ArticlePDF Available

Activation of Tumor-Specific Splice Variant Rac1b by Dishevelled Promotes Canonical Wnt Signaling and Decreased Adhesion of Colorectal Cancer Cells

Authors:

Abstract and Figures

Rac1b is a tumor-specific splice variant of the Rac1 GTPase that displays limited functional similarities to Rac1. We have shown previously a novel cross-talk between Rac1 and beta-catenin, which induces canonical Wnt pathway activation in colorectal cancer cells. This prompted us to investigate if Rac1b, frequently overexpressed in colon tumors, contributes to Wnt pathway dysregulation. We show that Rac1b overexpression stimulates Tcf-mediated gene transcription, whereas depletion of Rac1b results in decreased expression of the Wnt target gene cyclin D1. Reconstitution experiments revealed an important difference between Rac1 and Rac1b such that Rac1b was capable of functionally interacting with Dishevelled-3 (Dvl-3) but not beta-catenin to mediate synergistic induction of Wnt target genes. In agreement, Dvl-3 but not beta-catenin caused increased activation of Rac1b levels, which may explain the functional cooperativity displayed in transcription assays. Furthermore, we show that Rac1b negatively regulates E-cadherin expression and results in decreased adhesion of colorectal cancer cells. RNA interference-mediated suppression of Rac1b resulted in reduced expression of Slug, a specific transcriptional repressor of E-cadherin, and a concomitant increase in E-cadherin transcript levels was observed. Intriguingly, mutation of the polybasic region of Rac1b resulted in complete loss of Rac1b stimulatory effects on transcription and suppressive effects on adhesion, indicating the importance of nuclear and membrane localization of Rac1b. Our results suggest that Rac1b overexpression may facilitate tumor progression by enhancing Dvl-3-mediated Wnt pathway signaling and induction of Wnt target genes specifically involved in decreasing the adhesive properties of colorectal cancer cells.
Rac1b expression stimulates transcription of Wnt-responsive promoter TOPflash in HCT116 colorectal cancer cells. A, top, endogenous Rac1b expression was analyzed by RT-PCR in a panel of human colorectal and breast cancer cell lines: breast (MDA-MB-231 and MCF-7) and colon (SW480, HT29, HCT116, and SW48); bottom, endogenous protein expression and activation status of Rac1b was analyzed by Western blotting of whole-cell lysates (10 Ag) and PBD pull-down lysates, respectively, in human embryonic kidney HEK293 cells, HCT116 and SW480 colorectal cancer cells, and MCF-7 breast cancer cells. B, pTOPflash (5) or pFOPflash (n) reporter constructs were transiently transfected with increasing amount of Rac1b expression vector (0.2, 0.4, and 0.9 Ag) or WT Rac1 (0.4 and 0.9 Ag) for 24 h in HCT116 cells. Luciferase activity is expressed as total relative light units (RLU ). Columns, average of experiments carried out in triplicate; bars, SE. Empty vector (À) was transfected to establish basal TOPflash/ FOPflash activity. C, quantification of knockdown of endogenous Rac1b gene expression. HT29 cells were transfected with a scrambled control siRNA (negative control), Rac1 siRNA (targets both Rac1 and Rac1b), or siRNA selectively targeting Rac1b for 24 h and then subjected to real-time RT-PCR to quantitate Rac1/ Rac1b gene expression. Data are triplicate values of at least two independent experiments. D, endogenous cyclin D1 transcript expression was analyzed following specific Rac1b knockdown in HT29 cells under serum-starved conditions. Cells were transfected with Rac1b siRNA and then changed to low-serum conditions (0.1% fetal bovine serum) 4 h later. RNA was harvested for real-time RT-PCR after 20 h.
… 
Dvl-3 stimulates activation of Rac1b and cooperatively induces transcription of cyclin D1. A, FLAG-tagged Rac1b was transiently cotransfected with increasing concentrations of either h-catenin or Dvl-3 (0, 1, and 4 Ag) to determine activation status in transiently transfected HEK293 cells. Top, active GTP-bound Rac1b was determined by glutathione S -transferase (GST) pull-down assays using the PBD followed by immunoblot analysis using anti-FLAG antibody to detect exogenous Rac1b; bottom, total expression levels of exogenously transfected FLAG-tagged Rac1b were determined by immunoblot analysis of whole-cell lysates (10 Ag). B, HEK293 cells were transfected with cyclin D1 promoter-luciferase construct (CycD1-luc ) and Dvl-3 or Rac1b (0.4 Ag) alone or in combination. Luciferase activity is expressed as total relative light units. Columns, average of experiment carried out in triplicate; bars, SE. C, top, complex formation of Rac1b with Dvl-3 and h-catenin was examined in HEK293 cells cotransfected with FLAG-tagged Rac1b and Dvl-3 for 24 h. Whole-cell lysates (WCL ), cytoplasmic extracts (CE ), or nuclear extracts (NE ) were immunoprecipitated (IP ) with anti–Dvl-3 or anti–h-catenin antibody or agarose beads alone (negative control). Immunoblotting was done with indicated antibodies. FLAG-specific antibody was used to detect FLAG-tagged Rac1b. Bottom, 10 Ag WCL, CE, and NE were immunoblotted with h-catenin, Dvl, or FLAG antibodies to confirm expression of endogenous h-catenin and exogenous Rac1b and Dvl-3 in different cellular fractions.
… 
Content may be subject to copyright.
2007;67:2469-2479. Cancer Res
Susmita Esufali, George S. Charames, Vaijayanti V. Pethe, et al.
Decreased Adhesion of Colorectal Cancer Cells
Dishevelled Promotes Canonical Wnt Signaling and
Activation of Tumor-Specific Splice Variant Rac1b by
Updated version
http://cancerres.aacrjournals.org/content/67/6/2469
Access the most recent version of this article at:
Material
Supplementary
http://cancerres.aacrjournals.org/content/suppl/2007/03/16/67.6.2469.DC1.html
Access the most recent supplemental material at:
Cited Articles
http://cancerres.aacrjournals.org/content/67/6/2469.full.html#ref-list-1
This article cites by 48 articles, 17 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/67/6/2469.full.html#related-urls
This article has been cited by 6 HighWire-hosted articles. Access the articles at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
Activation of Tumor-Specific Splice Variant Rac1b by Dishevelled
Promotes Canonical Wnt Signaling and Decreased Adhesion of
Colorectal Cancer Cells
Susmita Esufali,1,2,3 George S. Charames,1,2,3 Vaijayanti V. Pethe,1,2 Pinella Buongiorno,1,2,3
and Bharati Bapat1,2,3
1Samuel Lunenfeld Research Institute and 2Department of Pathology and Laboratory Medicine, Mount Sinai Hospital; 3Department of
Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
Abstract
Rac1b is a tumor-specific splice variant of the Rac1 GTPase
that displays limited functional similarities to Rac1. We have
shown previously a novel cross-talk between Rac1 and B-
catenin, which induces canonical Wnt pathway activation in
colorectal cancer cells. This prompted us to investigate if
Rac1b, frequently overexpressed in colon tumors, contributes
to Wnt pathway dysregulation. We show that Rac1b over-
expression stimulates Tcf-mediated gene transcription, where-
as depletion of Rac1b results in decreased expression of the
Wnt target gene cyclin D1. Reconstitution experiments
revealed an important difference between Rac1 and Rac1b
such that Rac1b was capable of functionally interacting
with Dishevelled-3 (Dvl-3) but not B-catenin to mediate
synergistic induction of Wnt target genes. In agreement, Dvl-
3 but not B-catenin caused increased activation of Rac1b
levels, which may explain the functional cooperativity dis-
played in transcription assays. Furthermore, we show that
Rac1b negatively regulates E-cadherin expression and results
in decreased adhesion of colorectal cancer cells. RNA
interference–mediated suppression of Rac1b resulted in
reduced expression of Slug, a specific transcriptional repres-
sor of E-cadherin, and a concomitant increase in E-cadherin
transcript levels was observed. Intriguingly, mutation of the
polybasic region of Rac1b resulted in complete loss of Rac1b
stimulatory effects on transcription and suppressive effects
on adhesion, indicating the importance of nuclear and
membrane localization of Rac1b. Our results suggest that
Rac1b overexpression may facilitate tumor progression by
enhancing Dvl-3–mediated Wnt pathway signaling and induc-
tion of Wnt target genes specifically involved in decreasing the
adhesive properties of colorectal cancer cells. [Cancer Res
2007;67(6):2469–79]
Introduction
Rac1 is one of the most extensively studied members of the Rho
family of small GTP binding proteins. It regulates diverse cellular
processes, which include actin cytoskeleton organization, mem-
brane trafficking, proliferation, and gene expression (1–4). Similar
to all members of the Ras superfamily proteins, the GTP binding/
GTP hydrolysis state of Rac1 is tightly controlled by guanine
nucleotide exchange factors (GEF) and GTPase-activating proteins
(GAP), respectively. An additional level of regulation exists for Rho
GTPases, whereby Rho-GDP dissociation inhibitors (Rho-GDI)
sequester GDP-bound forms in the cytoplasm, preventing their
activation. Exhaustive characterization of this GTPase has revealed
the pivotal role it plays in the genesis of many cancers. A recently
discovered splice variant of Rac1, designated as Rac1b, has also
received much attention for its plausible role in tumorigenesis of
colon and breast tissues. However, deciphering the functional
consequences of its expression and contribution to tumorigenesis
has been challenging because Rac1b shows limited similarities to
known Rac1 functions.
Overexpression of Rac1, as well as altered function of Rac1-
specific regulators (GEFs, GAPs, and GDIs) or downstream effec-
tors, have been found in several cancers (5). Studies using
activated point mutants of Rac1 (G12V or Q61L) indicate that
aberrant activation of Rac1 can alter many cellular processes
important for cancer progression. Rac1 is essential for cell cycle
progression and activates several pathways important for cellular
proliferation, such as serum response factor, cyclin D1, and E2F
(6, 7). Rac1 also promotes cell survival by activating the nuclear
factor-nB (NF-nB) pathway and by preventing anoikis and apop-
tosis (8–10). Rac1 can induce cellular transformation in rodent
fibroblast models and is required for Ras-induced transformation
(11–14). Rac1 also regulates cell-cell and cell-matrix adhesion as
well as stimulates motility and invasion by modulating the actin
cytoskeleton, activities important for tumor metastasis (15).
Rac1b is a naturally occurring splice variant, preferentially
expressed in colon and breast tumors (16, 17). It is created by
alternative splicing of an additional exon, resulting in a 19–amino
acid insertion between codon 75 and 76 of Rac1, directly C-
terminus to the switch II region. This structural modification
enables it to behave as a constitutively activated GTPase, largely
because of an accelerated GEF-independent guanine nucleotide
exchange rate, a decreased intrinsic GTPase activity, and an
inability to interact with Rho-GDI (18, 19). It has been postulated
that the insertion may create a novel effector binding site, which
may enable Rac1b to participate in signaling pathways related to
neoplastic growth (19). Most studies to date indicate impaired
effector signaling activity of Rac1b. Unlike Rac1, Rac1b does not
activate the protein kinases c-Jun NH2-terminal kinase (JNK) and
p21-activated kinase 1 and is not involved in lamellipodia
formation. However, similar to Rac1, Rac1b can promote growth
transformation of NIH3T3 cells as well as a loss of density-
dependent and anchorage-dependent growth and stimulates AKT
serine/threonine kinase, hence promoting cell survival (20).
Furthermore, a constitutively activated Q61L Rac1b mutant, but
Note: Supplementary data for this article are available at Cancer Research Online
(http://cancerres.aacrjournals.org/).
Requests for reprints: Bharati Bapat, Samuel Lunenfeld Research Institute, Mount
Sinai Hospital, Room L6-304B, 60 Murray Street, Toronto, Ontario, Canada M5T 3L9.
