedbinding of BRCA1 to�-tubulin and centrosomes and an in-duction of multipolar spindles.

4. Centrosome replication defects versus failure ofcytokinesis

Centrosome amplification could proceed through eitherover-replication of the centrosomes or by a cell division orcytokinesis defect leading to amplified centrosome numbers( o beda de-f ationi e ofc -A isfia am-p NAr isa ctingt of

the cells with centrosomal amplification were multinucleatedand 20% were delayed in late mitosis or cytokinesis consis-tent with a cytokinesis defect. Surprisingly, centrosomal am-plification was still observed in mutants with sharply reducedor absent kinase activity. Similar observations were madewith overexpression of Plk1 or Aurora-B kinases. p53−/−mutants showed a 66% correlation between cells with multi-polar spindles and multinucleation. These observations sug-gest that multipolarity in these mutants is a result of a failureof cytokinesis rather than a centrosomal replication defect.Further study is needed to define fully the relative role of thesetwo mechanisms in centrosomal amplification in tumor cells.

5. Centrosomal amplification and spindlemultipolarity

The main impact of centrosome amplification on cancercells is most likely the formation of multipolar spindles. Howtightly linked are centrosomal amplification and spindlemultipolarity? Both the defects are generally observedtogether in tumor tissue[47,48] however, the frequencyof mitotic cells in tissue is typically quite low, and strongcorrelations are difficult to make. We know that spindlemultipolarity can be induced in the absence of centrosomalamplification. For example, overexpression of the centroso-m andm theto mesa (N.Q ) anda mals

mala hea ncesa TheN dis-

Fig. 1). Centrosomal amplification is often suggested tue to centrosomal replication defects[42,54,57], yet ex-mples of over-replication in the absence of cytokinesis

ects are rare, therefore at least some of the amplificnterpreted as replication defects may be due to failurytokinesis. The centrosomal-associated kinase Aurorarequently amplified in tumors and cancer cell lines[78] ands associated with centrosomal amplification[79]. Meraldi etl. recently showed that Aurora-A induced centrosomallification is blocked in cells arrested in S-phase by the Deplication inhibitor hydroxyurea, suggesting Aurora-Acting at another point of the cell cycle and may not be a

o over-replicate centrosomes[80]. Seventy-five percent

al Nek2 kinase elicits a splitting of the centrosomesultipolarity [21] as does treatment of neutrophils with

umor promoter 12-0-tetradecanoylphorbol-13-acetate[81]r DNA damaging conditions. However, most centrosoppear to be unfragmented in cancer cells in cultureuintyne and W. Saunders, unpublished observationsfurther consideration of the relevance of centroso

plitting in tumor cells is needed.Are additional changes in the cell other than centroso

mplification required to induce spindle multipolarity? Tnswer remains unclear. It is known that in some instamplified centrosomes fail to form multipolar spindles.E-115 neuroblastoma cell line has large numbers of

Page 5: Centrosomal amplification and spindle multipolarity in cancer cells

W. Saunders / Seminars in Cancer Biology 15 (2005) 25–32 29

persed centrosomes in interphase, but at mitosis the cen-trosomes coalesce allowing a bipolar spindle to form[15].Induction of extra centrosomes with the actin inhibitor cy-tochalasin B in BSC-1 cells still lead to bipolar division in30–50% of the cells, showing that some mechanism acts toprevent full centrosome separation following amplification[83]. Bill Brinkley has recently proposed that this may repre-sent a centrosomal coalescence mechanism that may preventmultipolarity in some cells[82]. However, the nature of thisputative coalescence activity remains unknown. Specifically,it is unclear if the mechanism of clustering of complete cen-trosomes such as observed in the NE-115 cells is related tothe cohesion of the two centrioles found in the normal cen-triolar pair. Another possibility is that clustering may merelyrepresent the proximity of replicated centrosomes[83].

