Vous êtes sur la page 1sur 12

The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633

Contents lists available at ScienceDirect

The International Journal of Biochemistry


& Cell Biology
journal homepage: www.elsevier.com/locate/biocel

Review

Actin cytoskeleton dynamics and the cell division cycle


Yi-Wen Heng, Cheng-Gee Koh ∗
School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore

a r t i c l e i n f o a b s t r a c t

Article history: The network of actin filaments is one of the crucial cytoskeletal structures contributing to the morpho-
Received 21 January 2010 logical framework of a cell and which participates in the dynamic regulation of cellular functions. In
Received in revised form 12 April 2010 adherent cell types, cells adhere to the substratum during interphase and spread to assume their charac-
Accepted 14 April 2010
teristic shape supported by the actin cytoskeleton. This actin cytoskeleton is reorganized during mitosis
Available online 20 April 2010
to form rounded cells with increased cortical rigidity. The actin cytoskeleton is re-established after mito-
sis, allowing cells to regain their extended shape and attachment to the substratum. The modulation of
Keywords:
such drastic changes in cell shape in coordination with cell cycle progression suggests a tight regulatory
Actin cytoskeleton
Cell cycle
interaction between cytoskeleton signalling, cell–cell/cell–matrix adhesions and mitotic events. Here,
Mitosis we review the contribution of the actin cytoskeleton to cell cycle progression with an emphasis on the
Rho GTPases effectors responsible for the regulation of the actin cytoskeleton and integration of their activities with
Focal adhesion the cell cycle machinery.
© 2010 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1622
2. Actin cytoskeleton, myosin and the cell cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1623
2.1. Actin cytoskeleton in cell cycle control . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1623
2.2. Actin, myosin and the regulation of the mitotic spindle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1623
2.3. Septin, actin cytoskeleton, and the cell cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1627
3. Rho GTPases, their regulators and the cell cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1627
3.1. RhoA and partners in cytokinesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1627
3.2. RhoA and partners in other stages of mitosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1628
3.3. Cdc42 and partners . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1629
3.4. Cyclin-dependent kinase and Rho GTPases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1629
4. Cell attachment and the cell cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1629
4.1. Integrin signalling and the cell cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1630
4.2. Cadherin signalling and the cell cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1630
4.3. Focal adhesion proteins and the cell cycle . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1630
5. Conclusion and perspective . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1631
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1631
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1631

1. Introduction

The actin cytoskeleton of eukaryotic cells undergoes drastic


Abbreviations: APC, adenomatous polyposis coli; CDK1, cyclin-dependent kinase changes and remodelling during cell division. Interphase cells usu-
1; ECM, extra cellular matrix; FAK, focal adhesion kinase; GAP, GTPase-activating ally contain an extensive actin network but this network is rapidly
protein; GEF, guanine nucleotide exchange factor; MLCK, myosin light chain kinase;
dismantled and rearranged when cells enter mitosis, giving mitotic
PAK, p21-activate kinase; Plk1, polo-like kinase 1; ROCK, p160-Rho-associated
coiled-coil-containing protein kinase. cells their characteristic round shape. At the end of mitosis, actin
∗ Corresponding author. Tel.: +65 63162854. rearranges at the cleavage furrows and forms part of the contractile
E-mail address: cgkoh@ntu.edu.sg (C.-G. Koh). ring, which is central to the process of cytokinesis. Another mitotic

1357-2725/$ – see front matter © 2010 Elsevier Ltd. All rights reserved.
doi:10.1016/j.biocel.2010.04.007
Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633 1623

event in which the actin network plays an important role is the somes and may possibly lead to the observed cytokinesis defects.
separation of centrosomes, which is dependent on the cortical flow These observations demonstrate actin’s involvement in cell cycle
of cortical actin and the myosin network. Disruption of the actin progression. Various proteins known to function in both the regu-
and myosin II networks by inhibitory drugs such as latrunculin, lation of the actin cytoskeleton and the cell cycle progression are
ROCK (p160-Rho-associated coiled-coil-containing protein kinase) summarized in Table 2. Many of these proteins change their cellular
inhibitor and myosin II RNA interference (RNAi) causes failure in localization at different phases of the cell cycle (Fig. 1 and Table 3).
centrosome separation and proper spindle assembly (Rosenblatt et Recently, cortactin, an actin-binding protein, has been identified
al., 2004; Uzbekov et al., 2002). as an anchor between the centrosome and F-actin and is essential
Thus, the regulation of the actin cytoskeleton and of cell cycle for F-actin driven centrosome separation during mitosis. The triply
progression appears to be connected. However, the nature of their phosphorylated form (Tyr421-, Tyr466- and Tyr482-) of cortactin
functional integration is not well understood. Here, we review is found to be localized exclusively to the spindle poles during tran-
the current state of knowledge concerning the regulatory links sition to anaphase. Truncated cortactin lacking its actin-binding
between these two activities in proliferating cells. The emphasis is domain inhibits centrosome separation (Wang et al., 2008). Inter-
on proteins known to regulate the actin cytoskeleton and are impli- estingly, cortactin has also been identified as a substrate of CDK1 at
cated in cell cycle control. Our discussion on the actin cytoskeleton serine 405 (Blethrow et al., 2008). Although the significance of this
will not be limited to the acto-myosin filament or filamentous actin phosphorylation of cortactin during mitosis has not been exam-
(F-actin) but will also encompass proteins associated with the mod- ined, it raises the possibility of a link between CDK1 signalling and
ulation of the actin cytoskeleton such as the Rho GTPases and their cortactin-mediated centrosomes separation during mitosis.
regulators. We will also discuss cell adhesions and their effect on Links between actin cytoskeleton and transcription control have
the cell cycle. also emerged. Disruption of the actin cytoskeleton during mitosis
leads to changes in the G- to F-actin ratio and hence in transcription
activities mediated by the myocardin-related transcription factor
2. Actin cytoskeleton, myosin and the cell cycle and serum response factor, MAL/SRF (Miralles et al., 2003). It is
possible that cell cycle progression could be affected by the status of
2.1. Actin cytoskeleton in cell cycle control actin polymerization via MAL/SRF mediated transcription. A recent
report shows that in human uterine leiomyosarcoma cells, down-
Actin is a highly conserved globular protein found in almost regulation of MAL leads to reduction of p21 CDK inhibitor (Kimura
all eukaryotic cells. It forms cellular scaffold structures that pro- et al., 2010). However, work by Triesman’s group has shown that
vide cells with their shape, tension support, intracellular vesicular depletion of SRF or MAL affects cell spreading and adhesion without
transport, cell attachment, adhesion properties and the ability to affecting cell proliferation or apoptosis (Medjkane et al., 2009).
move. Apart from these well-studied mechanical functions, actin
also plays a more subtle role in chemical signal transduction. It 2.2. Actin, myosin and the regulation of the mitotic spindle
was once thought that the cell cycle machinery controls the state
of actin organization within the cell via an “inside-out” signalling In mitosis, microtubules have hogged the limelight with their
mechanism (Wang, 1991; Yamashiro et al., 1991). However, retro- beautiful arrays and precisely choreographed functions in organ-
grade signalling where the state of actin organization within the cell ising events from the establishment of the bipolar spindle to the
controls cell cycle progression has proven to be important as well capturing, alignment and accurate segregation of chromosomes.
(Assoian and Zhu, 1997; Thery and Bornens, 2006). The significance Most importantly, mitotic spindle assembly and chromosome seg-
of the actin cytoskeleton for cell cycle progression can be easily regation can be reconstituted in vitro in cell free extract. The
gleaned from experiments using drugs or chemicals that interfere studies on actin in mitosis have remained focused almost solely
with the actin filament in the cells. Depolymerization of actin fila- on its mechanical function during cytokinesis until recently, where
ments by toxins such as cytochalasin D and latrunculin B has been actin’s role in the biogenesis of the mitotic spindle has gained
reported to delay progression of mitosis in primary cells and fission increasing attention. Using different methods of interference with
yeast, suggesting that an intact actin cytoskeleton may be required actin polymerization and actin-myosin at the cell cortex, two ear-
for the efficient onset of mitosis (Gachet et al., 2001; Lee and Song, lier papers clearly showed the requirement of myosin II and actin in
2007). A summary of the different drugs and chemicals and their centrosome separation in higher eukaryotic and mammalian cells
effect on the actin cytoskeleton is listed in Table 1. While a mor- (Rosenblatt et al., 2004; Uzbekov et al., 2002). Upon actin depoly-
phogenesis checkpoint has been proposed in budding yeast which merization with latrunculin treatment, centrosome separation is
is activated in response to perturbation of the actin cytoskele- blocked and a proper spindle cannot be assembled (Uzbekov et al.,
ton leading to delays in chromosome segregation (McMillan et al., 2002). Proper spindle assembly is also disrupted when myosin II is
1998), a similar actin regulated checkpoint control has not been inhibited through the use of the ROCK inhibitor – Y26732, which
established in mammalian cells. Apart from causing a delay in mito- prevents ROCK-mediated phosphorylation of myosin light chain
sis, disruption of actin filaments also leads to G1 arrest. This actin phosphatase, eventually blocking myosin activity. A similar effect
cytoskeleton dependent arrest has been linked to cyclin expres- is seen in cells treated with blebbistatin which inhibits the ATPase
sion and cyclin-dependent kinase (CDK) activation (Reshetnikova activity of non-muscle myosin II (Rosenblatt et al., 2004). Silencing
et al., 2000). In a study in which disruption of the actin cytoskele- of non-muscle myosin II heavy or light chain using RNA interfer-
ton was induced by the over-expression of cofilin, a member of ence also disrupts spindle formation. When the cortical flow of
the actin depolymerization factor (ADF)/cofilin family, more than acto-myosin filaments is prevented by cross-linking the cell sur-
90% of H1299 lung carcinoma cells arrested at G1 phase of the cell face with lectins such as concanavalin A, centrosome separation
cycle (Lee and Keng, 2005). Excessive polymerization of F-actin by and movement are impeded, which results in a lopsided spindle
a mutant WASP or the drug Jasplakinolide, which interferes with (Rosenblatt et al., 2004).
actin depolymerization, causes an increase in multinucleate cells More recent studies have illustrated a closer direct link between
suggesting a possible defect in cytokinesis (Moulding et al., 2007). the actin filaments and the mitotic spindles where F-actin was
Similarly, expression of mutant WASPI294T which mis-regulates found localized to the mitotic apparatus (Woolner et al., 2008;
the Arp2/3 complex and enhances F-actin polymerization, results Yasuda et al., 2005). Additional reports have also suggested that
in abnormal accumulation of F-actin around the mitotic chromo- the stability of the cortical actin network is crucial in establishing
1624 Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633

Fig. 1. Different localization of proteins at interphase and mitosis. HeLa cells were harvested at different stages of the cell cycle and immunostained with (A) Top panels:
anti-␣-tubulin antibodies. Bottom panels: phalloidin for F-actin. (B) Non-muscle myosin II heavy chain antibodies (red), DAPI (blue). (C) Top panels: anti-phospho LIMK1
(T508)/LIMK2 (T505) antibodies (green). Bottom panels: Merged image with anti-␣-tubulin (red) and DAPI stain (blue). (D) Top panels: anti-phospho PAK1 (T423) antibodies
(red). Bottom panels: merged image with anti-␣-tubulin (green) and DAPI (blue). All bars: 10 ␮m.
Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633 1625

Table 1
Summary of different reagents used to perturb the actin cytoskeleton and their effects on cell cycle progression.