Phone: 416-586-4800, ext. 5175; Fax: 416-586-8869; E-mail: bapat@mshri.on.ca.
I2007 American Association for Cancer Research.
doi:10.1158/0008-5472.CAN-06-2843
www.aacrjournals.org 2469 Cancer Res 2007; 67: (6). March 15, 2007
Research Article
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
not wild-type (WT) Rac1b, can stimulate the transcription factor
NF-nB and cyclin D1 expression (19, 20). Rac1b has been shown to
induce epithelial-mesenchymal transition (EMT) in mouse mam-
mary carcinoma cells via reactive oxygen species–dependent
activation of the transcription factor Snail (21).
Although these studies implicate a role for Rac1b in cellular
transformation, downstream signaling pathways by which this
tumor-associated splice variant may contribute to colorectal
tumorigenesis remain to be identified. The canonical h-catenin–
mediated Wnt signaling pathway is aberrantly activated in the vast
majority of colorectal cancers (22, 23). A hallmark feature of Wnt
pathway activation is nuclear translocation of h-catenin, where it
complexes with Tcf4 and inappropriately activates transcription of
target genes important for tumorigenesis (24, 25). We have shown
previously that Rac1 can cooperate with h-catenin to augment
canonical Wnt pathway activation by promoting its nuclear
accumulation and amplifying its transcription coactivator function
(26). This prompted us to investigate if Rac1b, frequently overex-
pressed in colon tumors, contributes to Wnt pathway dysregula-
tion, which is characteristically activated in these tumors. We
describe that Rac1b is a downstream target of Dishevelled-3 (Dvl-3)
and amplifies Dvl-3–initiated Wnt pathway activation leading to
increased h-catenin/Tcf–dependent transcription. Furthermore, we
show that Rac1b negatively regulates E-cadherin expression and
results in decreased adhesion of colorectal cancer cells. Thus, our
results contribute to the understanding of Rac1b as a putative
accelerator of tumor progression by positively regulating the
expression of proliferation-promoting genes and decreasing the
adhesive properties of colorectal cancer cells.
Materials and Methods
Cell culture and transfection. HEK293, SW480, and MCF-7 cells were
maintained in DMEM. HCT116 and HT29 cells were cultured in McCoy’s 5A.
SW48 and MDA-MB-231 cells were cultured in Leibovitz’s L-15 medium. All
media were supplemented with 10% (v/v) fetal bovine serum (Invitrogen,
Carlsbad, CA) and cultured at 37jC in a humidified atmosphere of 5% CO
2
.
Cells were transfected as described previously (26). For endogenous gene
knockdown experiments, the following small interfering RNAs (siRNA) were
used: Rac1 siRNA was a SMARTpool reagent (Dharmacon, Lafayette, CO),
whereas Rac1b knockdown was achieved via an equal mixture of two
siRNAs against the target sequences 5-GAAACGUACGGUAAGGAUA-3and
5-GGCAAAGACAAGCCGAUUG-3. Gene expression data were normalized
against transfections with a siCONTROL nontargeting siRNA (Dharmacon).
Plasmids. Dvl-3 construct was purchased from American Type Culture
Collection (Manassas, VA). Activator protein (AP-1)–luc and serum
response element (SRE)–luc luciferase reporter constructs were purchased
from Stratagene (La Jolla, CA). To generate the Rac1b polybasic region
(PBR) mutants, site-directed mutagenesis was done using the QuikChange
Site-Directed Mutagenesis kit (Stratagene). Using a WT FLAG-tagged Rac1b
cDNA construct as template, the KKRKRK sequence at amino acids 202 to
207 of Rac1b was replaced sequentially by pairs of glutamines to create Q2,
Q4, and Q6 mutants as depicted in Fig. 4A.
Reverse transcription-PCR. cDNA synthesis was done using 5 Ag total
RNA, random primer p(dN
6
), and SuperScript II reverse transcriptase. Rac1
and Rac1b mRNA levels were analyzed using the following primers specific
for both Rac1 and Rac1b: 5-ATGCAGGCCATCAAGTGTGTG-3( forward)
and 5-CAACAGCAGGCATTTTCTCTT-3(reverse). The PCR conditions
used were as follows: 1 cycle at 94jC for 4 min; 35 cycles at 94jC for 30 s,
55jC for 45 s, and 72jC for 30 s; and 1 cycle at 72jC for 5 min. PCR products
were separated on 1.5% agarose gel stained with ethidium bromide.
Quantitative real-time reverse transcription-PCR. First-strand cDNA
was synthesized from 5 Ag RNA using SuperScript III reverse transcriptase
(Invitrogen). A mixture of 2SYBR Green (15 AL/well), 10 Amol/L of
forward and reverse primer (0.6 AL each/well), 1 AL cDNA, and DNase-free
water to bring the reaction to 30 AL was added to each well. Samples were
analyzed in triplicate and carried out in an Applied Biosystems 7500 Real-
time PCR System (Foster City, CA). Real-time results were collected and
analyzed (standard curve method) using the ABI 7500 System software
according to the manufacturer’s protocol. Expression values were
normalized with the b-actin gene expression values. Primer sequences
used to amplify cDNA are described in Supplementary Table S1.
Luciferase reporter gene assays. Cells were plated at a seeding density
of 2 10
5
per well of a 24-well dish 24 h before transfection. Luciferase
reporter constructs (0.1 Ag) were added to each well together with 0.03 Ag
pCMV h-galactosidase construct for normalization. For coexpression of
additional proteins, FLAG-tagged WT or PBR-mutated Rac1b, WT h-
catenin, or Dvl-3 constructs were added as indicated in figures. Cells were
harvested after 24 h in 100 AL reporter lysis buffer (Promega, Madison, WI).
Assays of luciferase and h-galactosidase activity were done as described
previously (26).
Cell fractionation, coimmunoprecipitation, and Western blotting.
Whole-cell lysates as well as cytoplasmic and nuclear extracts were
prepared and detected as described (26). Coimmunoprecipitation experi-
ments were done using both whole-cell lysates and fractionated cytoplasmic
and nuclear lysates. The antibodies used in this study were horseradish
peroxidase–conjugated anti-FLAG (Sigma, St. Louis, MO), monoclonal anti–
h-catenin and monoclonal anti-paxillin (Transduction Laboratories, BD
Biosciences, Franklin Lakes, NJ), monoclonal anti–topoisomerase II
(Oncogene Research Products, San Diego, CA), monoclonal Dvl-3 (Santa
Cruz Biotechnology, Santa Cruz, CA), and anti–h-actin (Ambion, Austin,
TX) as a loading control.
Active Rac pull-down experiments. Active GTP-bound Rac1 and
Rac1b levels were determined using a nonradioactive Rac1 activation
assay kit according to the manufacturer’s protocol (Pierce Biotechnology,
Rockford, IL). Following transfection, cells were washed in cold PBS and
lysed on ice in 600 AL lysis buffer. Total lysates were cleared by
centrifugation at 16,000 gfor 15 min, and 100 AL lysate was kept for
protein quantitation of total Rac1/Rac1b protein. The remaining lysate
was incubated for 30 min at 4jC with 20 Ag PAK binding domain (PBD)
agarose beads. Precipitated complexes were washed thrice with excess
lysis buffer. After the final wash, the supernatant was discarded and 40 AL
of 2Laemmli sample buffer were added to the beads. Total lysates and
precipitates were then analyzed by Western blot.
Fluorescence microscopy. HCT116 cells cultured on glass coverslips
were transfected with indicated constructs and harvested after 24 h by
fixing in formaldehyde (3.7%, 30 min, room temperature) and permeabiliz-
ing in Triton X-100 (0.2%, 10 min, room temperature). Cells were incubated
in blocking buffer (1% goat serum, 1 h, room temperature) followed by
incubation with appropriate primary and fluorescently labeled secondary
antibodies. FLAG-tagged constructs were detected with rabbit polyclonal
anti-FLAG antibody (Sigma) followed by Texas red–conjugated anti-
rabbit polyclonal antibody (Molecular Probes, Eugene, OR). Endogenous
E-cadherin was visualized using mouse monoclonal anti–E-cadherin
(Transduction Laboratories) and antimouse FITC-conjugated secondary
antibody (Molecular Probes). Actin was stained using an actin-phalloidin
conjugate (Molecular Probes). The level of nonspecific background
immunostaining was established by omitting primary antibodies. Coverslips
were mounted in Vectashield (Vector Laboratories, Burlingame, CA) and
examined using the Olympus (Markham, Ontario, Canada) 1X-70 inverted
deconvolution microscope (100 lens magnification).
Adhesion assay. HCT116 colorectal cancer cells were seeded at a cell
density of 2 10
6
per 60-mm tissue culture dish the day before transfection.
Cells were transfected with indicated constructs, and DNA concentration
was held constant at 8 Ag per dish. Cells were harvested 24 h post-
transfection by trypsinization, resuspended in serum-free McCoy’s 5A
medium at cell density of 100,000 and 400,000 cells/mL, and used in the
Innocyte Adhesion Assay from Calbiochem (San Diego, CA). Briefly, 100 AL
of prepared cell suspension were added, in triplicate, to wells of 96-well
dish, coated with poly-L-lysine (general attachment) or bovine serum
albumin (negative control), and incubated 2 h at 37jC. Wells were gently
Cancer Research
Cancer Res 2007; 67: (6). March 15, 2007 2470 www.aacrjournals.org
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
washed twice with PBS to remove nonadherent cells and then incubated
with 100 AL alcein-AM green fluorescent dye solution for an additional 1 h at
37jC. Adherent cells were quantitated by measuring fluorescence of the
samples using a fluorescent plate reader at excitation wavelength 485 nm
and emission wavelength 520 nm.
Results
Rac1b induces transcriptional activation of Wnt-responsive
promoters in colorectal cancer cells. The splice variant Rac1b
exhibits selective expression, largely confined to colonic mucosa
and crypt epithelial cells, and is overexpressed specifically in colon
and breast tumors (16, 17). To assess the role of Rac1b in Wnt
pathway regulation, we first determined its endogenous expression
profile in a panel of colon cancer cell lines harboring aberrant Wnt
pathway activation. Using reverse transcription-PCR (RT-PCR)
analysis, we observed expression of the Rac1b transcript in SW480
and HT29 colorectal cancer cells as well as MCF-7 breast cancer
cells and very low levels in HCT116 and SW48 colorectal cancer
cells (Fig. 1A, top). Concordantly, cells expressing Rac1b transcript
also showed Rac1b protein expression (Fig. 1A, bottom). The level of
activation of Rac1b, assessed by PBD pull down-assay, was
comparable with Rac1 in SW480 and MCF-7 cells, although
examination of corresponding whole-cell lysates indicated far
more Rac1 versus Rac1b total protein in these cells. This
emphasizes that Rac1b is more efficiently activated than Rac1,
which agrees with previously published reports (19, 20).