Ch-TOG belongs to the XMAP215 family of microtubulebinding proteins known to play a key role in microtubule sta-bility in Xenopusegg extracts[84]. Binding of ch-TOG to thecentrosome is mediated by a set of three TACC proteins[85].Changes in both TACC and ch-TOG human homologs havebeen implicated in cancer[36]. siRNA knockdown of humanTOG caused showed little or no change in microtubule stabil-ity but a dramatic rise in multipolar spindles to approximately40% of the metaphase population. Thus, in human cells thispathway may play an important role in multipolarity in tumorc

6

ssiono ingp orE l am-p tes[ en-t

fica-t tin-d andt mi ba auses b.H stilli thef notrT mpli-fi Rb,b lved[

mec t theym st ap-

pearance of structurally immature centriolar cores[67]. E6,on the other hand, takes weeks to amplify the centrosomesand typically results in multinucleated cells. These results canbe interpreted to indicate that E7 acts primarily to interferewith centrosomal duplication while E6 may interfere with celldivision[92]. However, E7 expression also increases the fre-quency of multinucleated cells, and cell cycle arrest preventsthe centrosomal amplification phenotype from cells express-ing E7 [67]. Thus, E7 apparently also acts at least in partthrough misregulation of cell division.

Recently, centrosomal amplification and spindle multipo-larity was also observed in cells expressing the Hepatitis Bvirus X (HBx) oncoprotein[93,94]. Forgues et al. found thatHBx bound to and sequestered the nuclear transport proteinCrm1 in the cytoplasm and that HBx mutants lacking nuclearlocalization failed to induce multipolarity. Furthermore, in-hibition of nuclear transport by leptomycin B also inducedspindle multipolarity while overexpression of Crm1 blockedHBx induction of multipolarity. Thus, HBx may act to blockCrm1 nuclear export function. HBx inactivates p53 and tran-scription of p21[95], and some Crm1 could be seen at thecentrosome and was frequently lost in cells treated with lep-tomycin B. Yun et al. suggest that cytoplasmic sequestrationof p53 may be the mechanism by which HBx induces cen-trosomal amplification[93]. Alternatively, centrioles in lep-t atedw ug-g mesi gt torsn g tom

7

cen-t lastf . Asw im-p fullyd s in-c somalf e ofc vesti-g ies ofs lls.O ulti-p fixedc encem like-l asa tipo-l ser-v ancyi les.

ells.

. Virally induced centrosomal amplification

It has been appreciated for many years that expref certain viral protein can induce mitotic errors includolyploidy [86]. Expression of variants of either HPV E67 in transgenic mice cooperatively lead to centrosomalification and spindle multipolarity in normal keratinocy

53,67,87]. Adenovirus E1A-expressing cells also show crosomal amplification and multipolar spindles[88].

What is the mechanism behind centrosomal ampliion from viral proteins? The E6 protein activates ubiquiependent degradation of p53 while the E7 protein binds

argets Rb for destruction[89]. E7 also prevents p21 fronhibiting Cdk2/cyclin E, thus interfering with both the Rnd p53 pathways. Thus, these viral oncoproteins may cpindle multipolarity by inhibiting the activity of p53 and Rowever, E7 variants that lack the pRb binding domain

nduce centrosomal amplification, albeit at only one thirdrequency[67]. Also, overexpression of Cdk4 alone caneplicate the centrosomal defects of E7 expressing cells[90].hese data suggests that E7 induction of centrosomal acation does not proceed simply by inactivating p53 andut that other unknown partners of E6/E7 are also invo88].

While both E7 and E6 can induce multipolarity, the tiourse of induction and the exact phenotypes suggesay act through different pathways[91]. E7 expression lead

o centrosome amplification in less than 48 h, with the

omycin B-treated cells were irregular, abnormally elongith extra “minicentrioles” separated from their parents, sesting a defect in centriolar replication. Some centroso

n HBx-expressing cells lacked�-tubulin staining suggestinhat HBx may interfere with Crm1-mediated export of facecessary to maintain centrosomal integrity thus leadinultipolarity [94].

