You are on page 1of 12

Oncogene (2006) 25, 2285–2296

& 2006 Nature Publishing Group All rights reserved 0950-9232/06 $30.00
www.nature.com/onc

ORIGINAL ARTICLE

RhoC GTPase is required for PC-3 prostate cancer cell invasion but not
motility

H Yao1, EJ Dashner1, CM van Golen2 and KL van Golen1


1
Division of Hematology/Oncology, The Department of Internal Medicine, The University of Michigan Comprehensive Cancer
Center, Ann Arbor, MI, USA and 2The Department of Neurology, The University of Michigan Medical School, Ann Arbor, MI, USA

It is projected that in 2005, approximately 220 900 men majority of them will have extensive bone metastasis
will be newly diagnosed with carcinoma of the prostate (Alcover et al., 1994; Abate-Shen and Shen, 2000;
(CaP). Men who are diagnosed with locally advanced or American Cancer Society, 2005). Metastatic bone
metastatic disease undergo androgen ablation therapy and lesions are associated with intractable pain, bone
most will relapse and progress within 18 months. fracture and spinal compression that can lead to
Metastasis to bone is the major clinical concern during paralysis (Cancer Management: A Multidisciplinary
CaP progression, as it is associated with intractable pain, Approach, 1999; Thalmann et al., 2000). Currently,
bone fracture and paralysis resulting from spinal cord the mechanisms that underlie CaP metastasis are poorly
compression. Therefore, an understanding of the key understood. Therefore, molecules involved in CaP cell
mechanisms involved in CaP cell bone metastasis is vital motility and invasion must be identified to design novel
to development of novel treatments. The Rho GTPases are therapeutic strategies for metastatic CaP. Recent studies
molecular switches involved in cell survival, motility and have focused on Ras homology (Rho) kinase (a.k.a.
invasion. Increased expression of RhoC GTPase is linked ROCK), the downstream effector protein of the mono-
to enhanced metastatic potential in multiple cancers; meric G-proteins RhoA and RhoC (Somlyo et al., 2000,
however, the role of RhoC GTPase in CaP metastasis has 2003). The ROCK inhibitor Wf-536, used in conjunction
not been addressed. In the current study, we demonstrate with the matrix metalloproteinase inhibitor Marimastat,
that RhoC GTPase is expressed and active in PC-3 CaP significantly inhibits experimental tumor growth, tumor
cells. RhoC inhibition, either pharmacologically with C3 angiogenesis and increased survival of mice bearing PC-
exotransferase or molecularly through expression of a 3 xenotransplants (Somlyo et al., 2003). Other investi-
dominant-negative RhoC, promotes IGF-I stimulated gators have demonstrated that treatment of PC-3 cells
random motility but decreases in vitro invasion and with the ROCK inhibitor Y27632 retards in vitro PC-3
experimental metastases. Inhibition of RhoC activity cell migration (Somlyo et al., 2000). These results
results in drastic morphologic changes and alterations in underscore the importance of the molecules that lie
the expression and distribution of focal adhesion-related upstream of ROCK, specifically, the Rho GTPases.
proteins. These data suggest that RhoC inhibition leads to The Rho GTPases belong to the Ras superfamily of
activation of other GTPases involved in nondirected small GTP binding proteins and act as molecular switches
motility and that expression of active RhoC is required involved in all aspects of cellular motility and invasion,
for the invasive phenotype of PC-3 cells. including polarity, cytoskeletal organization and signal
Oncogene (2006) 25, 2285–2296. doi:10.1038/sj.onc.1209260; transduction from the extracellular environment (Kjoller
published online 28 November 2005 and Hall, 1999; Price and Collard, 2001; Ridley, 2001;
Takai et al., 2001). Like Ras, Rho cycles between an
Keywords: RhoC GTPase; Rac1 GTPase; insulin-like inactive GDP-bound and active GTP-bound state that
growth factor-1; collagen 1 can activate downstream effector proteins (Olson, 1996;
Geyer and Wittinghofer, 1997). To be fully effective in
producing an invasive phenotype, Rho proteins need to
complete a full GTPase cycle and continue to cycle,
Introduction transitioning between inactive and active states (Hall,
1990; Moorman et al., 1999; Sander et al., 1999; Danen
This year alone, an estimated 29 900 men will die of et al., 2000). John Collard and his group elegantly
prostate cancer (CaP) in the United States, and the vast demonstrated a reciprocal relationship of activation
between RhoA GTPase and Rac1 (Sander et al., 1999;
Zondag et al., 2000). Therefore, although one isoform of
Correspondence: Assistant Professor KL van Golen, Department of Rho may predominate in a cancer cell, activation of other
Internal Medicine, The University of Michigan Cancer Center, 200 GTPases must occur during motility, migration and
Zina Pitcher Place, R-4048A Kresge II, Ann Arbor, MI 48109-0548.
E-mail: Kgolen@umich.edu
invasion, to effectively reorganize the cytoskeleton form-
Received 8 August 2005; revised 13 October 2005; accepted 13 October ing stress fibers, lamellipodia and filopodia (Nobes and
2005; published online 28 November 2005 Hall, 1995; Hall, 1998; Price and Collard, 2001).
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2286
RhoC GTPase is a Rho protein implicated in the
progression and metastasis of several cancers, including
breast, ovarian, pancreas, gastric cancer, hepatocellular
carcinoma, bladder, colorectal and cutaneous melano-
ma (Moscow et al., 1994; del Peso et al., 1997; Suwa
et al., 1998; Fritz et al., 1999; van Golen et al., 1999,
2000; Imamura et al., 1999; Clark et al., 2000; Sahai and
Marshall 2002; Carr et al., 2003; Horiuchi et al., 2003;
Kamai et al., 2003; Marionnet et al., 2003; Shinto et al.,
2003; Wang et al., 2003; Kondo et al., 2004). Our
laboratory has demonstrated that ectopic expression of
wild-type RhoC in immortalized human mammary
epithelial cells leads to a metastatic phenotype (van
Golen et al., 2000a, b). We have also demonstrated that
RhoC can be used as a prognostic marker to identify
small breast tumors (o1 cm) with a high likelihood to Figure 1 Expression of RhoA and RhoC GTPases in LNCaP,
metastasize (Kleer et al., 2002). Recently our attention DU145 and PC-3 CaP cell lines. Panel a shows the semiquantitative
RT-PCR analysis of 5 mg total RNA using primers specific for
has turned to the potential role of RhoC in CaP RhoA, RhoC and GAPDH (as a loading control). No noticeable
metastasis, particularly metastasis to bone. differences were seen in RhoA expression amongst the three cell
In several aggressive cancers, including CaP, activa- lines tested, whereas only the LNCaP and PC-3 cells expressed
tion of the Rho GTPases, including RhoC, occurs RhoC mRNA. Panel b shows the Western blot and GTPase
activation for the RhoA and RhoC proteins. A GST-fusion protein
through protein tyrosine kinase growth factor receptor of the Rho binding domain of rhotekin was used to selectively pull
stimulation (van Golen, 2003). Motility of many cancer out active RhoA or RhoC. Actin was used as a loading control of
cell types is stimulated by growth factors such as insulin- the immunoblot analysis. No differences in RhoA protein
like growth factor I (IGF-I) (Sahai and Marshall, 2002). expression or activation were observed for the three lines. Only
Increased levels of the type I IGF receptor (IGF-IR) are the PC-3 cell line expressed active GTP-bound RhoC protein.
Actin was used as a loading control.
found in the majority of primary and metastatic CaP
patient tumors (Hellawell et al., 2002). Functional IGF-
IR is expressed on PC-3 cells (Iwamura et al., 1993; (van Golen et al., 1999, 2000a, b, 2002). Increasing
Kimura et al., 1996), and bone is a rich source of IGF-I, evidence from several laboratories suggests that RhoC
which is thought to act as a chemoattractant in CaP expression is associated with particularly aggressive,
bone metastasis (Cooper and Pienta, 2000). CaP cells metastatic cancers; however, this has never been
further upregulate IGF-IR expression upon localization reported for CaP (Moscow et al., 1994; del Peso et al.,
to bone (Rubin et al., 2004). Although the link between 1997; Fritz et al., 1999; Imamura et al., 1999; Clark
the IGF system and CaP progression is well established, et al., 2000; Sahai and Marshall, 2002; Carr et al., 2003;
recent new drug developments have led to renewed Horiuchi et al., 2003; Kamai et al., 2003; Marionnet
pharmaceutical interest in IGF signaling pathways as et al., 2003; Shinto et al., 2003; Wang et al., 2003;
therapeutic targets (Zhang and Yee, 2004; Garber, 2005; Kondo et al., 2004). We hypothesized that RhoC may be
Mitsiades and Mitsiades, 2005). involved in CaP metastasis. To answer this question, we
In the present study, we demonstrate that RhoC is screened the LNCaP, DU145 and PC-3 cell lines for
highly expressed and active in the human PC-3 cell line, RhoC expression. RhoA GTPase, a close relative of
which is derived from a bone metastasis. Furthermore, RhoC, has been implicated in PC-3 cell invasion;
RhoC is responsible for IGF-I-mediated invasion of PC- therefore, we also examined RhoA expression. RhoA
3 cells but not for IGF-I-stimulated motility. Inhibition GTPase is highly expressed at the mRNA (Figure 1a,
of RhoC activity leads to increased Rac1 and Cdc42 left panel) and protein (Figure 1b, left panel) level in all
expression and activation. This in turn leads to an three CaP cell lines tested. However, RhoC message was
increase in linear motility, but decreased directed detected only in the LNCaP and PC-3 cell lines
migration and invasion. Finally, inhibition of RhoC (Figure 1a, right panel). Active, GTP-bound RhoA
activity results in a change in the expression and cellular was detected in all three prostate cancer cell lines. In
distribution of focal adhesion-associated proteins. These contrast, only the PC-3 cell line, which was originally
data provide insight into the potential mechanisms of derived from a skeletal metastasis, expressed active
CaP cell invasion, a key component in the metastatic RhoC protein (Figure 1b, right panel). Although the
cascade. LNCaP cell line has detectable levels of RhoC message,
it does not express appreciable levels protein, suggesting
that RhoC expression and activation may be associated
Results with the PC-3 metastatic phenotype.