Because Rac1b expression was minimally detected in HCT116
cells, they were used to evaluate the role of Rac1b overexpres-
sion in Wnt pathway regulation. A hallmark feature of canonical
Wnt pathway activation is h-catenin/Tcf–mediated transcription
of Wnt target genes. h-catenin/Tcf–dependent gene expression
was tested with the Wnt-responsive Tcf reporter TOPflash
together with the mutant reporter FOPflash as a negative
control. As shown in Fig. 1B(top), transient transfection of
Rac1b led up to a 7-fold activation of h-catenin/Tcf–mediated
transcription of TOPflash. Under similar conditions, WT Rac1
also stimulated TOPflash activity in HCT116 cells although much
less robustly compared with Rac1b, reaching only a maximal 2-
fold induction (Fig. 1B, bottom). To further elucidate the specific
contribution of Rac1b in Wnt pathway activation, we down-
regulated endogenous Rac1b in HT29 cells by transfection of
small interfering RNAs (siRNA) targeting the splice insertion
sequence within Rac1b. As shown in Fig. 1C(right), f70%
specific knockdown of Rac1b was achieved and did not affect
WT Rac1 transcript levels. We examined the effects of Rac1b
knockdown on the regulation of an endogenous Wnt target gene
Figure 1. Rac1b expression stimulates
transcription of Wnt-responsive promoter
TOPflash in HCT116 colorectal cancer
cells. A, top, endogenous Rac1b
expression was analyzed by RT-PCR in a
panel of human colorectal and breast
cancer cell lines: breast (MDA-MB-231 and
MCF-7) and colon (SW480, HT29,
HCT116, and SW48); bottom,
endogenous protein expression and
activation status of Rac1b was analyzed by
Western blotting of whole-cell lysates
(10 Ag) and PBD pull-down lysates,
respectively, in human embryonic kidney
HEK293 cells, HCT116 and SW480
colorectal cancer cells, and MCF-7 breast
cancer cells. B, pTOPflash (5)or
pFOPflash (n) reporter constructs were
transiently transfected with increasing
amount of Rac1b expression vector
(0.2, 0.4, and 0.9 Ag) or WT Rac1 (0.4 and
0.9 Ag) for 24 h in HCT116 cells.
Luciferase activity is expressed as total
relative light units (RLU ). Columns,
average of experiments carried out in
triplicate; bars, SE. Empty vector ()was
transfected to establish basal TOPflash/
FOPflash activity. C, quantification of
knockdown of endogenous Rac1b gene
expression. HT29 cells were transfected
with a scrambled control siRNA (negative
control), Rac1 siRNA (targets both Rac1
and Rac1b), or siRNA selectively targeting
Rac1b for 24 h and then subjected to
real-time RT-PCR to quantitate Rac1/
Rac1b gene expression. Data are triplicate
values of at least two independent
experiments. D, endogenous cyclin D1
transcript expression was analyzed
following specific Rac1b knockdown in
HT29 cells under serum-starved
conditions. Cells were transfected with
Rac1b siRNA and then changed to
low-serum conditions (0.1% fetal bovine
serum) 4 h later. RNA was harvested for
real-time RT-PCR after 20 h.
Dishevelled-Mediated Activation of Rac1b
www.aacrjournals.org 2471 Cancer Res 2007; 67: (6). March 15, 2007
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
cyclin D1 and observed f40% reduction (Fig. 1D). These findings
suggest that Rac1b expression contributes to inappropriate
transcription of Wnt target genes in colorectal cancer cells.
Rac1b augments Dishevelled-mediated activation of Tcf/
Lef–dependent transcription. We have shown previously that
constitutively active V12Rac1 synergizes with stabilized h-catenin
to activate transcription of Wnt target genes in HEK293 cells (26).
To see if Rac1b acts analogously, Rac1b was cotransfected with the
TOPflash reporter into HEK293 cells, which have an intact and
therefore tightly regulated Wnt pathway. As shown in Fig. 2A,
Rac1b failed to activate TOPflash activity on its own compared
with 6-fold activation by h-catenin. Surprisingly, Rac1b did not
show functional synergy with h-catenin as shown previously for
V12Rac1. In agreement with our V12Rac1 studies, WT Rac1 showed
a remarkable synergy with h-catenin, increasing TOPflash activity
up to 30-fold (data not shown).
Failure to see a functional interaction between Rac1b and h-
catenin in HEK293 cells that could account for the transcriptional
activity observed in HCT116 cells prompted us to explore other
Wnt signaling components that lay upstream of h-catenin along
the Wnt signaling axis. Dishevelled is one such protein that is a
well-established positive regulator of Wnt signaling, functioning
very early in the cascade. It is believed that Dishevelled transmits
Wnt signals from Frizzled receptors by inhibiting glycogen
synthase kinase-3hactivity through an unknown mechanism,
enabling h-catenin to escape proteasomal degradation. Coexpres-
sion of Rac1b with Dvl-3 caused a striking transcriptional
activation of up to 11-fold of TOPflash compared with 6-fold
induction with Dvl-3 alone (Fig. 2B). Taken together, these data
suggest that the functional cross-talk between Rac1b and Wnt
pathway activation likely occurs downstream of Dvl-3 but
upstream of h-catenin.
Having established a role for Rac1b as an upstream activator of
transcription of Wnt target genes, we wished to compare its
transcription regulatory function with respect to other well-
established Rac1-specific transcriptional targets. Canonical Rac1
effector pathways that result in transcriptional regulation include
NF-nB, mitogen-activated protein kinase (MAPK), and JNK path-
ways (8, 27, 28). As depicted in Fig. 2C, Rac1b very modestly
activated NF-nB–dependent transcription in a dose-dependent
manner and exerted no effect on AP-1– or SRE-dependent gene
transcription, corresponding to MAPK and JNK pathway activation,
respectively. Furthermore, Rac1b showed a dose-dependent
activation of transcription of the cyclinD1 promoter (Fig. 2D),
which agrees with our observations of Rac1b suppression causing
reduced endogenous cyclin D1 expression (Fig. 1D). These
Figure 2. Rac1b cooperates with Dvl-3 to stimulate Wnt target gene transcription. Aand B, pTOPflash (5) or pFOPflash (n) reporter constructs were transiently
transfected with increasing amounts of Rac1b, h-catenin, or Dvl-3 expression vectors for 24 h in HEK293 cells. Amounts of expression constructs used are 0.2, 0.4, and
0.9 Ag. The lowest amount of h-catenin or Dvl-3 (0.2 Ag) was used in cotransfection experiments with Rac1b. Cand D, HEK293 cells were cotransfected with
0.9 Ag of empty vector () or 0.2 and 0.9 Ag of Rac1b expression vector and either NF-nB–, AP-1–, or SRE-responsive promoter luciferase reporter constructs (C)ora
full-length cyclin D1 promoter construct, CycD1-luc (D). Results are representative of at least three or more independent experiments. Luciferase activity is
expressed as total relative light units. Columns, average; bars, SE (A–D ).
Cancer Research
Cancer Res 2007; 67: (6). March 15, 2007 2472 www.aacrjournals.org
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
differential transcriptional responses reiterate the selective signal-
ing capability of Rac1b versus Rac1.
Dvl-3 activates Rac1b and together they synergistically
stimulate cyclin D1 transcription. Because Rac1b was unable
to activate TOPflash activity on its own but showed significant
functional synergy in the presence of Dvl-3 in HEK293 cells, we
speculated whether Rac1b activity was being modified by Dvl-3.
Using the PBD assay, we found that expression of h-catenin did not
change the activity of exogenously transfected Rac1b (Fig. 3A, top).
In contrast, cotransfection with Dvl-3 resulted in a significant
increase in Rac1b activation levels. These findings help to explain
why we observed a differential response in our TOPflash assay
(Fig. 2B). Rac1b is activated by Dvl-3 but not h-catenin and
consequently proceeds to stimulate transcription from the Wnt-
responsive promoter in a Dishevelled-dependent manner. Cyclin
D1 is a well-characterized target gene of both the Wnt and Rac1
signaling pathways (12, 29, 30). We observed cooperativity between
Dvl-3 and Rac1b in regulation of the cyclin D1 promoter (Fig. 3B).
Combined expression of Dvl-3 with Rac1b yielded >5-fold
activation of cyclin D1 promoter activity, which was greater than
their effects individually, suggesting a synergistic interaction.
Expression of Dvl-3 and Rac1b has been shown in both the
cytoplasm and the nucleus. A recent study has shown that nuclear
translocation of Dvl is essential for its ability to activate Wnt/h-
catenin signaling (31). In addition, we as well as others have
reported previously the expression of Rac1 in the nucleus (26, 32,
33). For this reason, we examined complex formation of Dvl-3 and
Rac1b in these specific cellular fractions. We observed complexes of
Rac1b and Dvl-3 in the cytoplasm as well as the nucleus (Fig. 3C).
Interestingly, Rac1b coimmunoprecipitated with Dvl-3 more effi-
ciently in the nuclear fraction. Similarly, endogenous h-catenin also
preferentially coimmunoprecipitated with Rac1b in the nuclear
fraction. Protein expression levels of Rac1b, Dvl-3, and h-catenin in
whole-cell lysates and cytoplasmic and nuclear fractions are
shown. Blots were stripped and reprobed for marker proteins for
cytoplasm and nucleus, paxillin and topoisomerase II, respectively.
Considered together, our data suggest that Dvl-3 and h-catenin can
both form complexes with Rac1b in the nucleus and cytoplasm.
PBR of Rac1b regulates its nuclear localization. The C-
terminal PBR of Rac1 has been shown to function as a nuclear
localization signal (NLS) and mutation of this region drastically
reduces its nuclear entry (34). To examine if the PBR of Rac1b was
involved in mediating its nuclear localization, we generated various
FLAG-tagged Rac1b PBR mutant constructs, by substituting the
basic amino acids of the PBR with the neutral amino acid
glutamine, depicted in Fig. 4A. We examined cellular distribution
of these mutants in fractionated lysates of HCT116 cells. As shown
in Fig. 4B, progressive mutation of the PBR resulted in a significant
progressive decrease in nuclear expression of Rac1b and was
accompanied by a corresponding progressive increase in cytoplas-
mic expression of Rac1b. The Rac1b (Q6) mutant, with all six basic
amino acids mutated, showed the greatest reduction in nuclear
expression.
Mutation of the PBR did not compromise Rac1b activation
levels. As shown in Fig. 4C, all PBR mutants (Q2, Q4, and Q6)
showed similar levels of activation to WT PBR-intact Rac1b. We
also examined the distribution of Rac1b and Rac1b (Q6) by
fluorescence microscopy in HCT116 cells (Fig. 4D). Whereas Rac1b
was prominently localized to the plasma membrane, and to a lesser
extent in the cytoplasm and nucleus, Rac1b (Q6) transfectants
showed complete loss of membrane staining, distinct exclusion
Figure 3. Dvl-3 stimulates activation of Rac1b and cooperatively induces
transcription of cyclin D1. A, FLAG-tagged Rac1b was transiently cotransfected
with increasing concentrations of either h-catenin or Dvl-3 (0, 1, and 4 Ag) to
determine activation status in transiently transfected HEK293 cells. Top, active
GTP-bound Rac1b was determined by glutathione S-transferase (GST)
pull-down assays using the PBD followed by immunoblot analysis using
anti-FLAG antibody to detect exogenous Rac1b; bottom, total expression
levels of exogenously transfected FLAG-tagged Rac1b were determined by
immunoblot analysis of whole-cell lysates (10 Ag). B, HEK293 cells were
transfected with cyclin D1 promoter-luciferase construct (CycD1-luc ) and Dvl-3
or Rac1b (0.4 Ag) alone or in combination. Luciferase activity is expressed as
total relative light units. Columns, average of experiment carried out in triplicate;
bars, SE. C, top, complex formation of Rac1b with Dvl-3 and h-catenin was
examined in HEK293 cells cotransfected with FLAG-tagged Rac1b and Dvl-3 for
24 h. Whole-cell lysates (WCL ), cytoplasmic extracts (CE ), or nuclear extracts
(NE) were immunoprecipitated (IP) with anti–Dvl-3 or anti–h-catenin antibody or
agarose beads alone (negative control). Immunoblotting was done with indicated
antibodies. FLAG-specific antibody was used to detect FLAG-tagged Rac1b.
Bottom, 10 Ag WCL, CE, and NE were immunoblotted with h-catenin, Dvl, or
FLAG antibodies to confirm expression of endogenous h-catenin and exogenous
Rac1b and Dvl-3 in different cellular fractions.