. Future directions

Though Boveri suggested almost 100 years ago thatrosomal changes play a key role in cancer, only in theew years have we come to realize how right he wase continue to investigate this further, I suggest twoortant research issues to keep in mind. One is to careistinguish between centrosomal amplification, defined areased numbers of normal centrosomes, and centroragmentation or splitting. Furthermore, the significancentrosomal fragmentation in cancer cells needs to be inated. The second is to at least consider actual frequencpindle multipolarity and other mitotic defects in live ceur observations indicate that the real frequency of molarity may be substantially less than that observed inells, at least in some cancer cell lines. Real-time fluorescicroscopy of histone-GFP labeled cells showed that the

ihood that a cell will divide with a multipolar spindle wbout one half the frequency of metaphase cells with mul

ar spindles in fixed samples (Luo et al., unpublished obations). Preliminary analysis suggests that the discreps due to a delay in mitosis of cells with multipolar spind

Page 6: Centrosomal amplification and spindle multipolarity in cancer cells

30 W. Saunders / Seminars in Cancer Biology 15 (2005) 25–32

While the equipment for live cell microscopy may not be al-ways available, investigators need to keep in mind that the ac-tual frequency of mitotic defects like multipolar spindles maybe substantially different than that observed in fixed cells.

References

[1] Khodjakov A, Cole RW, Oakley BR, Rieder CL. Centrosome-independent mitotic spindle formation in vertebrates. Curr Biol2000;10(2):59–67.

[2] Heald R, Tournebize R, Blank T, Sandaltzopoulos R, Becker P, Hy-man A, et al. Self-organization of microtubules into bipolar spin-dles around artificial chromosomes inXenopusegg extracts. Nature1996;382:420–5.

[3] Hinchcliffe EH, Miller FJ, Cham M, Khodjakov A, Sluder G. Re-quirement of a centrosomal activity for cell cycle progression throughG1 into S phase. Science 2001;291(5508):1547–50.

[4] Piel M, Nordberg J, Euteneuer U, Bornens M. Centrosome-dependentexit of cytokinesis in animal cells. Science 2001;291(5508):1550–3.

[5] Bornens M. Centrosome composition and microtubule anchoringmechanisms. Curr Opin Cell Biol 2002;14(1):25–34.

[6] Niu MY, Mills JC, Nachmias VT. Development of polarity in humanerythroleukemia cells: roles of membrane ruffling and the centro-some. Cell Motil Cytoskeleton 1997;36(3):203–15.

[7] Yvon AM, Walker JW, Danowski B, Fagerstrom C, Khodjakov A,Wadsworth P. Centrosome reorientation in wound-edge cells is celltype specific. Mol Biol Cell 2002;13(6):1871–80.

[8] de Bettignies G, Johnston LH. The mitotic exit network. Curr Biol

tressBiol

[ Cell

[ rte-l Biol

[ Cell

[ ingycle.

[ ized

[ le

[ RL.icro-–62.

[ re-ction

[ naseSA

[ bal-14(Pt

[ one-am-ene

[ ancle

[22] Mayor T, Meraldi P, Stierhof YD, Nigg EA, Fry AM. Protein kinasesin control of the centrosome cycle. FEBS Lett 1999;452(1–2):92–5.

[23] Fisk HA, Winey M. The mouse Mps1p-like kinase regulates centro-some duplication. Cell 2001;106(1):95–104.

[24] Okuda M, Horn HF, Tarapore P, Tokuyama Y, Smulian AG, ChanPK, et al. Nucleophosmin/B23 is a target of CDK2/cyclin E in cen-trosome duplication. Cell 2000;103(1):127–40.

[25] Hannak E, Kirkham M, Hyman AA, Oegema K. Aurora-A kinaseis required for centrosome maturation inCaenorhabditis elegans. JCell Biol 2001;155(7):1109–16.

[26] Berdnik D, Knoblich JA.DrosophilaAurora-A is required for cen-trosome maturation and actin-dependent asymmetric protein local-ization during mitosis. Curr Biol 2002;12(8):640–7.