Rho GTPase expression in CaP cells RhoC GTPase activity affects PC-3 cell motility
We previously demonstrated that RhoC GTPase is a and invasion
putative mammary oncogene responsible for the motile To determine whether expression of RhoC facilitates
and invasive capabilities of aggressive breast cancer cells cellular motility stimulated by elements found in the
Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2287
bone environment, we compared cell motility using a
colloidal blue-fluorescent bead random motility assay
and measured the area of phagokinetic tracks produced
by the cells (Figure 2a). The conditions of this assay
were adjusted to test motility in response to insulin-like
growth factor-I (IGF-I) stimulation on a collagen type I
(col I)-coated surface. Both IGF-I and collagen I are
present in the bone microenvironment and are impli-
cated in facilitating CaP dissemination, adherence and
survival (Kiefer and Farach-Carson, 2001; Rubin et al.,
2004; Chung et al., 2005; Yin et al., 2005). Each of the
three CaP cell lines tested have functional IGF-IR
(Kimura et al., 1996). At 16 h after stimulation with
5 nM IGF-I, the PC-3 cells produced unique circular
phagokinetic tracks many times the diameter of their cell
bodies (Figure 2a). Interestingly, when PC-3 cells were
pretreated with C3 exotransferase, a potent inhibitor of
Rho GTPases, and then stimulated with IGF-I, PC-3
cells no longer displayed a circular motility but became
extremely linearly mobile (Figure 2a). Quantitation of
the phagokinetic tracks produced by PC-3 cells demon-
strated a significant increase in area in cells treated with
C3 exotransferase prior to IGF-I stimulation
(Figure 2a). This observation is contrary to what we
have observed in other cell systems such as breast (van
Golen et al., 2000a, b). LNCaP cells are nonmotile, and
DU145 cells demonstrate moderate linear motility that
is inhibited by C3 exotransferase treatment. Since C3
exotransferase has specific affinity for the Rho proteins
(RhoC>RhoA>>RhoB) and very little affinity for
Rac1 or Cdc42, these data suggest that RhoC activity
may negatively modulate PC-3 cell random linear
motility (Aktories and Hall, 1989; Aktories et al.,
1989; Aktories, 1997). In contrast, since RhoC is not
expressed in the DU145 cells, inhibition of RhoA by C3
exotransferase treatment retards DU145 random linear
motility.
Next, we determined whether C3 treatment also
affected migration in response to a chemoattractant Figure 2 Effect of Rho GTPase activation on PC-3 cell motility,
(also termed directed motility). In this assay a Transwell migration and invasion. Panel a shows a comparison of the motile
chamber was used to determine the migratory capabil- capabilities of the CaP cells using a blue-fluorescent bead random
motility assay after stimulation with IGF-I on a collagen I-coated
ities of PC-3 cells through a filter with 8 mm pores using surface. In this assay, a colloid of blue-fluorescent polymer beads
5 nM IGF-I as a chemoattractant (Figure 2b). We found are overlaid onto 96-well plates coated with collagen I. The cells are
that the PC-3 cells were highly migratory towards IGF- allowed to attach for 1 h in serum-free medium and then stimulated
I. However, in contrast to the random motility assay with 5 nM IGF-I. After growth factor stimulation, motile cells
phagocytize the colloid and the area of the resulting phagokinetic
described above, the ability of cells to migrate in tracks are measured 24 h later. To determine the role of Rho
response to IGF-I is significantly inhibited by C3 proteins in conferring a motile phenotype, cells were treated
exotransferase treatment. overnight with 10 mg C3 exotransferase, a potent inhibitor of Rho
Similar results were observed when the cells were proteins. Rho inhibition significantly increased the PC-3 cells linear
tested for haptotactic migration through an 8 mm filter random motility (*P ¼ 0.001). Panel b demonstrates the results of
directed migration, haptotaxis (with a gradient of 1 nM in the top
with a gradient of 1 nM IGF-I in the upper chamber to chamber to 5 nM IGF-I in the lower chamber) or invasion assay
5 nM IGF-I in the lower chamber (Figure 2b). Untreated through a Matrigel or collagen I-coated filter. In each assay the
PC-3 cells move readily toward the higher IGF-I cells were placed in the top of a Transwell filter and migrated
concentration; however, C3 treatment abrogates this towards a chemoattractant. In contrast to the random motility
assay, C3 treatment (gray bars) significantly decreased PC-3
affect. movement as compared with the untreated cells (black bars,
Finally, the cells were tested for their ability to invade *Po0.05).
through a Matrigel- or collagen I-coated filter in
response to 5 nM IGF-I as a chemoattractant. In these
systems, the PC-3 cells are highly invasive (Figure 2b). motility and invasion in PC-3 cells are particularly
C3 exotransferase treatment significantly inhibits CaP regulated by different GTPases. Specifically, RhoC
cell invasion. Taken together, these data suggest that activity is required for directional motility toward a
Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2288
chemoattractant (i.e. migration, haptotaxis and inva- reduced by B65% in the RhoC/dnRhoC cells and
sion), but limits linear random motility. B60% in C3 exotransferase-treated PC-3 cells com-
Since C3 exotransferase also inhibits RhoA GTPase, pared with the parental and vector control and cell lines
stable dominant-negative RhoC-expressing PC-3 (PC-3/ (Figure 3a). However, there are no significant changes in
dnRhoC) cells were produced. Dominant-negative total RhoC levels. These data, taken from four separate
GTPases act by entering into a nonproductive interac- analyses, suggest that RhoC and RhoA, although highly
tion with Rho guanine exchange factors (GEFs), the homologous, are activated by distinct RhoGEFs in the
proteins responsible for catalyzing the exchange of GDP PC-3 cells. Therefore, expression of a dominant-negative
for GTP on the GTPase. Given that RhoC and RhoA RhoC specifically decreases activity of RhoC without
are 91% homologous on the protein level, we speculated affecting RhoA activation. No differences in monolayer
that they may share a common activating GEF, and cell growth were observed between the parental, vector
therefore expression of a dominant-negative RhoC and dnRhoC-expressing PC-3 cells (Figure 3c).
would affect RhoA activity. No significant differences To determine whether expression of dominant-nega-
in active or total RhoA levels were detected in the PC-3/ tive RhoC recapitulates the effect of C3 exotransferase
dnRhoC cells compared with the parental PC-3 or PC-3/ treatment on PC-3 cells, we performed a random
pcDNA3.1 vector control cells (Figure 3b). In contrast motility assay as before. Parental PC-3, PC-3/
to the observed RhoA activity, GTP-bound RhoC is pcDNA3.1 vector control and PC-3/dnRhoC stable