Dishevelled-Mediated Activation of Rac1b
www.aacrjournals.org 2473 Cancer Res 2007; 67: (6). March 15, 2007
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
from the nucleus, and diffuse cytoplasmic staining. The cells were
costained with actin to distinguish individual cells. Taken together,
the PBR of Rac1b controls both membrane localization and nuclear
accumulation of Rac1b. We used the Rac1b (Q6) mutant in
subsequent experiments because it exhibited the greatest reduction
in plasma membrane and nuclear localization.
Mutation of PBR compromises the transcription activator
function of Rac1b and the association of Dishevelled and B-
catenin with Rac1b. To gain insight into the role of subcellular
localization on Rac1b functions, we examined the transcription
activator potential of PBR-mutated Rac1b. As shown in Fig. 5A,
Rac1b (Q6) was unable to activate transcription of the TOPflash
promoter in HCT116 cells compared with WT Rac1b (Fig. 1B).
Consistent with these findings, the Rac1b (Q6) mutant also failed to
stimulate transcription from the NF-nB promoter (Fig. 5B) and
cyclin D1 promoter (Fig. 5C). These data show that the PBR of
Rac1b is important for mediating transcriptional activation.
Furthermore, disrupting plasma membrane and/or nuclear local-
ization of Rac1b interferes with this activity.
Because PBR-mutated Rac1b (Q6) was unable to stimulate
TOPflash activity, we speculated that activation by Dvl-3 may also
be compromised. Comparison of the activation levels of WT Rac1b
and Rac1b (Q6) in the presence of Dvl-3 indicate that Dvl-3 is
unable to further stimulate the activation of Rac1b (Q6) (Fig. 5D,
left). Because we have shown that Rac1b can complex with Dvl-3
and h-catenin (Fig. 3C), we examined whether they were associated
with active Rac1b by reprobing the PBD blot with Dvl-3 and h-
catenin antibodies. As shown in Fig. 5D(left), Dvl-3 and h-catenin
were associated with active GTP-bound Rac1b. Interestingly, the
Rac1b (Q6) pull-down fractions contained considerably reduced
levels of Dvl-3 and h-catenin. The reduced binding to Rac1b (Q6)
was not due to lack of expression of Dvl-3 and h-catenin, which
was confirmed by Western blotting of whole-cell lysates (Fig. 5D,
right). Because Rac1b (Q6) was refractory to further activation by
Figure 4. The PBR of Rac1b regulates its nuclear and plasma membrane localization but not activity. A, schematic representation of the Rac1b protein sequence
and an enlarged view of the C-terminal PBR sequence of WT Rac1b and the substitutions that were made to alter PBR function (Q2, Q4, and Q6). B, the expression of
FLAG-tagged Rac1b and the various FLAG-tagged PBR mutants (Q2, Q4, and Q6) were determined by Western blotting of fractionated cytoplasmic and nuclear
lysates of transiently transfected HEK293 cells. Blots were stripped and reprobed with topoisomerase II, paxillin, or h-actin antibodies to assess purity of nuclear and
cytosolic fractions and to normalize for protein loading, respectively. Interestingly, a doublet was observed in the Rac1b (Q6)–transfected cells. The doublet
could also be detected by an anti-Rac1 antibody on Western blots (data not shown). In addition, RT-PCR analysis of Rac1b (Q6) plasmid DNA did not show an
additional band. We conclude that the doublet is specific for Rac1b and may result due to a posttranslational modification. C, the activation status of Rac1b and
PBR-mutated forms of Rac1b were assessed by PBD assay. Indicated FLAG-tagged constructs were transiently transfected into HEK293 cells for 24 h. Top, active
GTP-bound Rac1b expression levels determined by PBD-GST pull-down assays followed by immunoblot analysis using anti-FLAG antibody; bottom, total cellular
expression of transfected Rac1b constructs determined by immunoblot analysis of whole-cell lysates (10 Ag) with anti-FLAG antibody. D, the distributions of
FLAG-tagged Rac1b and Rac1b (Q6) in HCT116 cells were determined by fluorescence microscopy. Cells were stained with actin-phalloidin to demarcate individual
cells. Approximately 50 to 60 transfected cells were examined. Bar, 10 Am.
Cancer Research
Cancer Res 2007; 67: (6). March 15, 2007 2474 www.aacrjournals.org
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
Dvl-3 and we failed to see association of Dvl-3 or h-catenin with
Rac1b (Q6), these observations strongly support the role of the
PBR in mediating binding and activation of Rac1b by Dvl-3.
Rac1b reduces E-cadherin expression and cellular adhesion
of colorectal cancer cells. Transcriptional activation of Wnt
target genes requires h-catenin accumulation and translocation to
the nucleus. Rac1b does not likely modulate proteasomal
degradation of h-catenin because our observations were made in
HCT116 cells, which already contain stabilized h-catenin (35).
Recently, it was shown that loss of E-cadherin can lead to Wnt-
dependent transcription in colorectal cancer cells (36). Therefore,
we next explored whether Rac1b may mediate its positive effects
on transcription by altering E-cadherin expression in HCT116 cells.
We observed that the total amount of endogenous E-cadherin
protein was decreased in the presence of Rac1b (Fig. 6A).
Densitometry indicated a 40% reduction of endogenous E-cadherin
levels in the presence of Rac1b. In agreement, specific suppression
of Rac1b using Rac1b siRNA resulted in a modest increase in the
expression of E-cadherin (Supplementary Fig. S1). Interestingly,
E-cadherin levels gradually returned to basal (untransfected) levels
with progressive mutation of the PBR, such that Rac1b (Q6)
showed minimal effects on E-cadherin expression (Fig. 6A). This
shows that the PBR is required for Rac1b-mediated regulation of
E-cadherin. Because the PBR is important in regulating the tar-
geting of Rac1b, it is possible that the loss of E-cadherin regulation
is related to change in its cellular localization.
Next, we wished to see if the membrane pool of E-cadherin
reflected the same changes as total E-cadherin. Overexpression of
Rac1b changed the distribution of endogenous E-cadherin by
causing a marked reduction of E-cadherin staining at the plasma
membrane (Fig. 6B) compared with untransfected cells. In contrast,
PBR-mutated Rac1b (Q6) did not alter E-cadherin staining at the
plasma membrane, which remained intact at cell-cell junctions.
Changes in E-cadherin expression often translate to changes in
cell-cell adhesion. We wanted to assess whether expression of
Rac1b changed the adhesive properties of HCT116 cells. Expression
of Rac1b decreased the adhesion of HCT116 cells, whereas Rac1b
(Q6)–transfected cells showed no change (Fig. 6C, left). Interest-
ingly, V12Rac1 caused an even more robust reduction in cell
adhesion compared with Rac1b. Because V12Rac1 is a constitu-
tively active mutant, we predicted that the greater reduction in
adhesion was attributed to its greater level of activity. Because we
Figure 5. Mutation of Rac1b PBR disrupts Rac1b-mediated transcriptional activation and activation by Dvl-3. A, HCT116 cells were transfected with pTOPflash/
pFOPflash promoter luciferase constructs and increasing doses of PBR-mutated Rac1b (Q6; 0.2, 0.4, and 0.9 Ag). Band C, HEK293 cells were transfected with
indicated promoter-luciferase constructs and either WT Rac1b or Rac1b (Q6; 0.4 Ag). D, activation status of FLAG-tagged WT Rac1b or Rac1b (Q6) in the absence ()
or presence of Dvl-3 (1 or 4 Ag). Indicated constructs were transiently transfected into HEK293 cells for 24 h. Left, active GTP-bound Rac1b/Q6 expression levels
determined by PBD-GST pull-down assays followed by immunoblot analysis using anti-FLAG antibody. Blots were stripped and reprobed with Dvl-3 and h-catenin
antibodies. Right, total cellular expression of transfected Rac1b/Q6 constructs determined by immunoblot analysis of whole-cell lysates (10 Ag) with anti-FLAG
antibody. Blots were stripped and reprobed for Dvl-3, h-catenin, and h-actin. Luciferase activity is expressed as total relative light units. Columns, average of
experiments carried out in triplicate; bars, SE (A–C ). Data are representative of three independent experiments (A–D ).
Dishevelled-Mediated Activation of Rac1b
www.aacrjournals.org 2475 Cancer Res 2007; 67: (6). March 15, 2007
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
have shown that Dvl-3 can increase the activation level of Rac1b,
we speculated whether Rac1b would decrease adhesion to a greater
extent in the presence of Dvl-3. Indeed, cotransfection of Dvl-3 with
Rac1b caused f80% reduction in adhesion of HCT116 cells
compared with Rac1b or Dvl-3 alone, which individually caused a
50% reduction in adhesion (Fig. 6C, right). A primary mechanism of
E-cadherin down-regulation in tumors is a result of the action of
transcriptional repressors (e.g., proteins of the Snail/Slug family),
which bind to E-box elements within the promoter region of E-
cadherin (37). This prompted us to investigate whether such
underlying molecular mechanisms were involved in mediating the
Rac1b-associated decrease in E-cadherin expression. Suppression
of endogenous Rac1b in HT29 cells resulted in increased E-
cadherin transcript expression and a concomitant decrease in Slug
Figure 6. Rac1b decreases the expression of E-cadherin and cellular adhesion in HCT116 cells. A, top, Western blot analysis of the total amount of E-cadherin after
24 h of transfection with Rac1b or PBR mutants of Rac1b. FLAG-tagged Rac1b and PBR-mutant expression was detected by probing blots with FLAG antibody.
Expression of actin was used as a loading control. Bottom, the band of densitometry was normalized with actin to allow quantification. Data show a significant decrease
in E-cadherin expression mediated by Rac1b, which is abolished on mutation of the PBR region. Data are representative of three independent experiments.
B, distribution of endogenous E-cadherin (red fluorescence) was compared in HCT116 cells transfected with FLAG-tagged Rac1b versus FLAG-tagged Rac1b (Q6),
or untransfected cells by fluorescence microscopy. Rac1b and Rac1b (Q6) transfectants were identified by green fluorescence. Bar, 10 Am. C, HCT116 cells were
transfected for 24 h with indicated constructs followed by assessment of adhesion on poly-L-lysine–coated surface. Results are the percentage of adhesion compared
with empty vector–transfected cells. Representative data obtained from triplicate values. Data are representative of at least two independent experiments.
D, E-cadherin (top) and Slug (bottom ) transcript levels in response to RNA interference–mediated inhibition of Rac1b expression in HT29 cells.
Cancer Research
Cancer Res 2007; 67: (6). March 15, 2007 2476 www.aacrjournals.org
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
(Snai2) transcript levels (Fig. 6D). We did not observe any change in
Snail transcript levels under similar conditions (data not shown).
These data agree of a repressive role for Rac1b in cellular adhesion
by inhibition of E-cadherin transcription.
Discussion
Because its discovery as a splice variant specifically overex-
pressed in colon and breast tumors, there has been considerable
interest in characterizing the functions of Rac1b to understand
how it may contribute to tumorigenesis in these tissues (16, 17).
Our data show functionality of naturally occurring Rac1b in
modulating Wnt signaling by augmenting Dishevelled-mediated
effects on Tcf-dependent target gene transcription. In addition, we
show that Rac1b decreases membrane E-cadherin expression and
reduces adhesion of colorectal cancer cells and this is likely via up-
regulation of Slug, the E-cadherin-specific transcriptional repressor.
The contribution of Rac1b to Wnt pathway activation may
represent an important mechanism by which it promotes colon
tumorigenesis. We have shown previously that V12Rac1 requires
the presence of stabilized h-catenin to mediate its stimulatory
effects on transcription. In agreement with these data, coexpres-
sion of WT Rac1 with h-catenin also resulted in synergistic
induction of TOPflash promoter activity in HEK293 cells (data not
shown). Unlike Rac1, Rac1b was unable to stimulate Tcf-dependent
transcription in HEK293 cells, despite coexpression of h-catenin.