[27] Matsumoto Y, Maller JL. Calcium, calmodulin, and CaMKII re-quirement for initiation of centrosome duplication inXenopuseggextracts. Science 2002;295(5554):499–502.

[28] Haase SB, Winey M, Reed SI. Multi-step control of spindlepole body duplication by cyclin-dependent kinase. Nat Cell Biol2001;3(1):38–42.

[29] O’Connell KF, Caron C, Kopish KR, Hurd DD, Kemphues KJ, Li Y,et al. TheC. eleganszyg-1 gene encodes a regulator of centrosomeduplication with distinct maternal and paternal roles in the embryo.Cell 2001;105(4):547–58.

[30] Lingle WL, Lutz WL, Ingle JN, Maihle NJ, Salisbury JL. Cen-trosome hypertrophy in human breast tumors: implications forgenomic stability and cell polarity. Proc Natl Acad Sci USA1998;95(6):2950–5.

[31] Bailly E, Doree M, Nurse P, Bornens M. p34cdc2 is located in bothnucleus and cytoplasm; part is centrosomally associated at G2/Mand enters vesicles at anaphase. Embo J 1989;8(13):3985–95.

[ rst43–8.

[ ianof

clearSA

[ Bor-rt to–66.

[ res-

[ rends

[ can-ed

[ can-ton

[ oci-athol

[ M,uctal

[ eyneticer Res

[ eichmo-res-

[ nncurscell

2003;13(8):R301.[9] Lange BM. Integration of the centrosome in cell cycle control, s

response and signal transduction pathways. Curr Opin Cell2002;14(1):35–43.

10] Doxsey S. Re-evaluating centrosome function. Nat Rev MolBiol 2001;2(9):688–98.

11] Rieder CL, Faruki S, Khodjakov A. The centrosome in vebrates: more than a microtubule-organizing center. Trends Cel2001;11(10):413–9.

12] Stearns T. Centrosome duplication. A centriolar pas de deux.2001;105(4):417–20.

13] Hinchcliffe EH, Sluder G. “It takes two to tango”: understandhow centrosome duplication is regulated throughout the cell cGenes Dev 2001;15(10):1167–81.

14] Robbins E, Jentzsch G, Micali A. The centriole cycle in synchronHeLa cells. J Cell Biol 1968;36(2):329–39.

15] Ring D, Hubble R, Kirschner M. Mitosis in a cell with multipcentrioles. J Cell Biol 1982;94(3):549–56.

16] Brinkley BR, Cox SM, Pepper DA, Wible L, Brenner SL, PardueTubulin assembly sites and the organization of cytoplasmic mtubules in cultured mammalian cells. J Cell Biol 1981;90(3):554

17] Hinchcliffe EH, Li C, Thompson EA, Maller JL, Sluder G. Requiment of Cdk2-cyclin E activity for repeated centrosome reproduin Xenopusegg extracts. Science 1999;283(5403):851–4.

18] Lacey KR, Jackson PK, Stearns T. Cyclin-dependent kicontrol of centrosome duplication. Proc Natl Acad Sci U1999;96(6):2817–22.

19] Meraldi P, Nigg EA. Centrosome cohesion is regulated by aance of kinase and phosphatase activities. J Cell Sci 2001;120):3749–57.

20] Mussman JG, Horn HF, Carroll PE, Okuda M, Tarapore P, Dhower LA, et al. Synergistic induction of centrosome hyperplification by loss of p53 and cyclin E overexpression. Oncog2000;19(13):1635–46.

21] Fry AM, Meraldi P, Nigg EA. A centrosomal function for the humNek2 protein kinase, a member of the NIMA family of cell cyregulators. Embo J 1998;17(2):470–81.