Figure 3 Expression of a dominant-negative RhoC in PC-3 cells. RhoC containing a T19N mutation was introduced into PC-3 and
stable clones selected. The data are from a representative polyclonal population. Panel a shows the result of triplicate analysis of RhoC
protein expression (dark gray bars) and activation (light gray bars). RhoC activation was compared with a vector control transfectant
left either untreated or treated with C3 exotransferase. C3 treatment also leads to decreased RhoC activation comparable to the
dnRhoC expressing line. Panel b shows the result of triplicate analysis of a GTPase activation assay and Western blot analysis of
RhoA. This was performed to demonstrate that the dominant-negative version of RhoC did not interfere with RhoA activation. No
significant difference in RhoA expression (dark gray bars) or activation (light gray bars) was observed in the parental, vector control or
dominant RhoC expressing PC-3 cells. Panel c is an MTT assay comparing the monolayer growth rates of parental (’), vector control
(m) and dnRhoC () PC-3 cells.

Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2289
morphology. Figure 5A shows phase contrast, laser
confocal and scanning electron micrographs of parental
PC-3 (panels a, d and g), PC-3/pcDNA3.1 (panels b, e
and h) and PC-3/dnRhoC (panels c, f and i) cells. Both
control cell lines are relatively flat with very broad
lamellipodia nearly encompassing the circumference of
the cell. In contrast, the PC-3/dnRhoC cells are
elongated with smaller lamellipodia located on the
leading edge of the cell.
Immunofluorescent staining for actin and the cyto-
skeletal protein vinculin demonstrates profound differ-
ences between PC-3 and PC-3/dnRhoC cells when
grown on collagen I and stimulated with IGF-I
(Figure 5A, panels j–o). In PC-3 cells the cytoskeleton
appears diffuse and appears branched leading to broad
deformation of the cells’ lamellipodia. Vinculin that
does not appear to be associated with actin bundle ends
is localized in the center of the parental PC-3 cell. In
contrast, colocalized actin and vinculin in PC-3/
dnRhoC cells are isolated to the tips of the protrusions
at the leading and trailing ends of the cell. Actin is
highly organized along the periphery of the cells in an
arrangement reminiscent of concave actin bundles and
the bulk of vinculin is localized throughout the main
body of PC-3/dnRhoC cells.
The change in PC-3 morphology is reminiscent of the
Figure 4 RhoC GTPase inhibits PC-3 linear random motility but epithelial to mesenchymal transition (EMT) described
promotes directed migration and invasion. Panel a shows the for other tumor cell types (Chang et al., 2003; Bockhorn
quantitation of the area of phagokinetic tracks produced in a et al., 2004). Figure 5B shows the Western blot analysis
random motility assay comparing wild type, vector control and for molecular markers associated with EMT: ZO-1, E-
dnRhoC expressing PC-3 cells. The motility assay was performed
using collagen I as a base substrate and 5 nM IGF-I to stimulate and N-cadherin. As typically described for other cell
motility for 16 h. Similar to what was observed when the PC-3 cells types undergoing EMT, a switch in expression of E- to
were treated with the Rho inhibitor C3 exotransfersase, expression N-cadherin and a decrease in ZO-1 are observed in the
of dnRhoC led to increased linear motility. Panel b shows a PC-3/dnRhoC cells (Figure 5B), suggesting that RhoC
comparison of parental (light gray bars), vector control (dark gray
bars) and dominant-negative RhoC (black bars) PC-3 cells in a
may act as a molecular switch for an EMT-like change
directed motility, Matrigel and collagen I invasion assays. in PC-3 cells.
Compared with the control cell lines, the PC-3/dnRhoC cells
demonstrated a significant decrease in directed migration and RhoC GTPase activation influences expression
invasion (*Po0.05). and activation of Rac1 and Cdc42
A reciprocal relationship between RhoA and Rac1/
transfectants were plated onto blue fluorescent beads Cdc42 activity has been described for motile NIH3T3
overlaid onto a collagen I-coated surface, stimulated cells (Sander et al., 1999; Zondag et al., 2000). To
with 5 nM IGF-I and analysed 24 h later. Similar to C3 determine whether other members of the Rho GTPase
exotransferase-treated PC-3 cells, PC-3/dnRhoC trans- family are affected by changes in RhoC activity, we
fectants displayed increased linear motility (Figure 4a). performed Western blot analysis and GTPase activity
Quantitation of the areas of phagokinetic tracks from assays for Rac1 and Cdc42. Both Rac1 and Cdc42 are
three separate assays demonstrates that the PC-3/ implicated in cellular motility, specifically through the
dnRhoC cells have a threefold increase in the area of formation of lamellipodia and filopodia, respectively,
their phagokinetic tracks as compared with the control and are therefore prime candidates for mediating the
PC-3 cells. observed changes in the morphologic characteristics of
The effect of dnRhoC on migration and invasion was PC-3 cells (Ridley and Hall, 1992a, b; Ridley et al., 1992;
evaluated as described previously. Migration and inva- Nobes and Hall, 1995). The process of linear motility
sion in PC-3/dnRhoC cells are approximately 2- and requires coordinated changes in Rac1 and Cdc42
2.5-fold less, respectively, than that of the control PC-3 activity. Therefore, we determined the expression and
cells (Figure 4b). This suggests that RhoC activation activation profile of these two GTPases over an
promotes PC-3 cell-directed migration and invasion, but extended time course using conditions similar to the
limits linear random motility. random motility assay. Serum-starved PC-3 and PC-3/
dnRhoC cells were plated onto collagen I, stimulated
RhoC GTPase activity affects PC-3 cell morphology with IGF-I and analysed 0, 5, 15, 60, 120 and 180 min
One observable consequence of inhibiting RhoC after stimulation. Quantitative changes in Rac1 and
GTPase activity is a profound change in PC-3 cell Cdc42 expression and activation are observed in PC-3/
Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2290