However, when exogenous Dvl-3 was coexpressed with Rac1b in
these cells, we observed striking synergistic activation of TOPflash.
Interestingly, Dvl proteins have been noted to form cytoplasmic
puncta when overexpressed in cells, and their formation has been
correlated with their ability to induce Wnt pathway activation
(38, 39). In this regard, we observed a striking increase in the size
and number of Dvl-3 punctae in HCT116 cells cotransfected with
Dvl-3 and either Rac1b or L61Rac1 compared with Dvl-3 alone
(Supplementary Fig. S2). Interestingly, we also observed Rac1b and
L61Rac1 colocalize with Dvl-3 within these punctae. Understanding
how these vesicles form and correlate with Wnt signaling activity
may uncover new roles for Dvl and Rac1b/Rac1 proteins in
regulating the canonical arm of Wnt signaling.
It is tempting to speculate that Rac1b and Rac1 may act at
different stages of tumor progression to further exacerbate Wnt
signaling in colonic tissues. Our data show that Rac1b and Rac1
likely require different cellular cues, at which they can participate
in Wnt signaling. In contrast to Rac1, which can synergize with
h-catenin to augment Wnt signaling, Rac1b requires a signal
further upstream, likely stemming from Dishevelled proteins. This
implies that expression of Rac1b in colorectal tumor cells that
already harbor inherent Dishevelled-dependent activation of Wnt
pathway may increase the intensity of Wnt signaling within those
cells. This may be a critical step in tumor progression, assisting
tumor cells to metastasize to distal sites. Clinical evidence suggests
that Wnt signaling acts differently at different stages of tumor
progression (40). This is best exemplified by nuclear staining for h-
catenin, a hallmark of Wnt pathway activation, in colorectal
carcinomas, which often show a heterogeneous pattern, with
strongest nuclear enrichment at the invasion front. The molecular
basis for this differential distribution of h-catenin is not known, but
it has been speculated that signals from the mesenchymal tissue,
which surrounds the invasive tumor cells, might superactivate the
pathway by unknown ways. Matrix metalloproteinases (MMP) are
important microenvironmental factors present in mesenchymal
tissue, implicated in tumor initiation and progression. Interestingly,
some MMPs are Wnt target genes (41). Recently, Rac1b was shown
to be the key mediator of MMP-3–induced malignant transforma-
tion of mouse mammary epithelial cells (21). Specifically, MMP-3
treatment induced Rac1b expression, which was responsible for the
EMT of the cells. It is tempting to speculate that a positive feedback
loop exists, in which initial Wnt pathway activation induces
expression of a subset of gene products, such as MMPs, which
feedback in an autocrine or paracrine manner, on Wnt-dysregu-
lated cells to induce Rac1b expression. In turn, as our data suggest,
Rac1b expression would further augment nuclear Wnt signaling in
the cancer-initiated cells, promoting tumor progression by aiding
tumor cells in invasion and metastasis. Whether such a feedback
loop exists warrants future investigation.
Another important point worth noting is that overexpression of
the naturally occurring WT form of Rac1b was sufficient to induce
changes in Wnt signaling. Unlike the widely used ‘artificial’
constitutively active mutants (G12V or Q61L), WT forms of Rho
proteins are amenable to regulation and so their use can facilitate
identification of upstream or downstream signals that may be
critical in modulating expression and activity. In this context, our
findings are significant because overexpression of WT Rac1b had a
profound stimulatory effect, cooperating with Dvl-3 to increase
nuclear Wnt signaling events. Furthermore, we show that
expression of Dvl-3, but not h-catenin, leads to increased activation
of Rac1b. This is likely the reason why Rac1b augmented Tcf-
mediated transcription in the presence of Dvl-3 but not h-catenin.
Perhaps association of Dvl-3 with Rac1b not only further activates
Rac1b by modulating GEF/GAP activity but also may recruit other
proteins, such as h-catenin, to form functional signaling com-
plexes. Our findings that Dvl-3 and h-catenin were predominantly
present in the active GTP-bound fractions of Rac1b, in Dvl-
dependent manner, support this idea. Mechanisms of how Dvl-3
may increase activity of Rac1b are currently under investigation in
our laboratory. Aberrant expression of Dishevelled has been
observed in multiple cancers and has been linked to activation of
Wnt/h-catenin signaling and cell growth in these cancers. It would
be interesting to examine Rac1b expression and activity and
turnover in these tumors (42–44).
Because we observed nuclear expression of Rac1b, we wondered
if the putative NLS in the PBR was responsible. Substitution of all
six basic amino acids of the Rac1b PBR with neutral glutamines
resulted in almost complete exclusion of Rac1b from the nucleus,
as well as the plasma membrane. PBR-mutated Rac1b was unable
to activate transcription from TOPflash, cyclin D1, and NF-nB
promoters. It is tempting to speculate that the nuclear pool of
Rac1b is likely critical for transcription of Wnt target genes.
Interestingly, Dvl-3 was unable to augment activity of PBR-mutated
Rac1b, suggesting that it may mediate these effects within the
nucleus. In this regard, nuclear expression of many Rac1-specific
proteins, including GEFs and downstream effectors, further
supports the notion of Rac1 and Rac1b participating in nuclear
signaling pathways (33, 45). In agreement with this hypothesis, we
observed Dvl-3 coimmunoprecipitating with Rac1b mostly in the
nuclear fraction of cells, despite these proteins being present in the
cytoplasm, which lends further support for these effects occurring
in the nucleus.
In the colon, E-cadherin–mediated cell adhesion is critical in the
transition from adenoma to carcinoma, and reexpression of WT
E-cadherin in cancer cell lines reduces their invasiveness (46, 47).
Here, we show that expression of Rac1b in HCT116 colorectal
Dishevelled-Mediated Activation of Rac1b
www.aacrjournals.org 2477 Cancer Res 2007; 67: (6). March 15, 2007
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
cancer cells reduces endogenous E-cadherin expression and
cellular adhesion. Intriguingly, coexpression of Dvl-3 with Rac1b
resulted in a striking reduction of adhesion compared with effects
seen by either protein alone. Furthermore, we show that both
of these effects disappear on mutation of the PBR of Rac1b.
Because nuclear localization of Rac1b is disrupted by mutation of
the PBR, it is possible that Rac1b functions in the nucleus to alter
E-cadherin expression. We observed decreased Slug expression and
increased E-cadherin transcript expression on suppression of
Rac1b in HT29 cells. These data support an important nuclear
role for Rac1b, whereby it positively regulates Slug transcription,
which consequently represses E-cadherin expression leading to
diminished cellular adhesion. Interestingly, Slug has been reported
to be a Wnt target gene and shown to have Tcf-binding elements
within its promoter region (48). We propose that Rac1b expression
contributes to tumor progression by synergizing with Wnt
components to modulate Slug expression and therefore the
adhesive properties of cells. Future studies aimed at elucidating
how Rac1b regulates Slug and the discovery of other Rac1b/Wnt
target genes will further our understanding of the role of Rac1b in
tumor progression.
It is tempting to speculate that because Rac1b, being a tumor-
specific protein, seems to share only a selective repertoire of Rac1
functions, it is likely that those pathways retained by Rac1b are
critical for tumorigenesis, by perhaps imparting a growth advantage
and/or increased capacity for invasion and metastasis. In this regard,
further activation of Wnt signaling in colon cancer cells by Rac1b
may accelerate tumor progression in this tissue. Tumorigenesis is an
evolutionary process, where successive genetic changes transform a
normal cell into a self-sufficient, resilient ‘‘super’’ cancer cell capable
of metastasis. Mounting evidence suggests that deregulation of Rac1
signaling, through overexpression of itself or its regulators, has dire
consequences on cell physiology, changing signaling circuitry and
contributing to cellular transformation. Changes in Rac1 splicing to
create Rac1b, a much more efficient and active GTPase, likely
represents a further evolutionary advantage for cancer development.
Future studies aimed at identifying the mechanisms involved in
activation of Rac1b expression as well as clinical studies correlating
Rac1b expression with extent of colorectal cancer will increase our
understanding of Rac1b in tumorigenesis and may also be of
prognostic value or provide novel therapeutic targets.
Acknowledgments
Received 8/1/2006; revised 12/21/2006; accepted 1/11/2007.
Grant support: Canadian Institutes for Health Research MOP 64225 (B. Bapat), the
Ontario Graduate Scholarship (S. Esufali and P. Buongiorno), the Samuel Lunenfeld
Research Institute Ontario Student Opportunity Trust Funds Fellowship (S. Esufali and
P. Buongiorno), and the University of Toronto Open Fellowship (S. Esufali, G.S.
Charames, and P. Buongiorno).
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
We thank the following colleagues for their kind gifts of cDNA constructs: FLAG-
Rac1b (Dr. M. Ahmadian, Max-Planck Institute, Heidelberg, Germany); GFP-Rac1b (Dr.
C. Der, University of North Carolina, Chapel Hill, NC); W T h-catenin and FLAG–Dvl-3
(Dr. L. Attisano, University of Toronto, Ontario, Canada); CycD1-luc (Dr. T. Akiyama,
University of Tokyo, Tokyo, Japan); NF-nB–luc (Dr. A. Bosserhoff, University of
Regensburg, Regensburg, Germany); and pTOPFLASH and pFOPFLASH (Dr. B. Alman,
Hospital for Sick Children, Toronto, Ontario, Canada).
Cancer Research
Cancer Res 2007; 67: (6). March 15, 2007 2478 www.aacrjournals.org
References
1. Sahai E, Marshall CJ. RHO-GTPases and cancer. Nat
Rev Cancer 2002;2:133–42.
2. Boettner B, Van Aelst L. The role of Rho GTPases in
disease development. Gene 2002;286:155–74.
3. Price LS, Collard JG. Regulation of the cytoskeleton by
Rho-family GTPases: implications for tumour cell
invasion. Semin Cancer Biol 2001;11:167–73.
4. Bar-Sagi D, Hall A. Ras and Rho GTPases: a family
reunion. Cell 2000;103:227–38.
5. Gomez del Pulgar T, Benitah SA, Valeron PF, Espina C,
Lacal JC. Rho GTPase expression in tumourigenesis:
evidence for a significant link. BioEssays 2005;27:602–13.
6. Hill CS, Wynne J, Treisman R. The Rho family GTPases
RhoA, Rac1, and CDC42Hs regulate transcriptional
activation by SRF. Cell 1995;81:1159–70.
7. Welsh CF. Rho GTPases as key transducers of
proliferative signals in g1 cell cycle regulation. Breast
Cancer Res Treat 2004;84:33–42.
8. Perona R, Montaner S, Saniger L, Sanchez-Perez I,
Bravo R, Lacal JC. Activation of the nuclear factor-nBby
Rho, CDC42, and Rac-1 proteins. Genes Dev 1997;11:
463–75.
9. Coniglio SJ, Jou TS, Symons M. Rac1 protects epithelial
cells against anoikis. J Biol Chem 2001;276:28113–20.
10. Ruggieri R, Chuang YY, Symons M. The small GTPase
Rac suppresses apoptosis caused by serum deprivation
in fibroblasts. Mol Med 2001;7:293–300.
11. van Leeuwen FN, van der Kammen RA, Habets GG,
Collard JG. Oncogenic activity of Tiam1 and Rac1 in
NIH3T3 cells. Oncogene 1995;11:2215–21.
12. Westwick JK, Lambert QT, Clark GJ, et al. Rac
regulation of transformation, gene expression, and actin
organization by multiple, PAK-independent pathways.
Mol Cell Biol 1997;17:1324–35.
13. Khosravi-Far R, Solski PA, Clark GJ, Kinch MS, Der CJ.
Activation of Rac1, RhoA, and mitogen-activated protein
kinases is required for Ras transformation. Mol Cell Biol
1995;15:6443–53.