Drug Target Action Phenotype(s) reported Reference(s)

Clostridium difficile toxin B RhoA, Rac1, Cdc42 Glycosylates RhoA, Rac1 and G2 arrest Ando et al. (2007)
Cdc42, thereby inactivating
them.
Clostridium botulinum RhoA, RhoB, RhoC ADP-ribosylation on Slows down p21 degradation, at Olson et al. (1995), Yamamoto et
exoenzyme C3 transferase asparagine 41 in the effector mitosis caused re-spreading of al. (1993)
binding domain of the GTPase prometaphase cells
Y-27632-Rock inhibitor ROCK I, ROCK II Competitive binding with ATP Failure in centrosome separation Rosenblatt et al. (2004)
to active site of ROCK I and
ROCK II
ML-7, ML-9 MLCK Competitive binding with ATP G1 arrest, affect spindle organization Deng et al. (2005), Bhadriraju and
to active site of MLCK Hansen (2004)
2,3-Butanedione monoxime Myosin Myosin ATPase inhibitor G1 arrest, inhibits Forer et al. (2007), Bhadriraju and
(BDM) kinetochore-microtubule elongation Hansen (2004), Huang et al. (1998)
Blebbistatin Myosin II Binds to Myosin-ADP-Pi with Failure in centrosome separation Rosenblatt et al. (2004)
high affinity and interferes
with phosphate release process
Calyculin A Protein phosphatases Binds to active site of PP1 and Accelerate anaphase chromosome Fabian et al. (2007)
PP2A protein phosphatases separation
CEP1347 Targets PAK1 ATP-binding site G1 arrest, aberrant spindle Nheu
PAK1 formation, delay in mitosis et
WR-PAK18 Binds SH3 domain of PIX,
blocking PAK-PIX interaction transition al.
Cytochalasin D Actin Binds to the barbed, fast Mitosis delay, G1 arrest, inhibits (2004)
Gachet et al. (2001), Lee and Song
growing plus ends of kinetochore-microtubule elongation (2007), Forer et al. (2007)
microfilaments, inhibiting
actin monomer assembly and
disassembly
Latrunculin A/B Actin Binds to actin monomers near Mitosis delay, G1 arrest, inhibits Rosenblatt et al. (2004), Uzbekov
their ATP-binding site, kinetochore-microtubule elongation, et al. (2002), Gachet et al. (2001),
preventing actin failure in centrosome separation Lee and Song (2007)
polymerization
Jasplakinolide Actin Binds to F-actin, stabilizing and Cytokinesis defect Moulding et al. (2007)
promoting actin
polymerization

correct spindle orientation in mammalian cells. The activity of LIM Besides spindle formation, actin and myosin can exert their
Kinase-1 (LIMK1) was found to increase during mitosis resulting in effects at different stages of the cell division. For example,
the phosphorylation and inactivation of cofilin (Kaji et al., 2008). myosin light chain kinase (MLCK) has been shown to function
Knockdown of LIMK1 leads to more activation and mislocalization in the phosphorylation of myosin II which is required for its
of cofilin, which in turn results in mis-orientation of the spindle. bundling with actin for contractile ring formation and for acto-
Similar defects can be induced by inhibition of actin polymerization myosin contractility forces that are necessary for cytokinesis
using latrunculin A or by over-expression of a non-phophorylatable (Mabuchi, 1986). MLCK activity also plays an important role in
cofilin (S3A). In addition, phosphorylated LIM kinases, but not the early mitotic events. Microinjection of the catalytic fragment of
unphosphorylated form, have been reported to colocalize and co- MLCK into prophase cells delays the transition from nuclear enve-
immunoprecipitate with ␥-tubulin during early stages of mitosis lope breakdown to the onset of anaphase, but does not affect the
(Chakrabarti et al., 2007). During metaphase, LIMK1 localises to duration between anaphase onset and mid cytokinesis. Unregu-
the centrosomes while LIMK2 associates mainly with the mitotic lated MLCK activity is also found to cause reduced fluorescent
spindle (Sumi et al., 2006). These data suggest that the activity of staining of spindle microtubules in prometaphase and metaphase
LIM kinases may play an important role in the regulation of spindle cells (Fishkind et al., 1991). Similarly, treatment of mouse eggs
activity during mitosis. with MLCK inhibitor ML-7, or the auto-inhibitory Peptide 18,
Apart from the interactions between the astral microtubule affects localization of actin cap on the metaphase II spindles
and the cortical actin network, other sites of interaction between and subsequent cortical reorganization activities (Deng et al.,
the spindle microtubules and acto-myosin filaments have long 2005).
been reported (Maupin and Pollard, 1986; Wu et al., 1998). More Although ROCK and myosin activities are required for the
recently, a myosin isoform Myo10, which binds to both actin and rounding up of cells during mitosis (Maddox and Burridge, 2003),
microtubules, has been found to localize to the poles of mitotic spin- the actual mechanism remains elusive. Additional evidence that
dles in Xenopus embryos (Woolner et al., 2008). Knocking down the actin structures can affect cell morphology during mitosis
Myo10 causes mitotic spindle defects which include fragmenta- came from the study of moesin. Moesin is a member of the
tion of the spindle poles and lengthening of the spindles. Dynamic ezrin/radixin/moesin (ERM) family of actin-binding proteins and
F-actin cables are also found localized to the mitotic spindles and has been implicated in cell rounding in mitosis. Phosphorylation of
the spindle poles. The authors proposed that F-actin and Myo10 moesin by the Ste20-like protein kinase Slik in Drosophila S2 cells
regulate spindle lengthening and shortening, respectively. While causes cell rounding in mitosis mainly via cross-linking actin to
the F-actin-mediated spindle lengthening is independent of Myo10, the membrane at the cortex to increase cortical rigidity (Carreno
spindle shortening induced by Myo10 requires F-actin. Thus F-actin et al., 2008; Kunda et al., 2008). Knocking down of moesin and
and Myo10 have both overlapping and distinct roles in mitosis. its upstream kinase Slik leads to defects in cell cortex organiza-
These observations suggest that the actin and microtubule struc- tion as well as metaphase spindle stability. Increasing cell rigidity
tures may act synergistically in the assembly and positioning of the externally by using lectin to crosslink the cell membrane can par-
mitotic apparatus. tially rescue the moesin knockdown phenotype suggesting that
1626 Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633

Table 2
Summary of the role of different groups of proteins in actin cytoskeleton regulation and cell cycle progression.

Protein Group Protein Roles in actin cytoskeleton Roles in cell cycle progression Reference(s)
regulation regulation

Myosin II Actin crosslinker; ATPase G1 -S progression; mitotic cell Rosenblatt et al. (2004),
dependent acto-myosin force rounding; centrosome separation; Uzbekov et al. (2002), Mabuchi
generation. mitotic spindle assembly; (1986), Fabian et al. (2007)
Actin binding
kinetochore-microtubule formation;
anaphase chromosome movement;
cytokinesis.
Myosin X Unconventional actin crosslinker. Mitotic spindle shortening. Woolner et al. (2008)
Anillin Actin and microtubule bundling. Spindle associated cleavage Piekny and Glotzer (2008),
specification. Gregory et al. (2008)
Cortactin Recruitment of Arp2/3 to F-actin. F-actin mediated centrosome Wang et al. (2008)
separation.
Moesin Actin binding; ERM protein. Mitotic cell rounding; mitotic cortical Carreno et al. (2008), Kunda et
rigidity; metaphase spindle stability. al. (2008)
Septin Actin binding; scaffold for Cytokinesis; chromosome congression Joo et al. (2007), Spiliotis et al.
non-muscle MyoII and its and segregation at mitosis (2005)
kinases.
RhoA Stress fibres formation. G1 -S transition; mitotic cell rounding; Coleman et al. (2006),
Rho GTPase mitotic cortical rigidity; cytokinesis. Nishimura and Yonemura
(2006), Birkenfeld et al. (2007)
Rac1 Lamellapodia formation. G1 -S transition; cytokinesis. Canman et al. (2008), Klein et
al. (2009)
Cdc42 Filopodia formation. G1 -S transition; Yasuda et al. (2004, 2006), Jaffe
kinetochore-microtubule stabilization; et al. (2008), Mitsushima et al.
spindle biorientation; metaphase (2009)
chromosome alignment; cytokinesis.
p190RhoGAP RhoA GAP. Mitotic cell rounding. Maddox and Burridge (2003)
Ect2 RhoA GEF, Cdc42 GEF during Contractile ring formation; contraction Nishimura and Yonemura
mitosis. of contractile ring at cleavage furrow. (2006), Oceguera-Yanez et al.
Rho GTPase Regulator
(2005)
GEF-H1 RhoA GEF; microtubule RhoA related cytokinesis. Birkenfeld et al. (2007)
dynamics.
MgcRacGAP Rac1 and Cdc42 GAP; RhoA GAP Formation of ingression furrow; Minoshima et al. (2003), Zhao
during mitosis. interacts with Ect2. and Fang (2005), Miller and
Bement (2009)
Lfc RhoA GEF. Mitotic spindle assembly. Bakal et al. (2005)
MyoGEF RhoA GEF; myosin II binding. Localize RhoA and Ect2 to contractile Asiedu et al. (2009)
ring.
ROCK RhoA effector; myosin S phase progression, mitotic cell Rosenblatt et al. (2004),
regulation; actin bundling. rounding; centrosome separation; Uzbekov et al. (2002), Croft and
mitotic spindle assembly; cytokinesis. Olson (2006)
Rho GTPase effector and LIMK1 ROCK & PAK substrates; cofilin Mitotic spindle orientation. Kaji et al. (2008)
down-stream target LIMK2 regulation; actin bundling. Regulate cyclin A and p27 expression. Croft and Olson (2006)
Cofilin Downstream of RhoA pathway; G1 -S progression; mitotic spindle Lee and Keng (2005), Kaji et al.
actin severing. orientation. (2008)
mDia1 RhoA effector; actin nucleation Spindle formation in early mitosis Bakal et al. (2005)
and elongation. through an “Lfc-RhoA-mDia1”
pathway.
PRK2/PKN2 RhoA and Rac effector. Mitosis entry; mitosis exit. Schmidt et al. (2007)
PAK1 Rac and Cdc42 effector; cell G1 -S transition; G2 -M transition; Maroto et al. (2008), Zhao et al.
motility; focal adhesion turnover. centrosome maturation; regulation of (2005), Vadlamudi et al.
Plk1 and Aurora-A activity; regulate (2000), Nheu et al. (2004),
astral microtubule dynamics; spindle Balasenthil et al. (2004)
orientation.
N-WASP Rac and Cdc42 effector; Arp2/3 Cytokinesis. Moulding et al. (2007)
activator; actin nucleation.