Figure 5 Inhibition of RhoC GTPase alters the morphology of PC-3 cells. Panel A shows phase (a–c), laser confocal (d–f) and
scanning electron (g–i) micrographs of PC-3 (a, d and g), PC-3/pcDNA3.1 (b, e and h) and PC-3/dnRhoC (c, f and i). Panels j–o show
immunofluorescent (IF) staining of vinculin (j and m) and rhodamine-phallodin for actin stress fibers (k and n) with merge (l and o).
Cells were grown on collagen I, stimulated with 5 nM IGF-I and imaged as described in the Materials and methods section. Panel B
shows the Western blot analysis of 30 mg total protein from PC-3, vector control and PC-3/dnRhoC cells for expression of EMT-
associated proteins ZO-1, E- and N-cadherin. Densitometry measurements are the averages of three independent experiments. The
value for the PC-3 parental cells was set at 1 and all other values were measured against it.

dnRhoC cells compared with parental PC-3 cells GDP for GTP (Kwon et al., 2000). Thus, decreased
(Figure 6A). In the PC-3 cell line, a low, persistent level phosphorylation of Rac1 may contribute to an increase
of Rac1 and Cdc42 activation is observed throughout in Rac1 and Cdc42 activity.
the time course (Figure 6A). In contrast to parental
cells, PC-3/dnRhoC cells display a marked increase in RhoC GTPase activity modulates focal adhesion protein
expression and activation of both Rac1 and Cdc42 at activation and localization
60 min after IGF-I stimulation, which persists up to The phenotypic changes observed in PC-3/dnRhoC cells
180 min (Figure 6A). Of particular note is a striking are identical to those previously described for cells
increase in Cdc42 expression from 0 to 60 min after transfected with a paxillin deletion mutant (West et al.,
stimulation. 2004). Specifically, CHO.K1 fibroblasts transfected with
Interestingly, levels of phosphorylated Rac1 and paxillin lacking the LD4 domain, which binds paxillin
Cdc42 are reduced in PC-3/dnRhoC- and C3-treated kinase linker (PKL), demonstrate increased linear
PC-3 cells grown on collagen I and stimulated with IGF- motility, decreased invasion and increased, persistent
I for 60 min (Figure 6B). Rac1 and Cdc42 are Rac1 activation (West et al., 2004). With this in mind,
phosphorylated on serine 71 and in vitro assays suggest we determined the effect of RhoC activation on focal
that phosphorylation of Rac1 inhibits the exchange of adhesion proteins. Total and phosphorylated focal
Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2291

Figure 6 Expression and activation of Rac1 and Cdc42 in PC-3 cells are increased when RhoC GTPase is not active. Panel A, to
determine the duration of Rac1 and Cdc42 expression and activation, serum-starved PC-3 and PC-3/dnRhoC cells were plated on
collagen I, stimulated with IGF-I and harvested at 0, 5, 15, 60, 120 and 180 min. Immunoblot analysis is shown in (a). An aliquot of
30 mg total protein was taken to determine total Rac1 or Cdc42 levels; the remainder of the protein lysates was used to determine levels
of active GTPase. Densitometry was performed on each of the samples and a ratio of GTP versus total protein was determined and is
shown for the duration of the time course (b). PC-3 cells (’) are represented by the dashed line and the dnRhoC cells (m) by the solid
line. A separate panel (c) shows the changes for Rac1 observed in the first 15 min of the experiment. The t ¼ 0 value of parental PC-3
cells was set to 1 and all other values were measured against it. The values shown are averages from at least three separate experiments.
Panel B shows the immunoblot analysis for total and serine 71 phosphorylated Rac1 and Cdc42. PC-3, PC-3/pcDNA3.1 vector and
PC-3 dnRhoC cells were grown on collagen I, stimulated with 5 nM IGF-I and protein harvested 60 min later. A 50 mg aliquot of each
protein was analysed by immunoblotting with monoclonal antibodies specific for Rac1, Cdc42 and phospho-Rac1/Cdc42. GAPDH
was included as a loading control.

adhesion kinase (FAK) and phosphorylated paxillin adhesion proteins. Furthermore, taken with the altera-
levels were decreased in the PC-3/dnRhoC cells, whereas tions seen in the distribution of the actin cytoskeleton,
total paxillin and PKL protein levels essentially these data may indicate turnover of focal adhesions to
remained unchanged (Figure 7A). More interestingly, focal complexes (Kaverina et al., 2002).
immunoprecipitation experiments demonstrate that the
interaction of PKL with paxillin is decreased in PC-3/
dnRhoC cells (Figure 7B). In contrast, FAK and PKL Discussion
or FAK and paxillin coimmunopreciptated, indicating
that their interaction was not altered (Figure 7A). The ability of CaP cells to invade and metastasize,
Next, colocalization of paxillin with FAK (panel a), particularly to bone, has been an area of intense
PKL with FAK (panel b) and paxillin with PKL (panel research. Bone contains high levels of IGF ligands,
c) was determined (Figure 7C). All of the focal adhesion which are intimately involved in normal bone turnover
proteins colocalize within the cell, but the distribution of and are implicated in metastasis (Chung et al., 2005).
these proteins is altered in PC-3/dnRhoC cells compared IGF-I not only acts as a mitogenic factor for cells, but
with the parental PC-3 line (Figure 7C). Fluorescent also stimulates cellular motility (reviewed in (Cooper
localization demonstrated that FAK and PKL coloca- and Pienta, 2000; van Golen, 2003)). IGF-I potentially
lize in both PC-3 and PC-3/dnRhoC cells. Paxillin also regulates tumor cell motility through the activity of the
colocalized with both FAK and PKL in these cells. Rho GTPases (van Golen, 2003). In particular, IGF-I
However, in the PC-3/dnRhoC cells, PKL is found stimulation of Rac1 in neuroblastoma cells and RhoA
independent of paxillin in the cytoplasm of the cells and RhoC in breast cancer cells is well studied (Jackson
(white arrows; Figure 7C vi). These data suggest that the et al., 2001; Meyer et al., 2001; Tanno et al., 2001; Lopez
changes in cell motility, migration and invasion due to and Hanahan, 2002). Several laboratories, including our
inhibition of RhoC activity may be because of altera- own in the present study, have demonstrated that IGF-I
tions in the localization, activity and interaction of focal acts as a chemoattractant for CaP cells Cooper and
Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2292