14. Qiu RG, Chen J, Kirn D, McCormick F, Symons M. An
essential role for Rac in Ras transformation. Nature
1995;374:457–9.
15. Schmitz AA, Govek EE, Bottner B, Van Aelst L. Rho
GTPases: signaling, migration, and invasion. Exp Cell
Res 2000;261:1–12.
16. Schnelzer A, Prechtel D, Knaus U, et al. Rac1 in
human breast cancer: overexpression, mutation analy-
sis, and characterization of a new isoform, Rac1b.
Oncogene 2000;19:3013–20.
17. Jordan P, Brazao R, Boavida MG, Gespach C, Chastre
E. Cloning of a novel human Rac1b splice variant with
increased expression in colorectal tumors. Oncogene
1999;18:6835–9.
18. Fiegen D, Haeusler LC, Blumenstein L, et al.
Alternative splicing of Rac1 generates Rac1b, a self-
activating GTPase. J Biol Chem 2004;279:4743–9.
19. MatosP,CollardJG,JordanP.Tumor-related
alternatively spliced Rac1b is not regulated by Rho-
GDP dissociation inhibitors and exhibits selective
downstream signaling. J Biol Chem 2003;278:50442–8.
20. Singh A, Karnoub AE, Palmby TR, Lengyel E, Sondek
J, Der CJ. Rac1b, a tumor associated, constitutively
active Rac1 splice variant, promotes cellular transfor-
mation. Oncogene 2004;23:9369–80.
21. Radisky DC, Levy DD, Littlepage LE, et al. Rac1b and
reactive oxygen species mediate MMP-3-induced EMT
and genomic instability. Nature 2005;436:123–7.
22. Vogelstein B, Kinzler KW. Cancer genes and the
pathways they control. Nat Med 2004;10:789–99.
23. Bienz M, Clevers H. Linking colorectal cancer to Wnt
signaling. Cell 2000;103:311–20.
24. Korinek V, Barker N, Morin PJ, et al. Constitutive
transcriptional activation by a h-catenin-Tcf complex in
APC
/
colon carcinoma. Science 1997;275:1784–7.
25. Morin PJ, Sparks AB, Korinek V, et al. Activation of h-
catenin-Tcf signaling in colon cancer by mutations in h-
catenin or APC. Science 1997;275:1787–90.
26. Esufali S, Bapat B. Cross-talk between Rac1 GTPase
and dysregulated Wnt signaling pathway lead s to
cellular redistribution of h-catenin and TCF/LEF-
mediated transcriptional activation. Oncogene 2004;23:
8260–71.
27. Minden A, Lin A, Claret FX, Abo A, Karin M. Selective
activation of the JNK signaling cascade and c-Jun
transcriptional activity by the small GTPases Rac and
Cdc42Hs. Cell 1995;81:1147–57.
28. Zhang S, Han J, Sells MA, et al. Rho family GTPases
regulate p38 mitogen-activated protein kinase through
the downstream mediator Pak1. J Biol Chem 1995;270:
23934–6.
29. Shtutman M, Zhurinsky J, Simcha I, et al. The cyclin
D1 gene is a target of the h-catenin/LEF-1 pathway.
Proc Natl Acad Sci U S A 1999;96:5522–7.
30. Tetsu O, McCormick F. h-catenin regulates expres-
sion of cyclin D1 in colon carcinoma cells. Nature 1999;
398:422–6.
31. Itoh K, Brott BK, Bae GU, Ratcliffe MJ, Sokol SY.
Nuclear localization is required for Dishevelled function
in Wnt/h-catenin signaling. J Biol 2005;4:3.
32. Lanning CC, Daddona JL, Ruiz-Velasco R, Shafer SH,
Williams CL. The Rac1 C-terminal polybasic region
regulates the nuclear localization and protein degrada-
tion of Rac1. J Biol Chem 2004;279:44197–210.
33. Williams CL. The polybasic region of Ras and Rho
family small GTPases: a regulator of protein inter-
actions and membrane association and a site of
nuclear localization signal sequences. Cell Signal 2003;
15:1071–80.
34. Lanning CC, Ruiz-Velasco R, Williams CL. Novel
mechanism of the co-regulation of nuclear transport of
SmgGDS and Rac1. J Biol Chem 2003;278:12495–506.
35. Ilyas M, Tomlinson IP, Rowan A, Pignatelli M,
Bodmer WF. h-catenin mutations in cell lines estab-
lished from human colorectal cancers. Proc Natl Acad
Sci U S A 1997;94:10330–4.
36. Kuphal F, Behrens J. E-cadherin modulates Wnt-
dependent transcription in colorectal cancer cells but
does not alter Wnt-independent gene expression in
fibroblasts. Exp Cell Res 2006;312:457–67.
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
Dishevelled-Mediated Activation of Rac1b
www.aacrjournals.org 2479 Cancer Res 2007; 67: (6). March 15, 2007
37. Thiery JP. Epith elial-mesenchymal transitions in
tumour progression. Nat Rev Cancer 2002;2:442–54.
38. Axelrod JD, Miller JR, Shulman JM, Moon RT, Perrimon
N. Differential recruitment of Dishevelle d provides
signaling specificity in the planar cell polarity and
Wingless signaling pathways. Genes Dev 1998;12:2610–22.
39. Capelluto DG, Kutateladze TG, Habas R, Finkielstein
CV, He X, Overduin M. The DIXdomain targets dishevelled
to actin stress fibres and vesicular membranes. Nature
2002;419:726–9.
40. Behrens J. The role of the Wnt signalling pathway
in colorectal tumorigenesis. Biochem Soc Trans 2005;
33:672–5.
41. Lustig B, Behrens J. The Wnt signaling pathway and
its role in tumor development. J Cancer Res Clin Oncol
2003;129:199–221.
42. Uematsu K, He B, You L, Xu Z, McCormick F, Jablons
DM. Activation of the Wnt pathway in non small cell
lung cancer: evidence of dishevelled overexpression.
Oncogene 2003;22:7218–21.
43. Uematsu K, Kanazawa S, You L, et al. Wnt pathway
activation in mesothelioma: evidence of Dishevelled
overexpression and transcriptional activity of h-catenin.
Cancer Res 2003;63:4547–51.
44. Okino K, Nagai H, Hatta M, et al. Up-regulation and
overproduction of DVL-1, the human counterpart of the
Drosophila Dishevelled gene, in cervical squamous cell
carcinoma. Oncol Rep 2003;10:1219–23.
45. Singh RR, Song C, Yang Z, Kumar R. Nuclear
localization and chromatin targets of p21-activated
kinase 1. J Biol Chem 2005;280:18130–7.
46. Perl AK, Wilgenbus P, Dahl U, Semb H, Christofori G.
A causal role for E-cadherin in the transition from
adenoma to carcinoma. Nature 1998;392:190–3.
47. Birchmeier W, Behrens J. Cadherin expression in
carcinomas: role in the formation of cell junctions and
the prevention of invasiveness. Biochim Biophys Acta
1994;1198:11–26.
48. Vallin J, Thuret R, Giacomello E, Faraldo MM, Thiery
JP, Broders F. Cloning and characterization of three
Xenopus slug promoters reveal direct regulation by Lef/
h-catenin signaling. J Biol Chem 2001;276:30350–8.
Research.
on June 12, 2013. © 2007 American Association for Cancercancerres.aacrjournals.org Downloaded from
... Likewise, RAC1B is necessary for EMT-bypass of senescence in lung adenocarcinoma [20]. Other suggested mechanisms driven by RAC1B to promote invasion and EMT include downregulation of E-cadherin and/or interaction with the GAP protein ArhGAP11A [21,22]. Emerging in vivo evidence indicates an oncogenic role of RAC1B. ...
... There are a number of mechanistic reasons that could explain the increased tumourigenesis seen in both models. For example, RAC1B might enhance epithelial proliferation through activation of the Wnt signalling pathway [17,21]. However, overexpression of RAC1B did not increase the number of proliferative cells in the normal small intestine nor in the early adenomas (Figs. ...
... Therefore, it appears that suppression of TGFβ signalling is a key downstream effect of RAC1B signalling and whether this leads to tumour promoting or suppressing effects is dependent on the context of this overexpression. In the context of colorectal cancer, the building evidence from the literature [14,[19][20][21][22]24] along with our in vivo mouse experiments and the analysis of human cancer expression data [32] suggests that RAC1B acts as a tumour promoting factor. ...
Article
Full-text available
RAC1B is a tumour-related alternative splice isoform of the small GTPase RAC1, found overexpressed in a large number of tumour types. Building evidence suggests it promotes tumour progression but compelling in vivo evidence, demonstrating a role in driving tumour invasion, is currently lacking. In the present study, we have overexpressed RAC1B in a colorectal cancer mouse model with potential invasive properties. Interestingly, RAC1B overexpression did not trigger tumour invasion, rather it led to an acceleration of tumour initiation and reduced mouse survival. By modelling early stages of adenoma initiation we observed a reduced apoptotic rate in RAC1B overexpressing tumours, suggesting protection from apoptosis as a mediator of this phenotype. RAC1B overexpressing tumours displayed attenuated TGFβ signalling and functional analysis in ex vivo organoid cultures demonstrated that RAC1B negatively modulates TGFβ signalling and confers resistance to TGFβ-driven cell death. This work defines a novel mechanism by which early adenoma cells can overcome the cytostatic and cytotoxic effects of TGFβ signalling and characterises a new oncogenic function of RAC1B in vivo.
... The extra 19-amino acid sequence, however, endows RAC1B with novel functions as a consequence of enhanced binding to proteins involved in cell-cell adhesion, motility, and transcriptional regulation such as SmgGDS, RACK1, and p120 catenin [22]. RAC1B has also been reported to interact with Dishevelled-3 and to form a tetramer with β-catenin/TCF that is recruited to the promoter of canonical Wnt target genes [23]. As a consequence of impaired binding to Rho-GDI, RAC1B is primarily localized at the inner leaflet of the plasma membrane, which brings it into contact with specific interaction partners or substrates, especially membrane receptors and their signalosomes, i.e., those for transforming growth factor β (TGFβ). ...
... It is well documented that a portion of both RAC1 [105] and RAC1B [23] is localized to the nucleus; however, very little is known regarding the function and biological significance of nuclear RAC1 or RAC1B. In the nucleus, both isoforms can interact with a very different set of proteins and may, therefore, display novel functional activities. ...
Article
Full-text available
Simple Summary Transforming growth factor β (TGFβ) promotes pancreatic ductal adenocarcinoma (PDAC) primarily through its non-canonical (non-Smad) signaling arms, including signaling by the small GTPase RAC1. The human RAC1 gene also encodes for another protein, designated RAC1B, but whether this isoform also interacts with TGFβ signaling has remained unknown. In a series of studies in PDAC-derived cells, we found that RAC1B also cross-talks with TGFβ signaling, but unlike RAC1 antagonizes TGFβ-induced responses, i.e., epithelial–mesenchymal transition, through multiple mechanisms. However, rather than being uniformly inhibitory, RAC1B selectively blocks tumor-promoting pathways, while concomitantly allowing tumor-suppressive pathways to proceed. In this review article, we discuss the specific interactions between RAC1B and TGFβ signaling, which occur at multiple levels and include various components of both the canonical Smad and non-Smad pathways. In addition to emerging as a novel tumor suppressor in PDAC, RAC1B turned out to be a useful tool to dissect TGFβ signaling. Abstract RAC1 and its alternatively spliced isoform, RAC1B, are members of the Rho family of GTPases. Both isoforms are involved in the regulation of actin cytoskeleton remodeling, cell motility, cell proliferation, and epithelial–mesenchymal transition (EMT). Compared to RAC1, RAC1B exhibits a number of distinctive features with respect to tissue distribution, downstream signaling and a role in disease conditions like inflammation and cancer. The subcellular locations and interaction partners of RAC1 and RAC1B vary depending on their activation state, which makes RAC1 and RAC1B ideal candidates to establish cross-talk with cancer-associated signaling pathways—for instance, interactions with signaling by transforming growth factor β (TGFβ), a known tumor promoter. Although RAC1 has been found to promote TGFβ-driven tumor progression, recent observations in pancreatic carcinoma cells surprisingly revealed that RAC1B confers anti-oncogenic properties, i.e., through inhibiting TGFβ-induced EMT. Since then, an unexpected array of mechanisms through which RAC1B cross-talks with TGFβ signaling has been demonstrated. However, rather than being uniformly inhibitory, RAC1B interacts with TGFβ signaling in a way that results in the selective blockade of tumor-promoting pathways, while concomitantly allowing tumor-suppressive pathways to proceed. In this review article, we are going to discuss the specific interactions between RAC1B and TGFβ signaling, which occur at multiple levels and include various components such as ligands, receptors, cytosolic mediators, transcription factors, and extracellular inhibitors of TGFβ ligands.