the maintenance of cell shape and rigidity is sufficient to stabi- results in the stabilization of myosin II phosphorylation and the
lize the mitotic spindles. Moesin’s function in rounding up of cells subsequent acceleration of pole-ward movement of chromosomes
appears to be independent of myosin II because cells expressing during anaphase (Fabian et al., 2007).
active T559D moesin but lacking myosin light chain are still able These studies strongly imply that the formation of mitotic
to round up during mitosis (Kunda et al., 2008). However, it is spindle is highly dependent on the actin and myosin networks
very likely that both moesin-actin and myosin-actin activities are within the cell. Actin reorganization during mitosis helps cre-
required to establish the rounded cell shape and rigidity for proper ate the cellular environment required for the mitotic spindle
spindle assembly and positioning. to serve its function. Disruption of the cortical actin architec-
Actin and myosin structures also participate in generating ture during mitosis can severely affect spindle orientation which
the forces required for chromosome segregation. Treatment of may result in cell cycle arrest. There may well exist in mam-
crane-fly spermatocytes with actin depolymerization drugs such malian cells, like in the budding yeast, a morphogenesis checkpoint
as cytochalasin D and lantrunculin A or myosin ATPase inhibitors which is dependent on the actin cytoskeleton (Lew, 2003) or
like butanedione monoxime (BDM) causes inhibition of spindle a spindle orientation checkpoint (Gachet et al., 2006) which
microtubule elongation (Forer et al., 2007). Conversely, nonspe- hinges on the interaction between the microtubule and cortical
cific inhibition of myosin light chain phosphatase by calyculin A actin, although the existence of a spindle orientation checkpoint
Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633 1627

Table 3
The localization of different proteins linked to the actin cytoskeleton in interphase and mitosis.

Protein Subcellular localization by immunofluorescence staining Reference(s)

Interphase (adherent cell type) M-phase

F-actin Stress fibers; actin structures Contractile ring Schroeder (1968)


Myosin II Stress fibers; actin structures Contractile ring Mabuchi and Okuno (1977)
Anillin Ubiquitous Contractile ring Piekny and Glotzer (2008)
Septin Stress fibres Contractile ring Joo et al. (2007)
Cortactin Ubiquitous, actin structures Centrosomes (phosphorylated from) Wang et al. (2008)
Ect2 Ubiquitous Central spindle Nishimura and Yonemura (2006)
GEF-H1 Ubiquitous Mitotic spindle; midzone Birkenfeld et al. (2007)
LIMK1 Cell–cell contacts Centrosomes; equatorial cortex; contractile ring Sumi et al. (2006)
LIMK2 Ubiquitous Mitotic spindle, contractile ring Sumi et al. (2006)
MyoGEF Ubiquitous Central spindle Asiedu et al. (2009)
PAK1 Ubiquitous; actin structures; centrosomes Centrosomes; contractile ring; mid-body Zhao et al. (2005)
RhoA Ubiquitous Contractile ring Yuce et al. (2005)
HEF1 Focal adhesions Mitotic spindle; mid-body Law et al. (1998)
Integrin-linked kinase (ILK) Focal adhesions Centrosomes Fielding et al. (2008)
Focal adhesion kinase (FAK) Focal adhesions Centrosones Rodriguez-Fernandez et al. (1999)
Pyk2 Focal adhesions Centrosomes Rodriguez-Fernandez et al. (1999)
Paxillin Focal adhesions Centrosomes Herreros et al. (2000)
Zyxin Focal adhesions Mitotic spindle; central spindle Hirota et al. (2000)

in the fission yeast is now questionable (Meadows and Millar, increasing acto-myosin contraction and bundling of actin filament
2008). whereas active Cdc42 and Rac1 increase filopodia and lamellipo-
dia formation, respectively. Apart from their roles in modulating
the actin cytoskeleton, Rho GTPases have also been found to reg-
2.3. Septin, actin cytoskeleton, and the cell cycle
ulate cell cycle progression with two main sites of action: one at
the G1 /S transition and the other during cytokinesis. For example,
Septins are a family of GTPase which can polymerize to form fil-
inhibition of Cdc42, Rac1 and RhoA results in cell cycle arrest at G1
amentous structures. They were originally discovered in the screen
phase of the cell cycle (Olson et al., 1995; Yamamoto et al., 1993). In
for cell division mutants in the budding yeast (Hartwell, 1971).
human capillary endothelial cells, active RhoA causes an increase
Their main role is in the control of cytokinesis. A ring of septin
in the expression of the F-box containing protein Skp2 which is
polymers is assembled during early stages of the cell cycle at the
required for ubiquitinylation-dependent degradation of the CDK
bud neck and remains till cytokinesis (Cid et al., 2001). Apart from
inhibitor p27kip1 (Mammoto et al., 2004). p27kip1 binds to and inac-
cytokinesis, septins have also been implicated in the regulation of
tivates the cyclin D1/CDK4 and cyclin E/CDK2 complexes. Absence
GIN4 kinase activation required for bud growth (Carroll et al., 1998).
of active RhoA leads to high levels of p27kip1 , resulting in cell cycle
Mammalian septins are found to associate with the plasma
arrest in G1 . Active RhoA acts via the balance of activities of its
membrane, actin cytosketon and the microtubules (Spiliotis and
two effectors, ROCK and mDia, to activate the Skp2-p27kip1 path-
Nelson, 2006). Mammalian Septin2 colocalizes with actin stress
way. It has also been shown that inhibition of RhoA or disruption
fibres during the interphase and the contractile ring at cytokine-
of F-actin drastically slows down the degradation of another CDK
sis. It has also been shown to bind to non-muscle myosin II (Joo
inhibitor p21Waf/Cip1 (Coleman et al., 2006). Specifically, ROCK has
et al., 2007). In HeLa and MDCK cells, septins are localized to the
been shown to regulate cyclin A expression via the Ras/MAPK path-
metaphase plate during mitosis (Spiliotis et al., 2005). Knocking
way and via LIMK2 (Croft and Olson, 2006). More recently, a study
down of septins results in loss of chromosomes from the metaphase
using the Clostridium difficile toxin B to inhibit the Rho GTPases at
plate. It has been suggested that the septins form a scaffold at the
the G2 phase of the cell cycle also reveals possible involvement
midplane of mitotic spindle to maintain CENP-E motor protein at
of the Rho GTPases in the control of multiple signalling pathways
the kinetochores thereby facilitating the congression of chromo-
involved in the progression to mitosis (Ando et al., 2007). The dif-
somes at the metaphase plate. Interestingly, Kremer et al. have
ferent pathways regulated by Rho GTPases at different phases of
shown that septins present in the cytoplasm bind to and act as a
the cell cycle are summarized in Fig. 2.
reservoir for SOCS7 (suppressor of cytokine signalling 7). Knock-
ing down of septins 2, 6 and 7 causes loss of stress fibres and
also nuclear accumulation of NCK (Kremer et al., 2007). SOCS7 is
3.1. RhoA and partners in cytokinesis
responsible for the import of NCK into the nucleus. DNA dam-
ages also result in the nuclear accumulation of SOCS7 and NCK.
Although RhoA activities have been reported to be required for
Hence septins are also linked to the DNA damage checkpoint via
cell cycle progression at different phases, most studies have con-
the septin-SOCS7-NCK pathway. Given that NCK is associated with
centrated on the role of RhoA in cytokinesis where the activity
the control of actin cytoskeleton, there is a possibility that cell cycle
of RhoA is essential. RhoA organises the assembly of the contrac-
progression and actin cytoskeleton can influence each other via this
tile ring and induces the acto-myosin-driven constriction of the
pathway.
cleavage furrow [reviewed by Barr and Gruneberg, 2007]. ECT2,
a guanine nucleotide exchange factor (GEF) which activates RhoA,
3. Rho GTPases, their regulators and the cell cycle localizes and activates RhoA at the cleavage furrow (Nishimura and
Yonemura, 2006). GEF-H1, a microtubule-regulated GEF for RhoA,
The small GTPases belonging to the Rho family have long has also been shown to modulate RhoA activity during cytokine-
been associated with the regulation and remodelling of the actin sis (Birkenfeld et al., 2007). These GEFs are, in turn, activated by
cytoskeleton and are important for cell motility, morphogene- CDK1/cyclin B and Aurora-A/B kinases (Birkenfeld et al., 2007; Hara
sis and neurite development (Etienne-Manneville and Hall, 2002; et al., 2006; Niiya et al., 2006). Through the use of chemical genetics
Koh, 2006). Active RhoA causes the formation of stress fibres by and specific inhibitors, Polo-like kinase Plk1, originally thought to
1628 Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633

Fig. 2. Functional connections between proteins involved in the regulation of Rho GTPases, the actin cytoskeleton and cell cycle progression. The activities of different
proteins and how they can influence one another at different phases of the cell cycle are summarized.