32] Jackman M, Lindon C, Nigg EA, Pines J. Active cyclin B1-Cdk1 fiappears on centrosomes in prophase. Nat Cell Biol 2003;5(2):1

33] Pockwinse SM, Krockmalnic G, Doxsey SJ, Nickerson J, LJB, van Wijnen AJ, et al. Cell cycle independent interactionCDC2 with the centrosome, which is associated with the numatrix-intermediate filament scaffold. Proc Natl Acad Sci U1997;94(7):3022–7.

34] Zatsepina OV, Rousselet A, Chan PK, Olson MO, Jordon EG,nens M. The nucleolar phosphoprotein B23 redistributes in pathe spindle poles during mitosis. J Cell Sci 1999;112(Pt 4):455

35] Fisk HA, Mattison CP, Winey M. Centrosomes and tumour suppsors. Curr Opin Cell Biol 2002;14(6):700–5.

36] Raff JW. Centrosomes and cancer: lessons from a TACC. TCell Biol 2002;12(5):222–5.

37] Wunderlich V. JMM—past and present chromosomes andcer: Theodor Boveri’s predictions 100 years later. J Mol M2002;80(9):545–8.

38] Brinkley BR, Goepfert TM. Supernumerary centrosomes andcer: Boveri’s hypothesis resurrected. Cell Motil Cytoskele1998;41(4):281–8.

39] Lingle WL, Salisbury JL. Altered centrosome structure is assated with abnormal mitoses in human breast tumors. Am J P1999;155(6):1941–51.

40] Sato N, Mizumoto K, Nakamura M, Nakamura K, KusumotoNiiyama H, et al. Centrosome abnormalities in pancreatic dcarcinoma. Clin Cancer Res 1999;5(5):963–70.

41] Pihan GA, Purohit A, Wallace J, Malhotra R, Liotta L, DoxsSJ. Centrosome defects can account for cellular and gechanges that characterize prostate cancer progression. Canc2001;61(5):2212–9.

42] Carroll PE, Okuda M, Horn HF, Biddinger P, Stambrook PJ, GlLL, et al. Centrosome hyperamplification in human cancer: chrosome instability induced by p53 mutation and/or Mdm2 overexpsion. Oncogene 1999;18(11):1935–44.

43] Ghadimi BM, Sackett DL, Difilippantonio MJ, Schrock E, NeumaT, Jauho A, et al. Centrosome amplification and instability ocexclusively in aneuploid, but not in diploid colorectal cancer

Page 7: Centrosomal amplification and spindle multipolarity in cancer cells

W. Saunders / Seminars in Cancer Biology 15 (2005) 25–32 31

lines, and correlates with numerical chromosomal aberrations. GenesChromosomes Cancer 2000;27(2):183–90.

[44] Kuo KK, Sato N, Mizumoto K, Maehara N, Yonemasu H, KerCG, et al. Centrosome abnormalities in human carcinomas of thegallbladder and intrahepatic and extrahepatic bile ducts. Hepatology2000;31(1):59–64.

[45] Haruki N, Harano T, Masuda A, Kiyono T, Takahashi T, TatematsuY, et al. Persistent increase in chromosome instability in lung can-cer: possible indirect involvement of p53 inactivation. Am J Pathol2001;159(4):1345–52.

[46] Lingle WL, Barrett SL, Negron VC, D’Assoro AB, Boeneman K,Liu W, et al. Centrosome amplification drives chromosomal in-stability in breast tumor development. Proc Natl Acad Sci USA2002;99(4):1978–83.

[47] Pihan GA, Wallace J, Zhou Y, Doxsey SJ. Centrosome abnormalitiesand chromosome instability occur together in pre-invasive carcino-mas. Cancer Res 2003;63(6):1398–404.

[48] Gisselsson D, Palsson E, Yu C, Mertens F, Mandahl N. Mitoticinstability associated with late genomic changes in bone and softtissue tumours. Cancer Lett 2004;206(1):69–76.

[49] Goepfert TM, Adigun YE, Zhong L, Gay J, Medina D, Brink-ley WR. Centrosome amplification and overexpression of AuroraA are early events in rat mammary carcinogenesis. Cancer Res2002;62(14):4115–22.