Figure 7 Inhibition of RhoC leads to changes in expression and distribution of focal adhesion proteins. Panel A shows the Western
blot analysis for phosphorylated and total focal adhesion kinase (FAK), paxillin and paxillin kinase linker (PKL). Aliquots of 50 mg
total protein were analysed for phosphorylated and total protein levels. Relative ratios of phosphorylated to total protein levels are
shown for FAK and paxillin. Relative levels of total PKL are also shown. Densitometry measurements are the averages of three
independent experiments. The value for the PC-3 parental cells was set at 1 and all other values were measured against it. Panel B
interaction of focal adhesion proteins was determined by immunoprecipitation of 500 mg total protein that was pre-cleared with normal
IgG followed by immunoblotting. Normal IgG negative controls did not produce bands when immunoblotted with antibodies specific
for FAK, paxillin or PKL. Measurement of the relative levels of PKL detected in the paxillin immunopreciptiations is shown. The
value for the parental PC-3 cells was set at 1 and all other values were measured against it. Measurements are the average of three
independent experiments. Panel C shows the immunfluoresence for (a) paxillin (i and iv) and FAK (ii and v) with merge (iii and vi), (b)
PKL (i and iv) and FAK (ii and v) with merge (iii and vi), (c) paxillin (i and iv) and PKL (ii and v) with merge (iii and vi). The arrows
indicate that PKL is not associated with paxillin.

Pienta (2000). Furthermore, collagen I, an integral GTPases are temporally and spatially activated, allow-
component of mineralized bone matrix, affects CaP cell ing tumor cells to successfully invade, survive and grow
migration and invasion through integrin activation at distant sites (Keely et al., 1998; Jaffe and Hall, 2002).
(Keely et al., 1998; Jaffe and Hall, 2002). Through Thus, growth factor and integrin signaling provide two
interplay of growth factor and integrin signaling, Rho potential stimuli for Rho activation (Cooper and Pienta,
Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2293
2000). Therefore, we chose to investigate the role of 1, E- and N-cadherin. These data suggest that in PC-3
RhoC GTPase in IGF-I and/or collagen I-stimulated cells, RhoC may act as a determinant switch for EMT.
motility, migration, invasion and morphological altera- Collard’s group was the first to demonstrate that
tions in PC-3 cells. RhoA GTPase and Rac1 are in a reciprocal relationship
Clearly, the Rho GTPases play a central role in tumor of activation during cellular motility and invasion
cell invasion and metastasis (Sahai and Marshall 2002). (Sander et al., 1999; Zondag et al., 2000). In the current
To date, few studies have attempted to define the Rho study, we demonstrate that when RhoC is inhibited in
GTPase profile of metastatic CaP cells. A recent study PC-3 cells there is a marked upregulation of Rac1 and
by Hodge et al. (2003) suggests that increased RhoA Cdc42 expression and activation. In addition, in PC-3/
expression and activity are responsible for NF-kB- dnRhoC cells, we observe changes in expression,
mediated PC-3 cell invasion. The invasive capabilities activation and distribution of focal adhesion proteins.
of highly metastatic PC-3 variants, but not of low These changes likely represent a switch from focal
metastatic variants, were increased by introducing a adhesions, which are associated with stable adhesion, to
constitutively active form of RhoA into the cells (Hodge focal complexes, present during active cell motility, that
et al., 2003). Our data suggest that RhoA is a are typically formed through Rac1 and Cdc42 activity
component in the motile and invasive phenotype of (Kaverina et al., 2002). The changes in morphology,
PC-3 cells; however, we were unable to find a difference Rac1 GTPase activation, random motility and invasion
in RhoA expression in our PC-3/dnRhoC transfectants are similar to those observed in paxillin deletion mutants
that could account for the in vitro differences in invasion (West et al., 2004). Deletion of the LD4 domain of
detected, compared with parental PC-3 cells. Other paxillin results in redistribution of PKL to the cyto-
groups have demonstrated that RhoA and Cdc42 plasm of CHO.K1 fibroblasts. As a result, cellular
activation is involved in cytoskeletal reorganization of morphology changes, cellular protrusiveness increases,
CaP cells. Activation of the G-protein-coupled receptors random motility increases and invasion decreases,
called protease-activated receptors (PARs) results in suggesting an increase in focal complex formation.
activation of RhoA and other members of the Rho These cells also have increased Rac1 expression and
GTPase family (Greenberg et al., 2003). Stimulation of prolonged Rac1 activity. Therefore, disrupted interac-
LNCaP cells with PAR-activating peptides, trypsin or tion between paxillin and PKL promotes prolonged Rac
thrombin leads to increased stress fiber formation and activity and extended linear motility. In our study, we
filopodia formation (Greenberg et al., 2003). Similar observe a decreased interaction between paxillin and
cytoskeletal reorganization and activation of RhoA and PKL in the PC-3/dnRhoC cells. We have measured a
Cdc42 is observed when PC-3U cells (a subline of PC-3) slight decrease in PKL expression in the PC-3/dnRhoC
are stimulated with transforming growth factor-b cells, which could account for the decreased paxillin/
(Edlund et al., 2002). Although these studies demon- PKL interaction. However, we also observed a large
strate a role for these GTPases in CaP cell cytoskeletal amount of PKL in the cytoplasm of the cells suggesting
reorganization, they have not fully made the connection that PKL is localized away from paxillin.
with motility, invasion or metastasis. Further, it is Paxillin is present in both focal complexes and focal
becoming increasingly evident that the vast majority of adhesions and is one of the earliest proteins recruited to
human tumors overexpress RhoA, but overexpression of these structures by either integrin engagement or growth
RhoC is limited to particularly aggressive and metastatic factor stimulation (Turner, 2000; Zaidel-Bar et al.,