regulate only spindle assembly, has been found to also control the activities, thereby maintaining a RhoA-rich zone at the cell equator
localization of ECT2 to the central spindle and RhoA at the equator (Miller and Bement, 2009).
in anaphase (Brennan et al., 2007; Burkard et al., 2007; Petronczki et Anillin, a 124 kDa RhoA binding protein, has been shown to accu-
al., 2007). Inhibition of Plk1 blocks the interaction of ECT2 with the mulate at the cleavage furrow in a RhoA dependent manner (Piekny
midzone anchor HsCyk4 and the assembly of the contractile ring, and Glotzer, 2008). Anillin was first isolated as an actin binding and
resulting in the inhibition of cytokinesis and the formation of binu- bundling protein in Drosophila embryo (Field and Alberts, 1995).
cleate cells. Recently, centrosome/spindle pole-associated protein It was later shown to interact with myosin regulatory light chain
(CSPP) has been found to target MyoGEF (Myosin II-interacting GEF) (MLC) and septin (Kinoshita et al., 2002; Oegema et al., 2000;
to the central spindle during anaphase. MyoGEF also interacts with Straight et al., 2005). A recent report reveals that in Drosophila the
ECT2. Knockdown of MyoGEF results in mislocalization of ECT2 RhoGEF pebble controls the formation of a filamentous structure
and RhoA during cytokinesis (Asiedu et al., 2009). Centralspindlin containing Rho1, anillin and septin at the cleavage furrow, as well
which is localized to both the central spindle microtubules and the as the interaction of this filamentous structure with the plasma
tips of astral microtubules near the equatorial cortex is reported membrane and microtubules (Hickson and O’Farrell, 2008). Inter-
to recruit ECT2 to the central spindle (Nishimura and Yonemura, estingly, anillin also interacts with RacGAP50C, which is a spindle
2006). Knockdown of centralspindlin component, MKLP1, causes associated protein that specifies the cleavage site (Gregory et al.,
failure of ECT2 to localize to the equatorial cell cortex (Yuce et al., 2008). Depletion of anillin results in the loss of RacGAP50C from
2005). the equator of cleavage and the collapse of the cleavage furrow.
Another Rho GTPase regulator which participates in the con- Thus anillin acts as a linker between the contractile ring and the
trol of cytokinesis and contractile ring assembly is MgcRacGAP spindle.
(also known as HsCyk4). MgcRacGAP is a GTPase-activating pro-
tein for Rac and Cdc42 but is converted to a GAP for RhoA when 3.2. RhoA and partners in other stages of mitosis
phophorylated by Aurora-B in M phase of the cell cycle (Minoshima
et al., 2003). Silencing of MgcRacGAP by RNAi results in the loss of Other than cytokinesis, RhoA activities are also required in other
ingression of the cleavage furrow and hence failure of the cells to phases of the cell cycle. Microtubule-associated Rho GEF Lfc has
undergo cytokinesis (Zhao and Fang, 2005). MgcRacGAP interacts been implicated in spindle formation in early mitosis in Rat2 fibrob-
with ECT2 and therefore may exert its effect via ECT2. A more recent last cells (Bakal et al., 2005). Lfc knockdown or microinjection
report proposes a model where the GAP activities of MgcRacGAP of anti-Lfc antibody causes spindle assembly defects in the early
and GEF activity of ECT2 act concurrently to promote a flux of RhoA stages of mitosis. Both active RhoA and the formin mDia1 can res-
Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633 1629

cue these defects, suggesting that an “Lfc-RhoA-mDia1” pathway of the centrosome in the late G2 phase. PAK1 activity may also play
is working in Rat2 cells for spindle formation during early mito- an important role in the regulation of astral microtubule dynam-
sis. Additionally, via the use of novel covalent capturing method of ics during mitosis since over-expression of active PAK1 resulted in
kinase-specific phosphopeptides, mDia1 was found to contain an multiple spindle orientations (Vadlamudi et al., 2000).
optimal CDK1 phosphorylation consensus sequence (Blethrow et Studies have also linked PAK1 signalling to G1 to S phase transi-
al., 2008), suggesting a possible direct connection between CDK1 tion via the regulation of the cyclin D1 machinery. It has been shown
and mDia1 during mitosis. that in Ras transformed NIH 3T3 cells, the addition of two distinct
In addition to mDia1 and ROCK, another RhoA effector, and specific PAK1-3 inhibitors, CEP-1347 and WR-PAK18 was able
PRK2/PKN2, has also been implicated in the onset of mitosis and the to block malignant growth by down-regulation of cyclin D1 (Nheu
completion of cytokinesis (Schmidt et al., 2007). Silencing of PRK2 et al., 2004). Consistent with this, perturbation of PAK1 activity by
in HeLa S3 cells leads to accumulation of bi-nucleated cells and PAK1-KID, or knockdown of PAK1 by siRNA resulted in a marked
these cells also show delay in G2 /M progression. PRK2 contributes decrease in cyclin D1 expression (Balasenthil et al., 2004). So far,
positively towards the phosphorylation of Cdc25B, leading to the a direct linkage of Cdc42 or Rac1 activity to these activities of PAK
activation of CDK1/cyclin B. These effectors appear to constitute has not been documented, even though a concomitant increase in
a feedback loop as active CDK1/cyclinB can further phosphorylate Cdc42 and PAK1 activity has been reported (Oceguera-Yanez et al.,
PRK2 which can then participate in the abscission process during 2005). Thus PAK1 may exhibit its function in a GTPase-dependent
cytokinesis. or independent pathway.
Two recent papers have illustrated that Cdc42 is important in
3.3. Cdc42 and partners controlling spindle orientation in mitotic cells (Jaffe et al., 2008;
Mitsushima et al., 2009). Deletion of Cdc42 did not affect cell polar-
The level of the guanosine 5 -triphosphate (GTP) bound form ity but instead caused mis-orientation of the spindle leading to
of Cdc42 changes during cell cycle progression. The abundance of inappropriate positioning of the apical surfaces after cell division
Cdc42-GTP is low during pre-metaphase, peaks during metaphase (Jaffe et al., 2008). Mitsushima et al. (2009) went further to demon-
and declines again at telophase. However, there is no change in strate that two independent pathways downstream of Cdc42 are
Rac-GTP levels throughout the cell cycle (Oceguera-Yanez et al., involved in regulating spindle orientation: one involving Cdc42-
2005). The level of RhoA-GTP, on the other hand, peaks at telophase. PAK2-␤PIX and the other involving phosphatidylinositol 3 kinase
Over-expression of dominant-negative ECT2 and MgcRacGAP as (PI3K). It appears that the two independent pathways collectively
well as silencing of ECT2 by RNAi prevented the change in the affect the phosphatidyl 3,4,5 triphosphate (PIP3) levels and the
level of Cdc42-GTP in mitosis (Oceguera-Yanez et al., 2005). These cortical actin structures (Mitsushima et al., 2009).
observations suggest that both ECT2 and MgcRacGAP can regulate Not much has been reported about Rac1 and its role in the con-
both RhoA and Cdc42 at different stages of the cell cycle. More trol of the cell cycle. One recent report suggests that the inhibition
specifically, while ECT2 catalyses the formation of Cdc42-GTP at of Rac by the CYK-4/MgcRacGAP of the centralspindlin complex is
metaphase, MgcRacGAP increases the hydrolysis of Cdc42-GTP at essential for cytokinesis in C. elegans (Canman et al., 2008). Deple-
prometaphase. tion of Rac but not RhoA can rescue the cytokinesis defect of a CYK-4
The spatial and temporal function of Cdc42-GTP is required for GAP mutant. This work suggests a parallel inhibition of Rac and acti-
the progression through mitosis. Reduction in the activity of Cdc42 vation of RhoA during cytokinesis, most probably to prevent the
by RNAi causes a delay in mitotic progression. When Cdc42 RNAi activation of Arp2/3 complex by WASP or WAVE which is down-
is combined with the knockdown of other Cdc42-like GTPases, a stream of Rac. The net result is the prevention of the formation of
high proportion of the cells showed misalignment of chromosomes other actin networks which may interfere with the contractile ring.
(Yasuda et al., 2006). It has been suggested that one of the major
roles of Cdc42 in mitosis is to control biorientation and stabilization
3.4. Cyclin-dependent kinase and Rho GTPases
of the kinetochore-microtubule attachment via its association and
activation of the formin protein mDia3 (Yasuda et al., 2004), rather
That the onset of mitosis requires activation of CDK1 and is
than the modulation of the actin cytoskeleton. Another formin pro-
accompanied by drastic rearrangement of the actin cytoskeleton
tein mDia2 is reported to have microtubule stabilizing activities,
leading to the rounding up of the cell, suggests an intimate reg-
which is independent of its actin nucleation activity (Bartolini et
ulatory connection between CDK1 and actin cytoskeleton. While
al., 2008). mDia2 has been shown to bind to microtubules directly
the exact signalling pathways of CDK1 activity leading to mitotic
as well as to the microtubule tip proteins such as EB1 and APC. The
cytoskeletal changes remain poorly understood, p190 Rho GTPase-
actin cytoskeleton related role for mDia2 in the regulation of cell
activating protein (GAP) has been proposed as a major downstream
cycle has been attributed to the stabilization of the actin scaffold
effector of CDK1 (Maddox and Burridge, 2003). Activated CDK1
for the contractile ring during cytokinesis (Watanabe et al., 2008).
phosphorylates p190RhoGAP, down regulating its activity and thus
The p21-activated kinases (PAKs), a family of serine-threonine
decreases GTP hydrolysis by RhoA. This triggers a signalling cascade
kinases which are effectors of Cdc42 and Rac1, have been impli-
through ROCK and MLC phosphatase, that regulates cytoskeleton
cated in the control of G2 /M transition. It has been shown that
rearrangement observed in mitosis (Amano et al., 1996; Maddox
PAK1 regulates Plk1 activity. Inhibition of PAK1 activity leads to
and Burridge, 2003).
a delay in G2 /M and aberrant spindle assembly which are also the
phenotypes that result from Plk1 inactivation (Maroto et al., 2008).
PAK1 has also been shown to activate Aurora-A kinase at the cen- 4. Cell attachment and the cell cycle
trosome (Zhao et al., 2005). PAK1 is targeted to the centrosome by
the PIX-GIT1 complex. Studies have also proposed that PAK1 local- Attachment of cells to the extracellular matrix (ECM) or other
ization to the centrosome during metaphase to anaphase transition cells has long been implicated in cell cycle regulation. During cell
requires its kinase activity as the expression of a kinase inhibitory division, the cells undergo extensive cell shape changes to detach
domain, PAK1-KID, causes a drastic reduction in centrosomal tar- from and reattach to the ECM. While cell–matrix adhesions have
geting (Li et al., 2002). When PAK1 is activated at the centrosome, been reported to reduce during mitosis, cell–cell adhesions via the
it dissociates from PIX-GIT1 and is able to phosphorylate and acti- desmosomes, tight junctions and zonulae adherentes in epithelial
vate Aurora-A. Aurora-A activation is required for the maturation cells are maintained throughout cell divisions (Baker and Garrod,
1630 Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633