[50] Riley RR, Duensing S, Brake T, Munger K, Lambert PF, ArbeitJM. Dissection of human papillomavirus E6 and E7 function intransgenic mouse models of cervical carcinogenesis. Cancer Res2003;63(16):4862–71.

[51] D’Assoro AB, Barrett SL, Folk C, Negron VC, Boeneman K, BusbyR, et al. Amplified centrosomes in breast cancer: a potential indicator

25–34.[ gai

d withc car-vest

[ bertchro-ncer

[ GF.ience

[ n ofway.

[ D,enic

;17(1):

[ G,rchi-cro-lood

[ ri-45a-

[ etatl

[ loidalian

[ onsencer

[62] Casenghi M, Mangiacasale R, Tuynder M, Caillet-Fauquet P, Elha-jouji A, Lavia P, et al. p53-independent apoptosis and p53-dependentblock of DNA rereplication following mitotic spindle inhibition inhuman cells. Exp Cell Res 1999;250(2):339–50.

[63] Minn AJ, Boise LH, Thompson CB. Expression of Bcl-xL andloss of p53 can cooperate to overcome a cell cycle checkpoint in-duced by mitotic spindle damage. Genes Dev 1996;10(20):2621–31.

[64] Aubin JE, Osborn M, Weber K. Inhibition of cytokinesis and alteredcontractile ring morphology induced by cytochalasins in synchro-nized PtK2 cells. Exp Cell Res 1981;136(1):63–79.

[65] Bunz F, Fauth C, Speicher MR, Dutriaux A, Sedivy JM, KinzlerKW, et al. Targeted inactivation of p53 in human cells does notresult in aneuploidy. Cancer Res 2002;62(4):1129–33.

[66] Tarapore P, Tokuyama Y, Horn HF, Fukasawa K. Difference in thecentrosome duplication regulatory activity among p53 ‘hot spot’ mu-tants: potential role of Ser 315 phosphorylation-dependent centro-some binding of p53. Oncogene 2001;20(47):6851–63.

[67] Duensing S, Lee LY, Duensing A, Basile J, Piboonniyom S, GonzalezS, et al. The human papillomavirus type 16 E6 and E7 oncoproteinscooperate to induce mitotic defects and genomic instability by un-coupling centrosome duplication from the cell division cycle. ProcNatl Acad Sci USA 2000;97(18):10002–7.

[68] Meraldi P, Lukas J, Fry AM, Bartek J, Nigg EA. Centrosome du-plication in mammalian somatic cells requires E2F and Cdk2-cyclinA. Nat Cell Biol 1999;1(2):88–93.

[69] Hut HM, Lemstra W, Blaauw EH, Van Cappellen GW, KampingaHH, Sibon OC. Centrosomes split in the presence of impaired DNAintegrity during mitosis. Mol Biol Cell 2003;14(5):1993–2004.

[70] Sato N, Mizumoto K, Nakamura M, Ueno H, Minamishima YA,n in

[ tleyidy9(1):

[ k J,someCurr

[ en-oint

ells.

[ ssionile aloidygene

[ A-in

[ eomic

[ J, etomalres.

[ ki-lifiedogene

[ al.pli-enet

of tumor aggressiveness. Breast Cancer Res Treat 2002;75(1):52] Shono M, Sato N, Mizumoto K, Maehara N, Nakamura M, Na

E, et al. Stepwise progression of centrosome defects associatelocal tumor growth and metastatic process of human pancreaticinoma cells transplanted orthotopically into nude mice. Lab In2001;81(7):945–52.

53] Schaeffer AJ, Nguyen M, Liem A, Lee D, Montagna C, LamPF, et al. E6 and E7 oncoproteins induce distinct patterns ofmosomal aneuploidy in skin tumors from transgenic mice. CaRes 2004;64(2):538–46.

54] Fukasawa K, Choi T, Kuriyama R, Rulong S, Vande WoudeAbnormal centrosome amplification in the absence of p53. Sc1996;271(5256):1744–7.