forms of cancer (Moscow et al., 1994; del Peso et al., 2003). Rac-1 promotes focal complex formation,
1997; Fritz et al., 1999; Imamura et al., 1999; Clark whereas Rho kinase inhibition induces focal adhesion
et al., 2000; Sahai and Marshall, 2002; Carr et al., 2003; disassembly, also resulting in increased focal complex
Horiuchi et al., 2003; Kamai et al., 2003; Marionnet formation (Zaidel-Bar et al., 2003). Our observations
et al., 2003; Shinto et al., 2003; Wang et al., 2003; suggest that high levels of active RhoC in cells may
Kondo et al., 2004) The PC-3 cells, derived from a bone induce paxillin/PKL interaction, thus reducing the
metastasis, were the only CaP cell line to express active prolonged Rac activation associated with focal complex
RhoC protein. formation, leading to a reduction in cellular protrusions
Interestingly, we demonstrate that when PC-3 cells are and associated random motility. Since focal complexes
tested for random motility using collagen I as a are rather short-lived structures, active RhoC inhibition
substrate and IGF-I as a stimulant; the phagokinetic of focal complex formation could in turn promote more
tracks produced suggest that the cells are anchored at directed migration through stable focal adhesion for-
one end allowing them to only move in a circular mation and/or extended interactions with ECM.
pattern. Yet, they are highly motile in directed migra- In summary, our data suggest an inverse relationship
tion, haptotaxis and invasion assays. In contrast, when of active RhoC with Rac1 and Cdc42 in PC-3 cells.
RhoC is inhibited, the cells become highly randomly RhoC activity promotes IGF-I-stimulated migration
motile but do not invade. Changes in PC-3 cell and invasion while limiting linear motility. In contrast,
morphology, reminiscent of EMT described for other when RhoC activation is inhibited, Rac1 and Cdc42
tumor cell types, is observed when RhoC is inhibited activity increase, promoting linear motility. Concur-
(Chang et al., 2003; Bockhorn et al., 2004). Accom- rently, there is a switch reminiscent of an epithelial to
panying the changes in morphology, we distinguish mesenchymal morphology. Expression, activation and
changes in protein expression of the EMT markers ZO- localization of focal adhesion proteins are also affected.
Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2294
Thus, RhoC appears to act as a critical and dynamic MTT assay
switch controlling PC-3 cell linear and directed move- Monolayer growth was measured using MTT (Sigma Chemical
ment. Our laboratory is currently exploring the con- Co.) as previously described (van Golen et al., 2000). Briefly,
vergence of growth factor and integrin downstream cells were plated at densities ranging from 100 to 10 000 cells in
200 ml growth medium in six replicate wells of a 96-well plate
signaling and its effect on Rho GTPase activation.
and incubated in a 371C incubator under 10% CO2. Cell
growth was measured at 0, 1, 2, 3 and 5 days after plating. For
the 0 daytime point plates were centrifuged at 1000 r.p.m. in
Materials and methods Sorvall refrigerated tissue culture centrifuge. A 40 ml aliquot of
5 mg/ml MTT was added to the cultures for 1 h at 371C. The
Cell culture medium was aspirated and 100 ml DMSO (Sigma Chemical
All CaP cell lines were obtained from American Type Culture Co.) was added to each well to solubilize the formazon dye.
Collection (Manassas, VA, USA). The LNCaP cells were each Absorbance was read on a Dynatech microplate reader at
maintained in 90% RPMI-1640 with 2 mM L-glutamine, 1.5 g/l 590 nm.
sodium bicarbonate, 4.5 g/l glucose, 10 mM HEPES, 1 mM
sodium pyruvate and 10% fetal bovine serum (FBS, Gibco, Western blot analysis
Carlsbad, CA, USA). The DU145 cells were propagated in Protein was harvested from cells using RIPA buffer as
Eagles minimal essential medium with Earles BSS, 2 mM L- previously described (van Golen et al., 2000a, b). Lysates were
glutamine, 1.5 g/l sodium bicarbonate, 0.1 mM nonessential separated by SDS–PAGE on a 12% gel, transferred to
amino acids, 1.0 mM sodium pyruvate and 10% FBS (Gibco). nitrocellulose, blocked and probed with a polyclonal antibody
The PC-3 cells were maintained in Ham’s F-12 medium with for RhoC (Kleer et al., 2002) or monoclonal antibodies for
1.5 g/l sodium pyruvate, 2 mM L-glutamine and 10% FBS RhoA (Cytoskeleton, Denver, CO, USA), Rac1, Cdc42 and
(Gibco). PC-3 transfectants were maintained in the same phospho-Rac1/Cdc42 (Santa Cruz Biotechnology, Santa Cruz,
medium as the parental PC-3 cells with the addition of 250 mg/ CA, USA), E-cadherin (a gift from Dr Peggy Wheelock), N-
ml Neomycin (Gibco). All cell lines were maintained at 371C in cadherin, PKL (BD Transduction Laboratories, San Diego,
a 10% CO2 incubator. C3 exotransferase was introduced into CA, USA), FAK (Santa Cruz and Biolegend, San Diego, CA,
cells as previously described using a lipid transfer-mediated USA), phospho-FAK, paxillin and phospho-paxillin (Bio-
method and treated for 16 h before analysis (van Golen et al., legend). After incubation with a goat anti-rabbit-HRP or goat
2000). anti-mouse-HRP antibody (Santa Cruz), immunoblots were
developed with ECL, exposed to Hyperfilm (Amersham,
Plasmids and transfections Piscataway, NJ, USA) and images recorded on an Alpha
Dominant-negative RhoC (dnRhoC) in pcDNA3.1 was Image 90 documentation system (Alpha Innotech, San
obtained from Gutherie cDNA Resource center (Sayre, PA, Leandro, CA, USA). Actin or GAPDH were used as loading
USA). Empty pcDNA3.1 was used as a vector control controls (Sigma, St Louis, MO, USA). Whenever possible
transfectant (Invitrogen, Carlsbad, CA, USA). The dnRhoC blots were first probed with the phospho-specific antibody,
or empty vector was introduced into PC-3 cells using FuGene6 stripped and reprobed with an antibody to total protein.
transfection reagent (Roche, Indianapolis, IN, USA) and Densitometry was performed using Scion Image version 1.62.
selecting cells in 250 mg/ml Neomycin. Expression of the Values for the PC-3 parental line were set at 1 and all other
transgene was confirmed by RT-PCR. values were measured against it.