1993; Jinguji and Ishikawa, 1992; Reinsch and Karsenti, 1994). In ingly, ␤-catenin has also been shown to have an unexpected role
mammalian cells, inter-dependence of cell attachment and cell as a component of the inter-centrosomal linker and is essential
cycle signalling can be attributed to integrin and cadherin signals for the establishment of bipolar spindle (Bahmanyar et al., 2008).
(Pugacheva et al., 2006). Knocking-out or silencing of focal adhe- Although ␤-catenin is thought to be the major effector of cad-
sion proteins vinculin, paxillin and the adaptor protein CRK has herin engagement for cell cycle regulation, ␣-catenin has also been
been shown to result in the fusion of daughter cells leading to bin- reported to influence cell cycle regulation. ␣-catenin has been
ucleate cells, demonstrating that these proteins are essential for shown to bind to various actin-binding proteins such as ␣-actinin
the completion of cytokinesis (Nagasaki et al., 2009; Shafikhani et (Knudsen et al., 1995), vinculin (Hazan et al., 1997; Watabe-Uchida
al., 2008). et al., 1998; Weiss et al., 1998), Ajuba (Marie et al., 2003), spectrin
A recent paper reported a positive correlation between ECM (Pradhan et al., 2001), ZO-1 (Itoh et al., 1997), formin (Kobielak
stiffness and progression into S phase of the cell cycle in mam- et al., 2004) and afadin (Pokutta et al., 2002), suggesting these
mary epithelial cells and vascular smooth muscle cells (Klein et actin-binding proteins may play associative roles in the modula-
al., 2009). Using hydrogel to simulate physiological stiffness, it has tion of adherens junctions-mediated cell cycle control. Another
been shown that an increase in matrix stiffness results in selective component of the adherens junctions, p120 catenin, is also impli-
integrin activation, leading to localization and activation of focal cated in the regulation of mitosis. Loss of p120 catenin results in
adhesion kinase (FAK) which eventually induces RAC1 activation mitotic defects leading to extended M phase and binucleate cells
and cyclin D1 expression. (Perez-Moreno et al., 2008). This has been attributed to the abnor-
mally high RhoA activity in the p120 catenin conditional-knockout
4.1. Integrin signalling and the cell cycle cells.
Additional evidence that cell–cell adhesion plays an important
Integrins associate with numerous proteins and localise at the role in determining spindle orientation comes from the work of
focal contacts. Upon engagement with ECM, they activate proxi- den Elzen et al. (2009). They have reported that E-Cadherin can
mal signalling proteins such as FAK, SRC and CAS family members, provide cues to orient the mitotic spindle during symmetric cell
which in turn signal through several signalling cascades, e.g. the divisions in mammalian epithelia (den Elzen et al., 2009). When
RAP and B-RAF pathway, the PI3K, RAC, AKT and PAK pathway, dominant-negative E-Cadherin is introduced into MDCK cells, more
and the SHC, GRB2, RAS and RAF pathway. These three pathways than half of the cells showed mis-oriented spindles. Moreover,
converge to activate MEK and ERK kinases and together activate cortical APC (adenomatous polyposis coli) staining, which is consis-
G1 -specific cyclins D and E (Pugacheva et al., 2006). In addition, tently localized to the cell cortex at the apicolateral region, is lost in
integrin-activated FAK can directly phosphorylate transcription cells expressing dominant-negative E-Cadherin. Silencing of APC by
factor KLF8, leading to its nuclear translocation and the activation of siRNA abolishes junctional staining of APC and causes spindle mis-
cyclin D1 promoter (Zhao et al., 2003). Integrins also activate differ- orientation, suggesting that APC may mediate cadherin signalling
ent GEFs, which lead to the activation of the Rho GTPases. However, to orient the mitotic spindle.
it has been reported that focal adhesion signalling and cell spread- In another study using Drosophila neuroepithelial cells, sym-
ing are dispensable for progression through the cell cycle as long metric mode of cell division can be converted into asymmetric
as there is sufficient cyclin D1 (Margadant et al., 2007). division upon the disruption of the adherens junctions. The APC
Integrin mediated cell adhesion and the ECM can also control protein which is localized at the adherens junctions is involved in
the orientation of the spindles and hence determine the spindle the maintenance of the symmetric mode of division. The APC and
axis and the plane of cell division (Thery et al., 2005; Toyoshima and the microtubule-associated EB1 proteins function together to ori-
Nishida, 2007). ␤1-Integrin knockout results in random spindle ori- ent the mitotic spindle to provide the polarity cues for symmetric
entation and a high incidence of binucleate cells (Aszodi et al., 2003; division (Lu et al., 2001).
Lechler and Fuchs, 2005). The use of an inactive ␤1-integrin mutant
reveals a role for integrin in bipolar spindle assembly and cytoki- 4.3. Focal adhesion proteins and the cell cycle
nesis (Reverte et al., 2006). In addition to its role in focal adhesion
regulation, integrin-linked kinase (ILK) has also been localized to In adherent cell types, cell rounding upon entry into mitosis is
the centrosome and is implicated in mitotic spindle assembly and accompanied by a reduction in the focal contacts and an increase
chromosome segregation (Fielding et al., 2008). However, it is not in cortical rigidity. After cytokinesis, cells reattach to their substra-
clear if the roles of ILK at the focal adhesions and the centrosomes tum and re-establish cytoskeletal networks. The concerted changes
are dependent on each other. in cell shape and adhesion as cells prepare to enter mitosis sug-
Integrin signalling may also serve as an important link between gest intense cross-signalling events between focal contacts and cell
the small GTPase Rap1 and mitosis. The activity of Rap1 is regulated cycle signalling. This is evident from the observation that focal con-
during mitosis. Inhibition of Rap1 is required for focal adhesion dis- tacts are more densely distributed in the proximity of signalling
assembly at the onset of mitosis whereas Rap1 activation is needed molecules such as protein kinase C (Liao and Jaken, 1993), tyrosine
for cell spreading after mitosis (Dao et al., 2009). Rap1 has also kinases (Hanks et al., 1992; Schaller et al., 1992) and tyrosine phos-
been shown to activate many integrins (Bos, 2005; Caron, 2003). It phatases (Serra-Pages et al., 1995; Shen et al., 1998). It is possible
is very likely that Rap1 modulates integrin signalling and thereby that post translational modifications play a major role in elicit-
influences the cell shape changes which accompany mitosis. ing such drastic changes within a few minutes. Indeed, HEF1, FAK,
actopaxin, paxillin and PAK are phosphorylated at mitosis-specific
4.2. Cadherin signalling and the cell cycle sites (Clarke et al., 2004; Law et al., 1998; Yamakita et al., 1999)
during early M phase. In addition, proteins associated with focal
Cadherins are Ca2+ -dependent transmembrane proteins that adhesion contacts such as FAK (Rodriguez-Fernandez et al., 1999),
participate in cell–cell adhesion. In stable adherens junctions, cad- Pyk2 (Rodriguez-Fernandez et al., 1999), paxillin (Herreros et al.,
herin assembles with ␣-catenin, ␤-catenin and actin filaments 2000) and zyxin (Hirota et al., 2000) are also found to associate
to form stable quaternary complexes that limit cell growth via with the mitotic spindle or the microtubules-organizing centre dur-
contact-inhibition. In the absence of cell–cell contacts, ␤-catenin ing mitosis (Table 3). The dual roles focal adhesion proteins play in
is displaced from cadherin and translocates to the nucleus thereby adhesion-mediated signalling and mitosis-associated events sug-
activating cyclin D1 transcription (Yamada et al., 2005). Interest- gest a precise temporal control of molecular events ensuring that
Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633 1631