55] Tarapore P, Horn HF, Tokuyama Y, Fukasawa K. Direct regulatiothe centrosome duplication cycle by the p53-p21Waf1/Cip1 pathOncogene 2001;20(25):3173–84.

56] Wang XJ, Greenhalgh DA, Jiang A, He D, Zhong L, Medinaet al. Expression of a p53 mutant in the epidermis of transgmice accelerates chemical carcinogenesis. Oncogene 199835–45.

57] Mantel C, Braun SE, Reid S, Henegariu O, Liu L, Hangocet al. p21(cip-1/waf-1) deficiency causes deformed nuclear atecture, centriole overduplication, polyploidy, and relaxed mitubule damage checkpoints in human hematopoietic cells. B1999;93(4):1390–8.

58] Hollander MC, Sheikh MS, Bulavin DV, Lundgren K, AngeHenmueller L, Shehee R, et al. Genomic instability in Gadddeficient mice. Nat Genet 1999;23(2):176–84.

59] Wang XW, Zhan Q, Coursen JD, Khan MA, Kontny HU, Yu L,al. GADD45 induction of a G2/M cell cycle checkpoint. Proc NAcad Sci USA 1999;96(7):3706–11.

60] Andreassen PR, Lohez OD, Lacroix FB, Margolis RL. Tetrapstate induces p53-dependent arrest of nontransformed mammcells in G1. Mol Biol Cell 2001;12(5):1315–28.

61] Khan SH, Wahl GM. p53 and pRb prevent rereplication in respto microtubule inhibitors by mediating a reversible G1 arrest. CaRes 1998;58(3):396–401.

Farber JL, et al. A possible role for centrosome overduplicatioradiation-induced cell death. Oncogene 2000;19(46):5281–90.

71] Smith L, Liu SJ, Goodrich L, Jacobson D, Degnin C, BenN, et al. Duplication of ATR inhibits MyoD, induces aneuploand eliminates radiation-induced G1 arrest. Nat Genet 1998;139–46.

72] Tutt A, Gabriel A, Bertwistle D, Connor F, Paterson H, Peacocet al. Absence of Brca2 causes genome instability by chromobreakage and loss associated with centrosome amplification.Biol 1999;9(19):1107–10.

73] Xu X, Weaver Z, Linke SP, Li C, Gotay J, Wang XW, et al. Ctrosome amplification and a defective G2-M cell cycle checkpinduce genetic instability in BRCA1 exon 11 isoform-deficient cMol Cell 1999;3(3):389–95.

74] Bertrand P, Lambert S, Joubert C, Lopez BS. Overexpreof mammalian Rad51 does not stimulate tumorigenesis whdominant-negative Rad51 affects centrosome fragmentation, pand stimulates tumorigenesis, in p53-defective CHO cells. Onco2003;22(48):7587–92.

75] Sibon OC, Kelkar A, Lemstra W, Theurkauf WE. DNreplication/DNA-damage-dependent centrosome inactivationDrosophilaembryos. Nat Cell Biol 2000;2(2):90–5.

76] Takada S, Kelkar A, Theurkauf WE.Drosophila checkpoint kinas2 couples centrosome function and spindle assembly to genintegrity. Cell 2003;113(1):87–99.

77] Gisselsson D, Jonson T, Yu C, Martins C, Mandahl N, Wiegantal. Centrosomal abnormalities, multipolar mitoses, and chromosinstability in head and neck tumours with dysfunctional telomeBr J Cancer 2002;87(2):202–7.

78] Sen S, Zhou H, White RA. A putative serine/threoninenase encoding gene BTAK on chromosome 20q13 is ampand overexpressed in human breast cancer cell lines. Onc1997;14(18):2195–200.

79] Zhou H, Kuang J, Zhong L, Kuo WL, Gray JW, Sahin A, etTumour amplified kinase STK15/BTAK induces centrosome amfication, aneuploidy and transformation [see comments]. Nat G1998;20(2):189–93.