Anchorage-independent growth assays GTPase activation assays


For anchorage-independent growth assays, a 2% stock of Rho activation assay reagents were supplied to us by John
sterile, low-melt agarose was diluted 1:1 with 2  MEM. Collard, PhD, at The Netherlands Cancer Institute (Evers
Further dilution to 0.6% agarose was made using 10% FBS- et al., 2000; Diekmann and Hall, 1995) and used to determine
containing growth medium and 1 ml added to each well of a activation of RhoC in PC cells. Cells, 5–10  106 cells/assay,
six-well plate as a base layer. The cell layer was prepared by were grown in fresh serum-containing medium for 16 h and
diluting agarose to 0.3, 0.6 or 0.9% with 103 cells in 2.5% FBS- lysed with ice-cold GST-FISH buffer (10% Glycerol, 50 mM
containing growth medium/1.5 ml/well. A 1 ml layer of Tris pH 7.4, 100 mM NaCl, 1% NP-40 and 2 mM MgCl2).
medium was maintained on top of the agar to provide Protein lysates, 900 ml of 1 mg/ml, were precleared with
nutrients. Colonies X100 mm in diameter were counted after glutathione sepharose for 1 h at room temperature. The
2-week incubation at 371C in a 10% CO2 incubator. cleared lysates were incubated with GST-rhotekin-sepharose
conjugate at 41C for 30 min. The agarose conjugate was
RT-PCR collected by centrifugation, resuspended in Laemelli buffer,
Semi-quantitative RT-PCR for RhoC and RhoA expression separated by SDS–PAGE, transferred to nitrocellulose and
was performed as previously described using primers specific probed with a RhoC antibody developed by our laboratory
for RhoC, RhoA or as a control, GAPDH (van Golen et al., (Kleer et al., 2002). Densitometry was performed using an
2000a, b). Briefly, total RNA was harvested from cells using Alpha Image 950 imaging system.
Trizol reagent (Life Technologies, Gaithersburg, MD, USA)
and cDNA was made using the AMV-reverse transcriptase kit Motility, migration, haptotaxis and invasion assays
(Promega, Madison, WI, USA). RhoC and RhoA transcripts Random motility assays were also performed using blue
were PCR amplified from cDNA aliquots using a 1:1000 fluorescent bead motility HitKit (Cellomics, Pittsburg, PA,
dilution of Rho-specific primers mixed with GAPDH primers. USA), which is based on the colloidal gold random motility
PCR products were separated on a 1.2% TAE agarose gel and assay (Albrecht-Buehler, 1977; van Golen et al., 2000a, b).
visualized with ethidium bromide. The relative intensities of Cells, 200/well in SFM, were seeded onto a lawn of blue
the Rho and GAPDH bands were measured using an Alpha fluorescent beads in a BSA-coated 96-well plate (BD Bio-
Imager 2200 (Alpha Innotech, Co., San Leandro, CA, USA). sciences). Cells were stimulated 1 h later with 10% FBS,
Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2295
incubated 6 h at 371C and fixed. Cells were visualized by Jackson ImmunoResearch Labs, West Grove, PA, USA) in
fluorescent microscopy, digitally recorded and the areas of 1  PBS pH 7.4 with 0.3% Triton X-100 for 1 h at room
phagokenetic tracks measured using Scion Image. temperature. Immunfluorescence was performed using anti-
The directed migration assay was performed using an bodies specific for vinculin (monoclonal: Calbiochem, San
uncoated Transwell filter with 8 mm pores (Costar, Corning, Diego, CA, USA), FAK (monoclonal: Biolegend), paxillin
NY, USA). The lower chamber of the filter was filled with (monoclonal and polyclonal: Biolegend), PKL (monoclonal:
either serum-free medium or serum-free medium containing BD Transduction Laboratories) and rhodamine-phallodin
5 nm IGF-I. The Matrigel and collagen I invasion assays were (Invitrogen Molecular Probes). Double staining of cells was
performed as previously described using precoated Transwell performed using antibodies from different hosts. Primary
filters with 8 mm pores (Hendrix et al., 1987; van Golen et al., antibodies were diluted in 2%NGS in 1  PBS pH 7.4 with
2000a, b). Cells were harvested and resuspended in serum-free 0.3% Triton X-100 for 3 h at room temperature in a humidified
medium containing 0.1% BSA at a concentration of chamber. After rinsing with PBS, cells were incubated with a
3.75  105 cells/ml, and 0.5 ml was added to the top chambers. species-specific secondary antibody conjugated to a fluorescent
The haptotaxis assay was performed similar to the migration tag for 1.5 h at room temperature. Excess antibody was rinsed
assay, except that 1 nM IGF-I was added to the top chamber with PBS, and cells were treated with anti-Fade (Invitrogen
with the cells. The chambers were incubated for 24 h at 371C in Molecular Probes, Eugene, OR, USA), mounted onto Super-
a 10% CO2 incubator. The cell suspension was aspirated from Plus slides (Corning, NY, USA) with Gel Mount (Biomedia,
the top chamber of all three assays. Excess Matrigel or Foster City, CA, USA) and sealed with clear nail polish.
collagen was removed from the invasion assay filter using a Immunfluorescence was performed on a Zeiss Scanning Laser
cotton swab. The filters were then cut away from the Transwell Confocal Microscopy housed in the University of Michigan
assembly, fixed top side down with methanol to a glass MIL.
microscope slide, stained with H&E and the entire surface of
the filters counted. In the directed migration, haptotaxis and Statistical analysis
invasion assays, the number of cells that moved in the serum- All experiments were performed a minimum of three separate
free controls was considered as background and was sub- times and statistical analysis of the combined experiments was
tracted from the number of cells counted in the samples performed by the University of Michigan Biostatistics Core.
containing chemoattractant.

Scanning electron microscopy


Scanning electron microscopy (SEM) was performed as Abbreviations
previously described (van Golen et al., 2002). Briefly, cells
(12 000) grown on collagen I were fixed with buffered 2.5% CaP, prostate cancer; Rho, Ras homology; IGF-I, insulin-like
glutaraldehyde for 1 h, rinsed with phosphate-buffered saline growth factor I; col I, collagen type I; FAK, focal adhesion
(PBS) and postfixed for an additional hour with buffered kinase; PKL, paxillin kinase linker; EMT, epithelial to
osmium tetroxide. After dehydration in ascending strengths of mesenchymal transition; SEM, scanning electron microscopy.
ethanol, the cells were critical-point dried, mounted onto
standard SEM stubs and gold-sputter coated. They were Acknowledgements
viewed using an AMRAY 1000-B scanning electron micro-
scope housed at the University of Michigan Microscopy and This work was supported in part by the University of
Imaging Laboratories (MIL). Michigan Comprehensive Cancer Center support grant (5
P30 CA46592) and by a Career Development Award from the
Immunofluorescence Specialized Program of Research Excellence (P50 CA69568,
PC-3 and PC-3/dnRhoC cells were grown on collagen I-coated Kenneth Pienta Principal Investigator) at the University of
coverslips (BD Biosciences, San Diego, CA, USA) until 70% Michigan and the Department of Defense (W81XWH-04-1-
confluence, serum-starved for 16 h, stimulated with 5 nM IGF-I 0225) (to KLvG). We thank Chris Edwards, MS, for help with
for 60 min, rinsed three times with 1  PBS pH 7.4 and fixed the scanning electron micrographs, and Kate Groh, BS, for
with 4% paraformaldehyde in PBS for 20 min at room technical assistance, Drs Kenneth J Pienta, Carlton Cooper
temperature. After fixation, cells were rinsed three times with and Tim Ritty for insightful discussions, and Robyn Blanzy-
1  PBS and blocked with 5% normal goat serum (NGS, Hodges for manuscript preparation.

References

Abate-Shen C, Shen MM. (2000). Genes Dev 14: 2410–2434. Carr KM, Bittner M, Trent JM. (2003). Oncogene 22:
Aktories K, Hall A. (1989). Trends Pharmacol Sci 10: 3076–3080.
415–418. Chang F, Steelman LS, Shelton JG, Lee JT, Navolanic PM,
Aktories K, Braun U, Rosener S, Just I, Hall A. (1989). Blalock WL et al. (2003). Int J Oncol 22: 469–480.
Biochem Biophys Res Commun 158: 209–213. Chung LW, Baseman A, Assikis V, Zhau HE. (2005). J Urol
Aktories K. (1997). Trends Microbiol 5: 282–288. 173: 10–20.
Albrecht-Buehler G. (1977). Cell 11: 395–404. Clark EA, Golub TR, Lander ES, Hynes RO. (2000). Nature
Alcover J, Fernandez-Conde M, Carrere W, Serrallach M, 406: 532–535.
Carretero P. (1994). Actas Urol Esp 18(Suppl): 409–416. Cooper CR, Pienta KJ. (2000). Prostate Cancer Prostatic Dis
American Cancer Society Facts and Figures (2005). 3: 6–12.
Bockhorn M, Roberge S, Sousa C, Jain RK, Munn LL. (2004). Danen EH, Sonneveld P, Sonnenberg A, Yamada KM. (2000).
Cancer Res 64: 2469–2473. J Cell Biol 151: 1413–1422.
Cancer Management: A Multidisciplinary Approach (1999) del Peso L, Hernandez-Alcoceba R, Embade N, Carnero A,
3rd ed. Esteve P, Paje C et al. (1997). Oncogene 15: 3047–3057.