one set of events is completed before the next phase of the cell cycle affinity chromatography and characterized by mass spectrometry.
is initiated. In this screen, ROCK2 (an effector of RhoA) was found to be an inter-
FAK serves as an important signalling protein at focal adhe- acting protein of Plk1 as well as being its substrate. Plk1 can control
sion sites to mediate focal adhesion formation, cell migration and the local activation of RhoA during cytokinesis and phosphorylate
cell cycle progression. Over-expression of FAK has been associated ROCK2 directly and stimulate ROCK2 activity (Lowery et al., 2007;
with invasiveness in a variety of human tumours (Owens et al., Yoshida et al., 2006).
1995). In interphase, FAK mediates cyclin D1 and p21 CDK inhibitor It is very likely that CDK1 plays a central role in integrating the
expression downstream of integrin engagement. Over-expression mechanisms regulating the actin cytoskeleton and mitosis. CDK1,
of dominant-negative FAK causes G1 arrest in mouse fibroblasts through its activity on GEFs and GAPs, could affect the activities
(Zhao et al., 1998). During mitosis, FAK is targeted to the centro- of RhoGTPases. These GTPases can further regulate their effector
some (Rodriguez-Fernandez et al., 1999), suggesting that FAK may proteins such as PAK which in turn can regulate Plk1 and Aurora-A
regulate centrosome functions. This is consistent with the obser- kinases to modulate spindle dynamics. The activities of the RhoGT-
vation that deletion of FAK in endothelial cells leads to multiple Pases and actin-myosin filaments have been shown to be essential
centrosome formation, multi-polar and disorganized mitotic spin- for spindle formation and for cytokinesis. The same proteins may
dles and misalignment of chromosomes during metaphase (Park et be required at different stages of the cell cycle and may function
al., 2009). differently to ensure successful cell division (Fig. 2). A better under-
At the end of mitosis, integrins function to reattach the cells standing of the mutual regulatory relationship between the cell
to its substratum, followed by the establishment of a spread-out cycle and the actin cytoskeleton will have important implications
shape and cytoplasmic tension. Similar to cell rounding during for many diseases. This is especially so for cancer biology as many
mitosis, these processes are likely regulated by post transla- therapeutic interventions involve interference with cell division of
tional modifications of the attachment proteins. Activation of the cancer cells.
anaphase promoting complex/cyclosome (APC/C) during late mito-
sis may degrade proteins that target the HEF1, zyxin and other Acknowledgements
attachment proteins to mitotic structures, thereby allowing focal
contacts to reform. However, the exact mechanism of focal con- We thank the Academic Research Fund, MOE Tier 2 and the
tact re-establishment and reformation of stress fibers following Biomedical Research Council, ASTAR Singapore for research fund-
cytokinesis is poorly understood. It remains unclear if the formation ing.
of focal contacts precedes cytoskeletal tension or vice versa. Cells
treated with trypsin also show loss of focal contacts and stress fibers References
accompanied by cell rounding. However, when replated, these cells
re-establish focal contacts and spreading. Mitotic cells, on the other Amano M, Ito M, Kimura K, Fukata Y, Chihara K, Nakano T, et al. Phosphorylation
hand, stay detached until the completion of mitotic events. It is and activation of myosin by Rho-associated kinase (Rho-kinase). J Biol Chem
1996;271:20246–9.
likely that cells use distinct mechanisms to promote or inhibit Ando Y, Yasuda S, Oceguera-Yanez F, Narumiya S. Inactivation of Rho GTPases with
cytoskeletal events at different stages of cell cycle. Clostridium difficile toxin B impairs centrosomal activation of Aurora-A in G2 /M
transition of HeLa cells. Mol Biol Cell 2007;18:3752–63.
Asiedu M, Wu D, Matsumura F, Wei Q. Centrosome/spindle pole-associated protein
regulates cytokinesis via promoting the recruitment of MyoGEF to the central
5. Conclusion and perspective spindle. Mol Biol Cell 2009;20:1428–40.
Assoian RK, Zhu X. Cell anchorage and the cytoskeleton as partners in growth factor
dependent cell cycle progression. Curr Opin Cell Biol 1997;9:93–8.
Despite the extensive evidence suggesting that the disruption of Aszodi A, Hunziker EB, Brakebusch C, Fassler R. Beta1 integrins regulate chondrocyte
the actin cytoskeleton can lead to cellular arrest, many aspects of rotation, G1 progression, and cytokinesis. Genes Dev 2003;17:2465–79.
the regulatory relationship between cytoskeletal integrity and cell Bahmanyar S, Kaplan DD, Deluca JG, Giddings Jr TH, O’Toole ET, Winey M, et al.
beta-Catenin is a Nek2 substrate involved in centrosome separation. Genes Dev
cycle progression remain to be elucidated. In particular, it is unclear
2008;22:91–105.
if a checkpoint-like mechanism is responsible to ensure the coor- Bakal CJ, Finan D, LaRose J, Wells CD, Gish G, Kulkarni S, et al. The Rho GTP exchange
dination of these two sets of events. More detailed studies of the factor Lfc promotes spindle assembly in early mitosis. Proc Natl Acad Sci USA
2005;102:9529–34.
direct targets of some key cell cycle regulators such as CDK1, Plk1,
Baker J, Garrod D. Epithelial cells retain junctions during mitosis. J Cell Sci
Aurora kinases which are also actin cytoskeleton regulators should 1993;104(Pt 2):415–25.
provide further insight into how cell cycle progression can regulate Balasenthil S, Sahin AA, Barnes CJ, Wang RA, Pestell RG, Vadlamudi RK, et al.
the actin cytoskeleton and vice versa. Cdc28, the CDK responsible p21-activated kinase-1 signaling mediates cyclin D1 expression in mammary
epithelial and cancer cells. J Biol Chem 2004;279:1422–8.
for cell cycle progression in Saccharomyces cerevisiae and Candida Barr FA, Gruneberg U. Cytokinesis: placing and making the final cut. Cell
albicans, has been reported to phosphorylate and regulate the activ- 2007;131:847–60.
ity of a Rho GEF and IQGAP1, respectively (Kono et al., 2008; Li et Bartolini F, Moseley JB, Schmoranzer J, Cassimeris L, Goode BL, Gundersen GG.
The formin mDia2 stabilizes microtubules independently of its actin nucleation
al., 2008). Since CDK1 alone is sufficient to drive cell division in activity. J Cell Biol 2008;181:523–36.
mammalian cells (Santamaria et al., 2007), it is possible that some Bhadriraju K, Hansen LK. Extracellular matrix dependent myosin dynamics during
regulatory proteins implicated in maintaining the actin cytoskele- G1-S phase cell cycle progression in hepatocytes. Exp Cell Res 2004;300:259–71.
Birkenfeld J, Nalbant P, Bohl BP, Pertz O, Hahn KM, Bokoch GM. GEF-H1 modu-
ton could be direct targets of CDK1. Many possible CDK1(Cdc28) lates localized RhoA activation during cytokinesis under the control of mitotic
targets have been identified in S. cerevisiae (Ubersax et al., 2003) kinases. Dev Cell 2007;12:699–712.
and in HeLa cells (Blethrow et al., 2008). A thorough analysis of the Blethrow JD, Glavy JS, Morgan DO, Shokat KM. Covalent capture of kinase-specific
phosphopeptides reveals Cdk1-cyclin B substrates. Proc Natl Acad Sci USA
roles of these CDK1 targets could lead to the identification of pos-
2008;105:1442–7.
sible regulators of the actin cytoskeleton. Other key effectors, such Bos JL. Linking Rap to cell adhesion. Curr Opin Cell Biol 2005;17:123–8.
as polo kinase, Aurora-A and ubiquitin ligases that play important Brennan IM, Peters U, Kapoor TM, Straight AF. Polo-like kinase controls vertebrate
spindle elongation and cytokinesis. PLoS ONE 2007;2:e409.
roles in mitotic progression, may also serve as critical regulatory
Burkard ME, Randall CL, Larochelle S, Zhang C, Shokat KM, Fisher RP, et al. Chemical
links between the actin cytoskeleton and the mitotic machinery. A genetics reveals the requirement for Polo-like kinase 1 activity in position-
connection between Plk1 and the actin cytoskeleton regulators has ing RhoA and triggering cytokinesis in human cells. Proc Natl Acad Sci USA
been found in a screen for the Plk1 interactome in U2OS cells at 2007;104:4383–8.
Canman JC, Lewellyn L, Laband K, Smerdon SJ, Desai A, Bowerman B, et al. Inhibition
different stages of the cell cycle (Lowery et al., 2007). The interac- of Rac by the GAP activity of centralspindlin is essential for cytokinesis. Science
tome represents partners for a particular protein, isolated through 2008;322:1543–6.
1632 Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633