Page 8: Centrosomal amplification and spindle multipolarity in cancer cells

32 W. Saunders / Seminars in Cancer Biology 15 (2005) 25–32

[80] Meraldi P, Honda R, Nigg EA. Aurora-A overexpression revealstetraploidization as a major route to centrosome amplification inp53−/− cells. Embo J 2002;21(4):483–92.

[81] Schliwa M, Pryzwansky KB, Borisy GG. Tumor promoter-inducedcentrosome splitting in human polymorphonuclear leukocytes. Eur JCell Biol 1983;32(1):75–85.

[82] Brinkley BR. Managing the centrosome numbers game: fromchaos to stability in cancer cell division. Trends Cell Biol2001;11(1):18–21.

[83] Sluder G, Nordberg JJ. The good, the bad and the ugly: the practi-cal consequences of centrosome amplification. Curr Opin Cell Biol2004;16(1):49–54.

[84] Tournebize R, Popov A, Kinoshita K, Ashford AJ, Rybina S, Poz-niakovsky A, et al. Control of microtubule dynamics by the antago-nistic activities of XMAP215 and XKCM1 inXenopusegg extracts.Nat Cell Biol 2000;2(1):13–9.

[85] Gergely F, Draviam VM, Raff JW. The ch-TOG/XMAP215 proteinis essential for spindle pole organization in human somatic cells.Genes Dev 2003;17(3):336–41.

[86] Braithwaite AW, Cheetham BF, Li P, Parish CR, Waldron-StevensLK, Bellett AJ. Adenovirus-induced alterations of the cell growthcycle: a requirement for expression of E1A but not of E1B. J Virol1983;45(1):192–9.

[87] Duensing S, Munger K. The human papillomavirus type 16 E6 andE7 oncoproteins independently induce numerical and structural chro-mosome instability. Cancer Res 2002;62(23):7075–82.

[88] Lavia P, Mileo AM, Giordano A, Paggi MG. Emerging roles ofDNA tumor viruses in cell proliferation: new insights into genomicinstability. Oncogene 2003;22(42):6508–16.

[89] Boyer SN, Wazer DE, Band V. E7 protein of human papillomavirus-16 induces degradation of retinoblastoma protein through theubiquitin-proteasome pathway. Cancer Res 1996;56(20):4620–4.

[90] Piboonniyom SO, Duensing S, Swilling NW, Hasskarl J, HindsPW, Munger K. Abrogation of the retinoblastoma tumor suppres-sor checkpoint during keratinocyte immortalization is not sufficientfor induction of centrosome-mediated genomic instability. CancerRes 2003;63(2):476–83.

[91] Duensing S, Munger K. Human papillomaviruses and centrosomeduplication errors: modeling the origins of genomic instability. Onco-gene 2002;21(40):6241–8.

[92] Duensing S, Duensing A, Crum CP, Munger K. Human papillo-mavirus type 16 E7 oncoprotein-induced abnormal centrosome syn-thesis is an early event in the evolving malignant phenotype. CancerRes 2001;61(6):2356–60.

[93] Yun C, Cho H, Kim SJ, Lee JH, Park SY, Chan GK. Mitotic aber-ration coupled with centrosome amplification is induced by hep-atitis B virus X oncoprotein via the Ras-mitogen-activated pro-tein/extracellular signal-regulated kinase-mitogen-activated proteinpathway. Mol Cancer Res 2004;2(3):159–69.

[94] Forgues M, Difilippantonio MJ, Linke SP, Ried T, Nagashima K,Feden J, et al. Involvement of Crm1 in hepatitis B virus X protein-induced aberrant centriole replication and abnormal mitotic spindles.Mol Cell Biol 2003;23(15):5282–92.

[95] Elmore LW, Hancock AR, Chang SF, Wang XW, Chang S, CallahanCP, et al. Hepatitis B virus X protein and p53 tumor suppressorinteractions in the modulation of apoptosis. Proc Natl Acad Sci USA1997;94(26):14707–12.