Oncogene
Requirement of RhoC GTPase for prostate cancer invasion
H Yao et al
2296
Diekmann D, Hall A. (1995). Meth Enzymol 256: 207–215. Nobes CD, Hall A. (1995). Cell 81: 53–62.
Edlund S, Landstrom M, Heldin CH, Aspenstrom P. (2002). Olson MF. (1996). J Mol Med 74: 563–571.
Mol Biol Cell 13: 902–914. Pienta KJ, Sandler H, Sanda MG. In: Pazdur R, Coja LR,
Evers EE, van der Kammen RA, ten Klooster JP, Collard JG. Hoskins WJ, Wagman LD (eds). Cancer management: a
(2000). Meth Enzymol 325: 403–415. multidisciplinary approach. 3rd edn. PRR Publishing: Mel-
Fritz G, Just I, Kaina B. (1999). Int J Cancer 81: 682–687. ville, NY, 1999, pp 193–203.
Garber K. (2005). J Natl Cancer Inst 97: 790–792. Price LS, Collard JG. (2001). Semin Cancer Biol 11: 167–173.
Geyer M, Wittinghofer A. (1997). Curr Opin Struct Biol 7: Ridley AJ, Paterson HF, Johnston CL, Diekmann D, Hall A.
786–792. (1992). Cell 70: 401–410.
Greenberg DL, Mize GJ, Takayama TK. (2003). Biochemistry Ridley AJ. (2001). J Cell Sci 114: 2713–2722.
42: 702–709. Ridley AJ, Hall A. (1992a). Cell 70: 389–399.
Hall A. (1990). Science 249: 635–640. Ridley AJ, Hall A. (1992b). Cold Spring Harb Symp Quant Biol
Hall A. (1998). Science 279: 509–514. 57: 661–671.
Hellawell GO, Turner GD, Davies DR, Poulsom R, Brewster Rubin J, Chung LW, Fan X, Zhu L, Murphy TC, Nanes MS
SF, Macaulay VM. (2002). Cancer Res 62: 2942–2950. et al. (2004). Prostate 58: 41–49.
Hendrix MJ, Seftor EA, Seftor RE, Fidler IJ. (1987). Cancer Sahai E, Marshall CJ. (2002). Nat Rev Cancer 2: 133–142.
Lett 38: 137–147. Sander EE, ten Klooster JP, van Delft S, van der Kammen
Hodge JC, Bub J, Kaul S, Kajdacsy-Balla A, Lindholm PF. RA, Collard JG. (1999). J Cell Biol 147: 1009–1022.
(2003). Cancer Res 63: 1359–1364. Shinto E, Tsuda H, Matsubara O, Mochizuki H. (2003).
Horiuchi A, Imai T, Wang C, Ohira S, Feng Y, Nikaido T Nippon Rinsho 61(Suppl 7): 215–219.
et al. (2003). Lab Invest 83: 861–870. Somlyo AV, Phelps C, Dipierro C, Eto M, Read P, Barrett M
Imamura F, Mukai M, Ayaki M, Takemura K, Horai T, et al. (2003). FASEB J 17: 223–234.
Shinkai K et al. (1999). Clin Exp Metast 17: 141–148. Somlyo AV, Bradshaw D, Ramos S, Murphy C, Myers CE,
Iwamura M, Sluss PM, Casamento JB, Cockett AT. (1993). Somlyo AP. (2000). Biochem Biophys Res Commun 269:
Prostate 22: 243–252. 652–659.
Jackson JG, Zhang X, Yoneda T, Yee D. (2001). Oncogene 20: Suwa H, Ohshio G, Imamura T, Watanabe G, Arii S,
7318–7325. Imamura M et al. (1998). Br J Cancer 77: 147–152.
Jaffe AB, Hall A. (2002). Adv Cancer Res 84: 57–80. Takai Y, Sasaki T, Matozaki T. (2001). Physiol Rev 81:
Kamai T, Tsujii T, Arai K, Takagi K, Asami H, Ito Y et al. 153–208.
(2003). Clin Cancer Res 9: 2632–2641. Tanno S, Tanno S, Mitsuuchi Y, Altomare DA, Xiao GH,
Kaverina I, Krylyshkina O, Small JV. (2002). Int J Biochem Testa JR. (2001). Cancer Res 61: 589–593.
Cell Biol 34: 746–761. Thalmann GN, Sikes RA, Wu TT, Degeorges A, Chang SM,
Keely P, Parise L, Juliano R. (1998). Trends Cell Biol 8: Ozen M et al. (2000). Prostate 44: 91–103.
101–106. Turner CE. (2000). J Cell Sci 113(Part 23): 4139–4140.
Kiefer JA, Farach-Carson MC. (2001). Matrix Biol 20: van Golen KL, Davies S, Wu ZF, Wang Y, Bucana CD, Root
429–437. H et al. (1999). Clin Cancer Res 5: 2511–2519.
Kimura G, Kasua J, Giannini S, Honda Y, Mohan S, Kawachi van Golen KL, Wu ZF, Qiao XT, Bao LW, Merajver SD.
M et al. (1996). Int J Urol 3: 39–46. (2000a). Cancer Res 60: 5832–5838.
Kjoller L, Hall A. (1999). Exp Cell Res 253: 166–179. van Golen KL, Wu ZF, Qiao XT, Bao L, Merajver SD.
Kleer CG, van Golen KL, Zhang Y, Wu ZF, Rubin MA, (2000b). Neoplasia 2: 418–425.
Merajver SD. (2002). Am J Pathol 160: 579–584. van Golen KL, Bao L, DiVito MM, Wu Z, Prendergast GC,
Kondo T, Sentani K, Oue N, Yoshida K, Nakayama H, Yasui Merajver SD. (2002). Mol Cancer Ther 1: 575–583.
W. (2004). Pathobiology 71: 19–25. van Golen KL. (2003). Breast Cancer Res 5: 174–179.
Kwon T, Kwon DY, Chun J, Kim JH, Kang SS. (2000). J Biol van Golen KL, Bao LW, Pan Q, Miller FR, Wu ZF, Merajver
Chem 275: 423–428. SD. (2002). Clin Exp Metastasis 19: 301–311.
Lopez T, Hanahan D. (2002). Cancer Cell 1: 339–353. Wang W, Yang LY, Yang ZL, Huang GW, Lu WQ. (2003).
Marionnet C, Lalou C, Mollier K, Chazal M, Delestaing G, World J Gastroenterol 9: 1950–1953.
Compan D et al. (2003). Oncogene 22: 3500–3505. West K, Zhang H, Brown M, Nikolopoulos S, Riedy M,
Meyer GE, Shelden E, Kim B, Feldman EL. (2001). Oncogene Horwitz A et al. (2004). J Cell Biol 154: 161–176.
20: 7542–7550. Yin JJ, Pollock CB, Kelly K. (2005). Cell Res 15: 57–62.
Mitsiades CS, Mitsiades N. (2005). Expert Rev Anticancer Ther Zaidel-Bar R, Ballestrem C, Kam Z, Geiger B. (2003). J Cell
5: 487–499. Sci 116: 4605–4613.
Moorman JP, Luu D, Wickham J, Bobak DA, Hahn CS. Zhang H, Yee D. (2004). Expert Opin Investig Drugs 13:
(1999). Oncogene 18: 47–57. 1569–1577.
Moscow JA, He R, Gnarra JR, Knutsen T, Weng Y, Zhao WP Zondag GC, Evers EE, ten Klooster JP, Janssen L, van der
et al. (1994). Oncogene 9: 189–194. Kammen RA, Collard JG. (2000). J Cell Biol 149: 775–782.

Oncogene

You might also like