Caron E. Cellular functions of the Rap1 GTP-binding protein: a pattern emerges. J Kimura Y, Morita T, Hayashi K, Miki T, Sobue K. Myocardin functions as an effective
Cell Sci 2003;116:435–40. inducer of growth arrest and differentiation in human uterine leiomyosarcoma
Carreno S, Kouranti I, Glusman ES, Fuller MT, Echard A, Payre F. Moesin and its acti- cells. Cancer Res 2010;70:501–11.
vating kinase Slik are required for cortical stability and microtubule organization Kinoshita M, Field CM, Coughlin ML, Straight AF, Mitchison TJ. Self- and actin-
in mitotic cells. J Cell Biol 2008;180:739–46. templated assembly of mammalian septins. Dev Cell 2002;3:791–802.
Carroll CW, Altman R, Schieltz D, Yates JR, Kellogg D. The septins are required for Klein EA, Yin L, Kothapalli D, Castagnino P, Byfield FJ, Xu T, et al. Cell-cycle con-
the mitosis-specific activation of the Gin4 kinase. J Cell Biol 1998;143:709–17. trol by physiological matrix elasticity and in vivo tissue stiffening. Curr Biol
Chakrabarti R, Jones JL, Oelschlager DK, Tapia T, Tousson A, Grizzle WE. Phosphory- 2009;19:1511–8.
lated LIM kinases colocalize with gamma-tubulin in centrosomes during early Knudsen KA, Soler AP, Johnson KR, Wheelock MJ. Interaction of alpha-actinin with
stages of mitosis. Cell Cycle 2007;6:2944–52. the cadherin/catenin cell–cell adhesion complex via alpha-catenin. J Cell Biol
Cid VJ, Adamikova L, Sanchez M, Molina M, Nombela C. Cell cycle control of 1995;130:67–77.
septin ring dynamics in the budding yeast. Microbiology 2001;147:1437– Kobielak A, Pasolli HA, Fuchs E. Mammalian formin-1 participates in adherens junc-
50. tions and polymerization of linear actin cables. Nat Cell Biol 2004;6:21–30.
Clarke DM, Brown MC, LaLonde DP, Turner CE. Phosphorylation of actopaxin regu- Koh CG. Rho GTPases and their regulators in neuronal functions and development.
lates cell spreading and migration. J Cell Biol 2004;166:901–12. Neurosignals 2006;15:228–37.
Coleman ML, Densham RM, Croft DR, Olson MF. Stability of p21Waf1/Cip1 CDK Kono K, Nogami S, Abe M, Nishizawa M, Morishita S, Pellman D, et al. G1 /S cyclin-
inhibitor protein is responsive to RhoA-mediated regulation of the actin dependent kinase regulates small GTPase Rho1p through phosphorylation of
cytoskeleton. Oncogene 2006;25:2708–16. RhoGEF Tus1p in Saccharomyces cerevisiae. Mol Biol Cell 2008;19:1763–71.
Croft DR, Olson MF. The Rho GTPase effector ROCK regulates cyclin A, cyclin D1, and Kremer BE, Adang LA, Macara IG. Septins regulate actin organization and cell-
p27Kip1 levels by distinct mechanisms. Mol Cell Biol 2006;26:4612–27. cycle arrest through nuclear accumulation of NCK mediated by SOCS7. Cell
Dao VT, Dupuy AG, Gavet O, Caron E, de Gunzburg J. Dynamic changes in Rap1 2007;130:837–50.
activity are required for cell retraction and spreading during mitosis. J Cell Sci Kunda P, Pelling AE, Liu T, Baum B. Moesin controls cortical rigidity, cell rounding,
2009;122:2996–3004. and spindle morphogenesis during mitosis. Curr Biol 2008;18:91–101.
den Elzen N, Buttery CV, Maddugoda MP, Ren G, Yap AS. Cadherin adhesion recep- Law SF, Zhang YZ, Klein-Szanto AJ, Golemis EA. Cell cycle-regulated processing of
tors orient the mitotic spindle during symmetric cell division in mammalian HEF1 to multiple protein forms differentially targeted to multiple subcellular
epithelia. Mol Biol Cell 2009;20:3740–50. compartments. Mol Cell Biol 1998;18:3540–51.
Deng M, Williams CJ, Schultz RM. Role of MAP kinase and myosin light chain Lechler T, Fuchs E. Asymmetric cell divisions promote stratification and differentia-
kinase in chromosome-induced development of mouse egg polarity. Dev Biol tion of mammalian skin. Nature 2005;437:275–80.
2005;278:358–66. Lee K, Song K. Actin dysfunction activates ERK1/2 and delays entry into mitosis in
Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature 2002;420:629–35. mammalian cells. Cell Cycle 2007;6:1487–95.
Fabian L, Troscianczuk J, Forer A. Calyculin A, an enhancer of myosin, speeds up Lee YJ, Keng PC. Studying the effects of actin cytoskeletal destabilization on cell cycle
anaphase chromosome movement. Cell Chromosome 2007;6:1. by cofilin overexpression. Mol Biotechnol 2005;31:1–10.
Field CM, Alberts BM. Anillin, a contractile ring protein that cycles from the nucleus Lew DJ. The morphogenesis checkpoint: how yeast cells watch their figures. Curr
to the cell cortex. J Cell Biol 1995;131:165–78. Opin Cell Biol 2003;15:648–53.
Fielding AB, Dobreva I, McDonald PC, Foster LJ, Dedhar S. Integrin-linked kinase Li CR, Wang YM, Wang Y. The IQGAP Iqg1 is a regulatory target of CDK for cytokinesis
localizes to the centrosome and regulates mitotic spindle organization. J Cell in Candida albicans. EMBO J 2008;27:2998–3010.
Biol 2008;180:681–9. Li F, Adam L, Vadlamudi RK, Zhou H, Sen S, Chernoff J, et al. p21-activated kinase 1
Fishkind DJ, Cao LG, Wang YL. Microinjection of the catalytic fragment of myosin interacts with and phosphorylates histone H3 in breast cancer cells. EMBO Rep
light chain kinase into dividing cells: effects on mitosis and cytokinesis. J Cell 2002;3:767–73.
Biol 1991;114:967–75. Liao L, Jaken S. Effect of alpha-protein kinase C neutralizing antibodies and the pseu-
Forer A, Spurck T, Pickett-Heaps JD. Actin and myosin inhibitors block elongation dosubstrate peptide on phosphorylation, migration, and growth of REF52 cells.
of kinetochore fibre stubs in metaphase crane-fly spermatocytes. Protoplasma Cell Growth Differ 1993;4:309–16.
2007;232:79–85. Lowery DM, Clauser KR, Hjerrild M, Lim D, Alexander J, Kishi K, et al. Proteomic
Gachet Y, Reyes C, Goldstone S, Tournier S. The fission yeast spindle orien- screen defines the Polo-box domain interactome and identifies Rock2 as a Plk1
tation checkpoint: a model that generates tension? Yeast 2006;23:1015– substrate. EMBO J 2007;26:2262–73.
29. Lu B, Roegiers F, Jan LY, Jan YN. Adherens junctions inhibit asymmetric division in
Gachet Y, Tournier S, Millar JB, Hyams JS. A MAP kinase-dependent actin checkpoint the Drosophila epithelium. Nature 2001;409:522–5.
ensures proper spindle orientation in fission yeast. Nature 2001;412:352–5. Mabuchi I. Biochemical aspects of cytokinesis. Int Rev Cytol 1986;101:175–213.
Gregory SL, Ebrahimi S, Milverton J, Jones WM, Bejsovec A, Saint R. Cell division Mabuchi I, Okuno M. The effect of myosin antibody on the division of starfish blas-
requires a direct link between microtubule-bound RacGAP and Anillin in the tomeres. J Cell Biol 1977;74:251–63.
contractile ring. Curr Biol 2008;18:25–9. Maddox AS, Burridge K. RhoA is required for cortical retraction and rigidity during
Hanks SK, Calalb MB, Harper MC, Patel SK. Focal adhesion protein-tyrosine kinase mitotic cell rounding. J Cell Biol 2003;160:255–65.
phosphorylated in response to cell attachment to fibronectin. Proc Natl Acad Sci Mammoto A, Huang S, Moore K, Oh P, Ingber DE. Role of RhoA, mDia, and ROCK in cell
USA 1992;89:8487–91. shape-dependent control of the Skp2-p27kip1 pathway and the G1 /S transition.
Hara T, Abe M, Inoue H, Yu LR, Veenstra TD, Kang YH, et al. Cytokinesis regulator ECT2 J Biol Chem 2004;279:26323–30.
changes its conformation through phosphorylation at Thr-341 in G2 /M phase. Margadant C, van Opstal A, Boonstra J. Focal adhesion signaling and actin stress
Oncogene 2006;25:566–78. fibers are dispensable for progression through the ongoing cell cycle. J Cell Sci
Hartwell LH. Genetic control of the cell division cycle in yeast. IV. Genes controlling 2007;120:66–76.
bud emergence and cytokinesis. Exp Cell Res 1971;69:265–76. Marie H, Pratt SJ, Betson M, Epple H, Kittler JT, Meek L, et al. The LIM protein Ajuba
Hazan RB, Kang L, Roe S, Borgen PI, Rimm DL. Vinculin is associated with the E- is recruited to cadherin-dependent cell junctions through an association with
cadherin adhesion complex. J Biol Chem 1997;272:32448–53. alpha-catenin. J Biol Chem 2003;278:1220–8.
Herreros L, Rodriguez-Fernandez JL, Brown MC, Alonso-Lebrero JL, Cabanas C, Maroto B, Ye MB, von Lohneysen K, Schnelzer A, Knaus UG. P21-activated kinase is
Sanchez-Madrid F, et al. Paxillin localizes to the lymphocyte microtubule orga- required for mitotic progression and regulates Plk1. Oncogene 2008;27:4900–8.
nizing center and associates with the microtubule cytoskeleton. J Biol Chem Maupin P, Pollard TD. Arrangement of actin filaments and myosin-like filaments in
2000;275:26436–40. the contractile ring and of actin-like filaments in the mitotic spindle of dividing
Hickson GR, O’Farrell PH. Rho-dependent control of anillin behavior during cytoki- HeLa cells. J Ultrastruct Mol Struct Res 1986;94:92–103.
nesis. J Cell Biol 2008;180:285–94. McMillan JN, Sia RA, Lew DJ. A morphogenesis checkpoint monitors the actin
Hirota T, Morisaki T, Nishiyama Y, Marumoto T, Tada K, Hara T, et al. Zyxin, a regulator cytoskeleton in yeast. J Cell Biol 1998;142:1487–99.
of actin filament assembly, targets the mitotic apparatus by interacting with Meadows JC, Millar J. Latrunculin A delays anaphase onset in fission yeast by dis-
h-warts/LATS1 tumor suppressor. J Cell Biol 2000;149:1073–86. rupting an Ase1-independent pathway controlling mitotic spindle stability. Mol
Huang S, Chen CS, Ingber DE. Control of cyclin D1, p27(Kip1), and cell cycle progres- Biol Cell 2008;19:3713–23.
sion in human capillary endothelial cells by cell shape and cytoskeletal tension. Medjkane S, Perez-Sanchez C, Gaggioli C, Sahai E, Treisman R. Myocardin-related
Mol Biol Cell 1998;9:3179–93. transcription factors and SRF are required for cytoskeletal dynamics and exper-
Itoh M, Nagafuchi A, Moroi S, Tsukita S. Involvement of ZO-1 in cadherin-based cell imental metastasis. Nat Cell Biol 2009;11:257–68.
adhesion through its direct binding to alpha catenin and actin filaments. J Cell Miller AL, Bement WM. Regulation of cytokinesis by Rho GTPase flux. Nat Cell Biol
Biol 1997;138:181–92. 2009;11:71–7.
Jaffe AB, Kaji N, Durgan J, Hall A. Cdc42 controls spindle orientation to position the Minoshima Y, Kawashima T, Hirose K, Tonozuka Y, Kawajiri A, Bao YC, et al. Phos-
apical surface during epithelial morphogenesis. J Cell Biol 2008;183:625–33. phorylation by Aurora B converts MgcRacGAP to a RhoGAP during cytokinesis.
Jinguji Y, Ishikawa H. Electron microscopic observations on the maintenance of the Dev Cell 2003;4:549–60.
tight junction during cell division in the epithelium of the mouse small intestine. Miralles F, Posern G, Zaromytidou AI, Treisman R. Actin dynamics control SRF activity
Cell Struct Funct 1992;17:27–37. by regulation of its coactivator MAL. Cell 2003;113:329–42.
Joo E, Surka MC, Trimble WS. Mammalian SEPT2 is required for scaffolding nonmus- Mitsushima M, Toyoshima F, Nishida E. Dual role of Cdc42 in spindle orientation
cle myosin II and its kinases. Dev Cell 2007;13:677–90. control of adherent cells. Mol Cell Biol 2009;29:2816–27.
Kaji N, Muramoto A, Mizuno K. LIM kinase-mediated cofilin phosphorylation Moulding DA, Blundell MP, Spiller DG, White MR, Cory GO, Calle Y, et al. Unregulated
during mitosis is required for precise spindle positioning. J Biol Chem actin polymerization by WASp causes defects of mitosis and cytokinesis in X-
2008;283:4983–92. linked neutropenia. J Exp Med 2007;204:2213–24.
Y.-W. Heng, C.-G. Koh / The International Journal of Biochemistry & Cell Biology 42 (2010) 1622–1633 1633