... It was indicated as an important stimulant in transcription and decreased adhesion of breast and colon cancer cells. Its overexpression results in increased TCF (T-cell factor)-mediated gene transcription, whereas suppression results in decreased expression of the Wnt target gene cyclin D, which is important in the regulation of cell cycle transitions and migration of macrophages [83,84]. Rac1b as a constitutively active form of Rac1 is therefore relevant in pathogenesis involving macrophage Rac signaling, such as in the case of atherosclerosis. ...
Article
Full-text available
Coronary artery disease caused by atherosclerosis is a major cause of morbidity and mortality around the world. Data from preclinical and clinical studies support the belief that atherosclerosis is an inflammatory disease that is mediated by innate and adaptive immune signaling mechanisms. This review sought to highlight the role of Rac-mediated inflammatory signaling in the mechanisms driving atherosclerotic calcification. In addition, current clinical treatment strategies that are related to targeting hypercholesterolemia as a critical risk factor for atherosclerotic vascular disease are addressed in relation to the effects on Rac immune signaling and the implications for the future of targeting immune responses in the treatment of calcific atherosclerosis.
... RAC1B results from the inclusion of exon 4 (alternatively designated exon 3b) encoding an additional 19 amino acids which leads to constitutive activation [22][23][24] . It is believed that RAC1B has a distinctive, more restricted set of effector pathways than RAC1, but appears to be more critical for tissue transformation [25][26][27][28][29] . Despite some in vitro evidence supporting a tumorigenic role for RAC1B, its in vivo function and mechanism of action is poorly understood and to date no studies have assessed whether RAC1B is required for tumorigenesis in vivo and thus the potential benefits of its therapeutic targeting. ...
Article
Full-text available
Current therapeutic options for treating colorectal cancer have little clinical efficacy and acquired resistance during treatment is common, even following patient stratification. Understanding the mechanisms that promote therapy resistance may lead to the development of novel therapeutic options that complement existing treatments and improve patient outcome. Here, we identify RAC1B as an important mediator of colorectal tumourigenesis and a potential target for enhancing the efficacy of EGFR inhibitor treatment. We find that high RAC1B expression in human colorectal cancer is associated with aggressive disease and poor prognosis and deletion of Rac1b in a mouse colorectal cancer model reduces tumourigenesis. We demonstrate that RAC1B interacts with, and is required for efficient activation of the EGFR signalling pathway. Moreover, RAC1B inhibition sensitises cetuximab resistant human tumour organoids to the effects of EGFR inhibition, outlining a potential therapeutic target for improving the clinical efficacy of EGFR inhibitors in colorectal cancer.
... RAC1B has been reported to be localized in the nucleus [33], a finding that would principally be compatible with a role as a transcription factor and, hence, direct (inhibitory) transcriptional effects of RAC1B on the TGFBR1 promoter. However, reporter gene assays showed that knockdown of RAC1B failed to alter transcriptional activity of a cotransfected reporter construct encompassing 392 base pairs upstream of the transcriptional start site in the TGFBR1 promoter. ...
Article
Full-text available
The small GTPase RAC1B has been shown to act as a powerful inhibitor of the transforming growth factor (TGF)β type I receptor ALK5 and TGFβ1/ALK5-induced epithelial–mesenchymal transition and cell motility. However, the precise mechanism has remained elusive. RNAi-mediated knockdown of RAC1B in the pancreatic ductal adenocarcinoma (PDAC)-derived cell line Panc1 failed to alter transcriptional activity from a transfected ALK5 promoter–reporter construct. In contrast, pharmacological inhibition of the proteasome decreased the abundance of ALK5 protein in cell lines of the mesenchymal subtype (Panc1, IMIM-PC-1, and breast cancer MDA-MB-231), but not in a PDAC cell line of the epithelial subtype (Colo357). Here, we focused on the inhibitory Smad protein, SMAD7, as a potential candidate for RAC1B-mediated inhibition of cell migration. In Panc1 cells devoid of RAC1B, SMAD7 protein was dramatically reduced and these cells were refractory to TGFβ1-induced upregulation of SMAD7 protein but not mRNA expression. Intriguingly, RNAi-mediated knockdown or ectopic overexpression of SMAD7 in Panc1 cells up- or downregulated, respectively, ALK5 protein expression and mimicked the suppressive effect of RAC1B on TGFβ/SMAD3-dependent transcriptional activity, target gene expression and cell migration. Transfection of SMAD7 was further able to partially rescue cells from the RAC1B knockdown-mediated increase in migratory properties. Conversely, knockdown of SMAD7 was able to partially rescue Panc1 and MDA-MB-231 cells from the antimigratory effect of ectopically expressed RAC1B. Finally, we demonstrate that RAC1B upregulation of SMAD7 protein requires intermittent transcriptional induction of the deubiquitinating enzyme USP26. Our data suggest that RAC1B induces SMAD7 by promoting its deubiquitination and establishes this Smad as one of RAC1B’s downstream effectors in negative regulation of ALK5 and TGFβ1-induced cell migration in mesenchymal-type carcinoma cells.
... In this connection, the Rac1b-dependent motility and spreading of the mouse mammary epithelial SCp2 cells proved to require p120 ctn . Rac1b has also been reported to interact with Dishevelled-3 and to form a tetramer with β-catenin/TCF that is recruited to the promoter of canonical Wnt target genes [112]. In this connection, the expression of the Rac1b splice variant in intestinal epithelial cells from transgenic mice is associated with an increase in the number of β-catenin nuclear positive cells at the bottom of the crypts [14]. ...
Article
Full-text available
The small GTPase Rac1 has been implicated in a variety of dynamic cell biological processes, including cell proliferation, cell survival, cell-cell contacts, epithelial mesenchymal transition (EMT), cell motility, and invasiveness. These processes are orchestrated through the fine tuning of Rac1 activity by upstream cell surface receptors and effectors that regulate the cycling Rac1-GDP (off state)/Rac1-GTP (on state), but also through the tuning of Rac1 accumulation, activity, and subcellular localization by post translational modifications or recruitment into molecular scaffolds. Another level of regulation involves Rac1 transcripts stability and splicing. Downstream, Rac1 initiates a series of signaling networks, including regulatory complex of actin cytoskeleton remodeling, activation of protein kinases (PAKs, MAPKs) and transcription factors (NFkB, Wnt/β-catenin/TCF, STAT3, Snail), production of reactive oxygen species (NADPH oxidase holoenzymes, mitochondrial ROS). Thus, this GTPase, its regulators, and effector systems might be involved at different steps of the neoplastic progression from dysplasia to the metastatic cascade. After briefly placing Rac1 and its effector systems in the more general context of intestinal homeostasis and in wound healing after intestinal injury, the present review mainly focuses on the several levels of Rac1 signaling pathway dysregulation in colorectal carcinogenesis, their biological significance, and their clinical impact.
... A recent publication by Li et al. supports the role of Rac1b in proliferation and inhibition of apoptosis by the activation of JNK2 (Li et al., 2016). However, earlier studies emphasize that Rac1b did not activate p21 protein kinase nor its downstream protein kinase JNK (Matos et al., 2003;Esufali et al., 2007). An interesting difference between Rac1 and Rac1b, not analyzed in this study, has been described regarding their sensitivity toward ubiquitinylation which is involved in regulation of protein stability (Visvikis et al., 2008). ...
Article
Full-text available
Rac1 is a ubiquitously expressed Rho GTPase and an important regulator of the actin cy-toskeleton. Its splice variant Rac1b exhibits a 19aa in-frame insertion and is predominantly active. Both proteins were described in tumorigenesis or metastasis. We investigated the contribution of Rac1 and Rac1b to tumor progression of human non-small-cell-lungadenocarcinoma (NSCLA). Rac1 protein was present in 8/8 NSCLA cell lines analyzed, whereas Rac1b was expressed in only 6/8. In wound healing assays, EGFP-Rac1 slightly de-creased cell migration, whereas proliferation was increased in both, Rac1- and Rac1bexpressing cells. In the in vivo chorioallantoic invasion model, EGFP-Rac1-expressing cells formed more invasive tumors compared to EGFP-Rac1b. This increased invasiveness correlated with enhanced phosphorylation of p38α, AKT, and GSK3β and activation of serum response- and Smad-dependent gene promoters by Rac1. In contrast, Rac1b solely activated the MAP kinase JNK2, together with TCF/LEF1- and NFκB-responsive gene reporters. Rac1b, as Rac1 phosphorylated p38α, AKT, and GSK3β. Knockdown of the splicing factor ESRP1, which mediates out-splicing of exon 3b from RAC1, resulted in increased Rac1b mRNA and suppression of the EMT-associated transcription factor ZEB1. Our data demonstrate different signaling and functional activities of Rac1 and Rac1b and an important role for Rac1 in lung cancer metastasis.
Article
Full-text available
Background The incidence of colorectal cancer (CRC) has been steadily increasing in younger individuals over the past several decades for reasons that are incompletely defined. Identifying differences in gene expression profiles, or transcriptomes, in early-onset colorectal cancer (EOCRC, < 50 years old) patients versus later-onset colorectal cancer (LOCRC, > 50 years old) patients is one approach to understanding molecular and genetic features that distinguish EOCRC. Methods We performed RNA-sequencing (RNA-seq) to characterize the transcriptomes of patient-matched tumors and adjacent, uninvolved (normal) colonic segments from EOCRC (n=21) and LOCRC (n=22) patients. The EOCRC and LOCRC cohorts were matched for demographic and clinical characteristics. We used The Cancer Genome Atlas Colon Adenocarcinoma (TCGA-COAD) database for validation. We used a series of computational and bioinformatic tools to identify EOCRC-specific differentially expressed genes, molecular pathways, predicted cell populations, differential gene splicing events, and predicted neoantigens. Results We identified an eight-gene signature in EOCRC comprised of ALDOB, FBXL16, IL1RN, MSLN, RAC3, SLC38A11, WBSCR27 and WNT11, from which we developed a score predictive of overall CRC patient survival. On the entire set of genes identified in normal tissues and tumors, cell type deconvolution analysis predicted a differential abundance of immune and non-immune populations in EOCRC versus LOCRC. Gene set enrichment analysis identified increased expression of splicing machinery in EOCRC. We further found differences in alternative splicing (AS) events, including one within the long non-coding RNA, HOTAIRM1. Additional analysis of AS found seven events specific to EOCRC that encode potential neoantigens. Conclusion Our transcriptome analyses identified genetic and molecular features specific to EOCRC which may inform future screening, development of prognostic indicators, and novel drug targets.