Nagasaki A, Kanada M, Uyeda TQ. Cell adhesion molecules regulate contractile ring- Spiliotis ET, Kinoshita M, Nelson WJ. A mitotic septin scaffold required for mam-
independent cytokinesis in Dictyostelium discoideum. Cell Res 2009;19:236–46. malian chromosome congression and segregation. Science 2005;307:1781–5.
Nheu T, He H, Hirokawa Y, Walker F, Wood J, Maruta H. PAK is essential for RAS- Spiliotis ET, Nelson WJ. Here come the septins: novel polymers that coordinate
induced upregulation of cyclin D1 during the G1 to S transition. Cell Cycle intracellular functions and organization. J Cell Sci 2006;119:4–10.
2004;3:71–4. Straight AF, Field CM, Mitchison TJ. Anillin binds nonmuscle myosin II and regulates
Niiya F, Tatsumoto T, Lee KS, Miki T. Phosphorylation of the cytokinesis regula- the contractile ring. Mol Biol Cell 2005;16:193–201.
tor ECT2 at G2 /M phase stimulates association of the mitotic kinase Plk1 and Sumi T, Hashigasako A, Matsumoto K, Nakamura T. Different activity regulation and
accumulation of GTP-bound RhoA. Oncogene 2006;25:827–37. subcellular localization of LIMK1 and LIMK2 during cell cycle transition. Exp Cell
Nishimura Y, Yonemura S. Centralspindlin regulates ECT2 and RhoA accumulation Res 2006;312:1021–30.
at the equatorial cortex during cytokinesis. J Cell Sci 2006;119:104–14. Thery M, Bornens M. Cell shape and cell division. Curr Opin Cell Biol 2006;18:648–57.
Oceguera-Yanez F, Kimura K, Yasuda S, Higashida C, Kitamura T, Hiraoka Y, et al. Thery M, Racine V, Pepin A, Piel M, Chen Y, Sibarita JB, et al. The extracellular matrix
Ect2 and MgcRacGAP regulate the activation and function of Cdc42 in mitosis. J guides the orientation of the cell division axis. Nat Cell Biol 2005;7:947–53.
Cell Biol 2005;168:221–32. Toyoshima F, Nishida E. Integrin-mediated adhesion orients the spindle paral-
Oegema K, Savoian MS, Mitchison TJ, Field CM. Functional analysis of a human homo- lel to the substratum in an EB1- and myosin X-dependent manner. EMBO J
logue of the Drosophila actin binding protein anillin suggests a role in cytokinesis. 2007;26:1487–98.
J Cell Biol 2000;150:539–52. Ubersax JA, Woodbury EL, Quang PN, Paraz M, Blethrow JD, Shah K, et al. Targets of
Olson MF, Ashworth A, Hall A. An essential role for Rho, Rac, and Cdc42 GTPases in the cyclin-dependent kinase Cdk1. Nature 2003;425:859–64.
cell cycle progression through G1 . Science 1995;269:1270–2. Uzbekov R, Kireyev I, Prigent C. Centrosome separation: respective role of micro-
Owens LV, Xu L, Craven RJ, Dent GA, Weiner TM, Kornberg L, et al. Overexpression tubules and actin filaments. Biol Cell 2002;94:275–88.
of the focal adhesion kinase (p125FAK) in invasive human tumors. Cancer Res Vadlamudi RK, Adam L, Wang RA, Mandal M, Nguyen D, Sahin A, et al. Regulatable
1995;55:2752–5. expression of p21-activated kinase-1 promotes anchorage-independent growth
Park AY, Shen TL, Chien S, Guan JL. Role of focal adhesion kinase Ser-732 phospho- and abnormal organization of mitotic spindles in human epithelial breast cancer
rylation in centrosome function during mitosis. J Biol Chem 2009;284:9418–25. cells. J Biol Chem 2000;275:36238–44.
Perez-Moreno M, Song W, Pasolli HA, Williams SE, Fuchs E. Loss of p120 catenin and Wang W, Chen L, Ding Y, Jin J, Liao K. Centrosome separation driven by
links to mitotic alterations, inflammation, and skin cancer. Proc Natl Acad Sci actin-microfilaments during mitosis is mediated by centrosome-associated
USA 2008;105:15399–404. tyrosine-phosphorylated cortactin. J Cell Sci 2008;121:1334–43.
Petronczki M, Glotzer M, Kraut N, Peters JM. Polo-like kinase 1 triggers the initiation Wang YL. Dynamics of the cytoskeleton in live cells. Curr Opin Cell Biol
of cytokinesis in human cells by promoting recruitment of the RhoGEF Ect2 to 1991;3:27–32.
the central spindle. Dev Cell 2007;12:713–25. Watabe-Uchida M, Uchida N, Imamura Y, Nagafuchi A, Fujimoto K, Uemura T, et al.
Piekny AJ, Glotzer M. Anillin is a scaffold protein that links RhoA, actin, and myosin alpha-Catenin-vinculin interaction functions to organize the apical junctional
during cytokinesis. Curr Biol 2008;18:30–6. complex in epithelial cells. J Cell Biol 1998;142:847–57.
Pokutta S, Drees F, Takai Y, Nelson WJ, Weis WI. Biochemical and structural def- Watanabe S, Ando Y, Yasuda S, Hosoya H, Watanabe N, Ishizaki T, et al. mDia2 induces
inition of the l-afadin- and actin-binding sites of alpha-catenin. J Biol Chem the actin scaffold for the contractile ring and stabilizes its position during cytoki-
2002;277:18868–74. nesis in NIH 3T3 cells. Mol Biol Cell 2008;19:2328–38.
Pradhan D, Lombardo CR, Roe S, Rimm DL, Morrow JS. alpha-Catenin binds directly Weiss EE, Kroemker M, Rudiger AH, Jockusch BM, Rudiger M. Vinculin is part of
to spectrin and facilitates spectrin-membrane assembly in vivo. J Biol Chem the cadherin-catenin junctional complex: complex formation between alpha-
2001;276:4175–81. catenin and vinculin. J Cell Biol 1998;141:755–64.
Pugacheva EN, Roegiers F, Golemis EA. Interdependence of cell attachment and cell Woolner S, O’Brien LL, Wiese C, Bement WM. Myosin-10 and actin filaments are
cycle signaling. Curr Opin Cell Biol 2006;18:507–15. essential for mitotic spindle function. J Cell Biol 2008;182:77–88.
Reinsch S, Karsenti E. Orientation of spindle axis and distribution of plasma Wu X, Kocher B, Wei Q, Hammer III JA. Myosin Va associates with microtubule-
membrane proteins during cell division in polarized MDCKII cells. J Cell Biol rich domains in both interphase and dividing cells. Cell Motil Cytoskeleton
1994;126:1509–26. 1998;40:286–303.
Reshetnikova G, Barkan R, Popov B, Nikolsky N, Chang LS. Disruption of the actin Yamada S, Pokutta S, Drees F, Weis WI, Nelson WJ. Deconstructing the cadherin-
cytoskeleton leads to inhibition of mitogen-induced cyclin E expression, Cdk2 catenin-actin complex. Cell 2005;123:889–901.
phosphorylation, and nuclear accumulation of the retinoblastoma protein- Yamakita Y, Totsukawa G, Yamashiro S, Fry D, Zhang X, Hanks SK, et al. Dissociation
related p107 protein. Exp Cell Res 2000;259:35–53. of FAK/p130(CAS)/c-Src complex during mitosis: role of mitosis-specific serine
Reverte CG, Benware A, Jones CW, LaFlamme SE. Perturbing integrin function inhibits phosphorylation of FAK. J Cell Biol 1999;144:315–24.
microtubule growth from centrosomes, spindle assembly, and cytokinesis. J Cell Yamamoto M, Marui N, Sakai T, Morii N, Kozaki S, Ikai K, et al. ADP-ribosylation of
Biol 2006;174:491–7. the rhoA gene product by botulinum C3 exoenzyme causes Swiss 3T3 cells to
Rodriguez-Fernandez JL, Gomez M, Luque A, Hogg N, Sanchez-Madrid F, Cabanas C. accumulate in the G1 phase of the cell cycle. Oncogene 1993;8:1449–55.
The interaction of activated integrin lymphocyte function-associated antigen 1 Yamashiro S, Yamakita Y, Hosoya H, Matsumura F. Phosphorylation of non-muscle
with ligand intercellular adhesion molecule 1 induces activation and redistribu- caldesmon by p34cdc2 kinase during mitosis. Nature 1991;349:169–72.
tion of focal adhesion kinase and proline-rich tyrosine kinase 2 in T lymphocytes. Yasuda H, Kanda K, Koiwa H, Suenaga K, Kidou S, Ejiri S. Localization of actin filaments
Mol Biol Cell 1999;10:1891–907. on mitotic apparatus in tobacco BY-2 cells. Planta 2005;222:118–29.
Rosenblatt J, Cramer LP, Baum B, McGee KM. Myosin II-dependent cortical move- Yasuda S, Oceguera-Yanez F, Kato T, Okamoto M, Yonemura S, Terada Y, et al.
ment is required for centrosome separation and positioning during mitotic Cdc42 and mDia3 regulate microtubule attachment to kinetochores. Nature
spindle assembly. Cell 2004;117:361–72. 2004;428:767–71.
Santamaria D, Barriere C, Cerqueira A, Hunt S, Tardy C, Newton K, et al. Yasuda S, Taniguchi H, Oceguera-Yanez F, Ando Y, Watanabe S, Monypenny J, et
Cdk1 is sufficient to drive the mammalian cell cycle. Nature 2007;448: al. An essential role of Cdc42-like GTPases in mitosis of HeLa cells. FEBS Lett
811–5. 2006;580:3375–80.
Schaller MD, Borgman CA, Cobb BS, Vines RR, Reynolds AB, Parsons JT. pp125FAK a Yoshida S, Kono K, Lowery DM, Bartolini S, Yaffe MB, Ohya Y, et al. Polo-like kinase
structurally distinctive protein-tyrosine kinase associated with focal adhesions. Cdc5 controls the local activation of Rho1 to promote cytokinesis. Science
Proc Natl Acad Sci USA 1992;89:5192–6. 2006;313:108–11.
Schmidt A, Durgan J, Magalhaes A, Hall A. Rho GTPases regulate PRK2/PKN2 to Yuce O, Piekny A, Glotzer M. An ECT2-centralspindlin complex regulates the local-
control entry into mitosis and exit from cytokinesis. EMBO J 2007;26:1624–36. ization and function of RhoA. J Cell Biol 2005;170:571–82.
Schroeder TE. Cytokinesis: filaments in the cleavage furrow. Exp Cell Res Zhao J, Bian ZC, Yee K, Chen BP, Chien S, Guan JL. Identification of transcription factor
1968;53:272–6. KLF8 as a downstream target of focal adhesion kinase in its regulation of cyclin
Serra-Pages C, Kedersha NL, Fazikas L, Medley Q, Debant A, Streuli M. The LAR D1 and cell cycle progression. Mol Cell 2003;11:1503–15.
transmembrane protein tyrosine phosphatase and a coiled-coil LAR-interacting Zhao JH, Reiske H, Guan JL. Regulation of the cell cycle by focal adhesion kinase. J
protein co-localize at focal adhesions. EMBO J 1995;14:2827–38. Cell Biol 1998;143:1997–2008.
Shafikhani SH, Mostov K, Engel J. Focal adhesion components are essential for mam- Zhao WM, Fang G. MgcRacGAP controls the assembly of the contractile ring and the
malian cell cytokinesis. Cell Cycle 2008;7:2868–76. initiation of cytokinesis. Proc Natl Acad Sci USA 2005;102:13158–63.
Shen Y, Schneider G, Cloutier JF, Veillette A, Schaller MD. Direct associa- Zhao ZS, Lim JP, Ng YW, Lim L, Manser E. The GIT-associated kinase PAK targets to
tion of protein-tyrosine phosphatase PTP-PEST with paxillin. J Biol Chem the centrosome and regulates Aurora-A. Mol Cell 2005;20:237–49.
1998;273:6474–81.

Vous aimerez peut-être aussi