Article
Alternative splicing (AS) is a critical mechanism for the aberrant biogenesis of long non-coding RNA (lncRNA). Although the role of Wnt signaling in AS has been implicated, it remains unclear how it mediates lncRNA splicing during cancer progression. Herein, we identify that Wnt3a induces a splicing switch of lncRNA-DGCR5 to generate a short variant (DGCR5-S) that correlates with poor prognosis in esophageal squamous cell carcinoma (ESCC). Upon Wnt3a stimulation, active nuclear β-catenin acts as a co-factor of FUS to facilitate the spliceosome assembly and the generation of DGCR5-S. DGCR5-S inhibits TTP's anti-inflammatory activity by protecting it from PP2A-mediated dephosphorylation, thus fostering tumor-promoting inflammation. Importantly, synthetic splice-switching oligonucleotides (SSOs) disrupt the splicing switch of DGCR5 and potently suppress ESCC tumor growth. These findings uncover the mechanism for Wnt signaling in lncRNA splicing and suggest that the DGCR5 splicing switch may be a targetable vulnerability in ESCC.
Article
Identifying events that regulate the prenylation and localization of small GTPases will help define new strategies for therapeutic targeting of these proteins in disorders such as cancer, cardiovascular disease, and neurological deficits. Splice variants of the chaperone protein SmgGDS (encoded by RAP1GDS1) are known to regulate prenylation and trafficking of small GTPases. The SmgGDS-607 splice variant regulates prenylation by binding pre-prenylated small GTPases, but the effects of SmgGDS binding to the small GTPase RAC1 versus the splice variant RAC1B are not well defined. Here we report unexpected differences in the prenylation and localization of RAC1 and RAC1B, and their binding to SmgGDS. Compared to RAC1, RAC1B more stably associates with SmgGDS-607, is less prenylated, and accumulates more in the nucleus. We show that the small GTPase DIRAS1 inhibits binding of RAC1 and RAC1B to SmgGDS and reduces their prenylation. These results suggest that prenylation of RAC1 and RAC1B is facilitated by binding to SmgGDS-607, but the greater retention of RAC1B by SmgGDS-607 slows RAC1B prenylation. We show that inhibiting RAC1 prenylation by mutating the CAAX motif promotes RAC1 nuclear accumulation, suggesting that differences in prenylation contribute to the different nuclear localization of RAC1 versus RAC1B. Finally, we demonstrate RAC1 and RAC1B that cannot be prenylated bind GTP in cells, indicating that prenylation is not a prerequisite for activation. We report differential expression of RAC1 and RAC1B transcripts in tissues, consistent with these two splice variants having unique functions that might arise in part from their differences in prenylation and localization.
Article
Full-text available
Development of malignant tumours is in part characterized by the ability of a tumour cell to overcome cell-cell adhesion and to invade surrounding tissue. E-cadherin is the main adhesion molecule of epithelia, and it has been implicated in carcinogenesis because it is frequently lost in human epithelial cancers. Re-establishing the functional cadherin complex in tumour cell lines results in a reversion from an invasive to a benign epithelial phenotype. However, it remained unresolved whether the loss of E-cadherin-mediated cell adhesion was a cause or a consequence of tumour progression in vivo. Here we report that the loss of E-cadherin expression coincides with the transition from well differentiated adenoma to invasive carcinoma in a transgenic mouse model of pancreatic beta-cell carcinogenesis (Rip1Tag2). Intercrossing Rip1Tag2 mice with transgenic mice that maintain E-cadherin expression in beta-tumour cells results in arrest of tumour development at the adenoma stage, whereas expression of a dominant-negative form of E-cadherin induces early invasion and metastasis. The results demonstrate that loss of E-cadherin-mediated cell adhesion is one rate-limiting step in the progression from adenoma to carcinoma.
Article
Full-text available
Mutations in the adenomatous polyposis coli (APC) tumour-suppressor gene occur in most human colon cancers. Loss of functional APC protein results in the accumulation of beta-catenin. Mutant forms of beta-catenin have been discovered in colon cancers that retain wild-type APC genes, and also in melanomas, medulloblastomas, prostate cancer and gastric and hepatocellular carcinomas. The accumulation of beta-catenin activates genes that are responsive to transcription factors of the TCF/LEF family, with which beta-catenin interacts. Here we show that beta-catenin activates transcription from the cyclin D1 promoter, and that sequences within the promoter that are related to consensus TCF/LEF-binding sites are necessary for activation. The oncoprotein p21ras further activates transcription of the cyclin D1 gene, through sites within the promoter that bind the transcriptional regulators Ets or CREB. Cells expressing mutant beta-catenin produce high levels of cyclin D1 messenger RNA and protein constitutively. Furthermore, expression of a dominant-negative form of TCF in colon-cancer cells strongly inhibits expression of cyclin D1 without affecting expression of cyclin D2, cyclin E, or cyclin-dependent kinases 2, 4 or 6. This dominant-negative TCF causes cells to arrest in the G1 phase of the cell cycle; this phenotype can be rescued by expression of cyclin D1 under the cytomegalovirus promoter. Abnormal levels of beta-catenin may therefore contribute to neoplastic transformation by causing accumulation of cyclin D1.
Article
Full-text available
β-Catenin plays a dual role in the cell: one in linking the cytoplasmic side of cadherin-mediated cell–cell contacts to the actin cytoskeleton and an additional role in signaling that involves transactivation in complex with transcription factors of the lymphoid enhancing factor (LEF-1) family. Elevated β-catenin levels in colorectal cancer caused by mutations in β-catenin or by the adenomatous polyposis coli molecule, which regulates β-catenin degradation, result in the binding of β-catenin to LEF-1 and increased transcriptional activation of mostly unknown target genes. Here, we show that the cyclin D1 gene is a direct target for transactivation by the β-catenin/LEF-1 pathway through a LEF-1 binding site in the cyclin D1 promoter. Inhibitors of β-catenin activation, wild-type adenomatous polyposis coli, axin, and the cytoplasmic tail of cadherin suppressed cyclin D1 promoter activity in colon cancer cells. Cyclin D1 protein levels were induced by β-catenin overexpression and reduced in cells overexpressing the cadherin cytoplasmic domain. Increased β-catenin levels may thus promote neoplastic conversion by triggering cyclin D1 gene expression and, consequently, uncontrolled progression into the cell cycle.
Article
Full-text available
Although substantial evidence supports a critical role for the activation of Raf-1 and mitogen-activated protein kinases (MAPKs) in oncogenic Ras-mediated transformation, recent evidence suggests that Ras may activate a second signaling pathway which involves the Ras-related proteins Rac1 and RhoA. Consequently, we used three complementary approaches to determine the contribution of Rac1 and RhoA function to oncogenic Ras-mediated transformation. First, whereas constitutively activated mutants of Rac1 and RhoA showed very weak transforming activity when transfected alone, their coexpression with a weakly transforming Raf-1 mutant caused a greater than 35-fold enhancement of transforming activity. Second, we observed that coexpression of dominant negative mutants of Rac1 and RhoA reduced oncogenic Ras transforming activity. Third, activated Rac1 and RhoA further enhanced oncogenic Ras-triggered morphologic transformation, as well as growth in soft agar and cell motility. Finally, we also observed that kinase-deficient MAPKs inhibited Ras transformation. Taken together, these data support the possibility that oncogenic Ras activation of Rac1 and RhoA, coupled with activation of the Raf/MAPK pathway, is required to trigger the full morphogenic and mitogenic consequences of oncogenic Ras transformation.
Article
Full-text available
The stress-activated p38 mitogen-activated protein (MAP) kinase defines a subgroup of the mammalian MAP kinases that appear to play a key role in regulating inflammatory responses. Co-expression of constitutively active forms of Rac and Cdc42 leads to activation of p38 while dominant negative Rac and Cdc42 inhibit the ability of interleukin-1 to increase p38 activity. p21-activated kinase 1 (Pak1) is a potential mediator of Rac/Cdc42 signaling, and we observe that Pak1 stimulates p38 activity. A dominant negative Pak1 suppresses both interleukin-1- and Rac/Cdc42-induced p38 activity. Rac and Cdc42 appear to regulate a protein kinase cascade initiated at the level of Pak and leading to activation of p38 and JNK.
Article
Rac1 is a member of the Ras superfamily of small guanosine triphosphatases (GTPases) that act as molecular switches to control cytoskeletal rearrangements and cell growth. Analogous to Ras, constitutively activating point mutations of Rac1 cause tumorigenic transformation of cell lines. However, there is no information about whether Rac1 is also mutated in vivo. After RT–PCR of Rac1, several clones of seven benign and 10 malignant breast cancer tissues as well as eight breast cancer cell lines were sequenced. Only single-nucleotide polymorphisms of Rac1 could be detected, and none of these corresponded to constitutively activating point mutations that have been used in cell lines for transformation. While sequencing Rac1 in breast tissues, a new Rac1 isoform with an insertion of 19 codons within the reading frame of Rac1 close to switch region II was identified and named Rac1b. The Rac1b protein acts like a fast cycling GTPase in GTP binding and hydrolysis assays. In Northern and Western blot experiments both Rac1 RNA and Rac1 protein had a significantly higher expression in breast cancer tissues compared to normal breast tissue samples. Immunohistochemical staining of Rac1 showed weak Rac1 expression in benign breast disease but high expression level in ductal carcinoma-in-situ, primary breast cancer, and lymph node metastases. In addition, breast tumor cells from patients with recurrent disease had Rac1 expression at the plasma membrane, suggesting activation of Rac1, in patients with aggressive breast cancer.
Article
Without epithelial-mesenchymal transitions, in which polarized epithelial cells are converted into motile cells, multicellular organisms would be incapable of getting past the blastula stage of embryonic development. However, this important developmental programme has a more sinister role in tumor progression. Epithelial-mesenchymal transition provides a new basis for understanding the progression of carcinoma towards dedifferentiated and more malignant states.
Article
Many small GTPases in the Ras and Rho families have a C-terminal polybasic region (PBR) comprised of multiple lysines or arginines. The PBR controls diverse functions of these small GTPases, including their ability to associate with membranes, interact with specific proteins, and localize in subcellular compartments. Different signaling pathways mediated by Ras and Rho family members may converge when the small GTPases are directed by their PBRs to shared binding sites in specific proteins or at cell membranes. The PBR promotes the interactions of small GTPases with SmgGDS, which is a nucleocytoplasmic shuttling protein that stimulates guanine nucleotide exchange by small GTPases. The PBR of Rac1 was recently found to have a functional nuclear localization signal (NLS) sequence, which enhances the nuclear accumulation of protein complexes containing SmgGDS and Rac1. Sequence analysis demonstrates that canonical NLS sequences (K-K/R-x-K/R) are present in the PBRs of additional Ras and Rho family members, and are evolutionarily conserved across several phyla. These findings suggest that the PBR regulates the nucleocytoplasmic shuttling of some Ras and Rho family members when they are in protein complexes that are too large to diffuse through nuclear pores. These diverse functions of the PBR indicate its critical role in signaling by Ras and Rho family GTPases.
Article
The c-fos serum response element (SRE) forms a ternary complex with the transcription factors SRF (serum response factor) and TCF (ternary complex factor). By itself, SRF can mediate transcriptional activation induced by serum, lysophosphatidic acid, or intracellular activation of heterotrimeric G proteins. Activated forms of the Rho family GTPases RhoA, Rac1, and CDC42Hs also activate transcription via SRF and act synergistically at the SRE with signals that activate TCF. Functional Rho is required for signaling to SRF by several stimuli, but not by activated CDC42Hs or Rac1. Activation of the SRF-linked signaling pathway does not correlate with activation of the MAP kinases ERK, SAPK/JNK, or MPK2/p38. Functional Rho is required for regulated activity of the c-fos promoter. These results establish SRF as a nuclear target of a novel Rho-mediated signaling pathway.