ArticlePDF Available

Abstract and Figures

Pancreatic ductal adenocarcinoma (PDAC) is the eighth largest cause of cancer-related mortality across the world, with a median 5-year survival rate of less than 3.5%. This is partly because the molecules and the molecular mechanisms that contribute to PDAC are not well understood. Our goal is to understand the role of p21-activated kinase 1 (Pak1) signaling axis in the progression of PDAC. Pak1, a serine/threonine kinase, is a well-known regulator of cytoskeletal remodeling, cell motility, cell proliferation and cell survival. Recent reports suggest that Pak1 by itself can have an oncogenic role in a wide variety of cancers. In this study, we analyzed the expression of Pak1 in human pancreatic cancer tissues and found that Pak1 levels are significantly upregulated in PDAC samples as compared with adjacent normals. Further, to study the functional role of Pak1 in pancreatic cancer model systems, we developed stable overexpression and lentiviral short hairpin RNA-mediated knockdown (KD) clones of Pak1 and studied the changes in transforming properties of the cells. We also observed that Pak1 KD clones failed to form tumors in nude mice. By adopting a quantitative PCR array-based approach, we identified fibronectin, a component of the extracellular matrix and a mesenchymal marker, as a transcriptional target of Pak1 signaling. The underlying molecular mechanism of Pak1-mediated transformation includes its nuclear import and recruitment to the fibronectin promoter via interaction with nuclear factor-κB (NF-κB)-p65 complex. To our knowledge, this is the first study illustrating Pak1-NF-κB-p65-mediated fibronectin regulation as a potent tumor-promoting mechanism in KRAS intact model.Oncogene advance online publication, 24 February 2014; doi:10.1038/onc.2013.576.
Content may be subject to copyright.
ORIGINAL ARTICLE
Transcriptional regulation of fibronectin by p21-activated kinase-1
modulates pancreatic tumorigenesis
S Jagadeeshan
1,6
, YR Krishnamoorthy
1,6
, M Singhal
1,6
, A Subramanian
1,6
, J Mavuluri
1
, A Lakshmi
1
, A Roshini
1
, G Baskar
2
,
M Ravi
2
, LD Joseph
3
, K Sadasivan
4
, A Krishnan
5
, AS Nair
5
, G Venkatraman
2
and SK Rayala
1
Pancreatic ductal adenocarcinoma (PDAC) is the eighth largest cause of cancer-related mortality across the world, with a median
5-year survival rate of less than 3.5%. This is partly because the molecules and the molecular mechanisms that contribute to PDAC
are not well understood. Our goal is to understand the role of p21-activated kinase 1 (Pak1) signaling axis in the progression of
PDAC. Pak1, a serine/threonine kinase, is a well-known regulator of cytoskeletal remodeling, cell motility, cell proliferation and cell
survival. Recent reports suggest that Pak1 by itself can have an oncogenic role in a wide variety of cancers. In this study, we
analyzed the expression of Pak1 in human pancreatic cancer tissues and found that Pak1 levels are significantly upregulated in
PDAC samples as compared with adjacent normals. Further, to study the functional role of Pak1 in pancreatic cancer model
systems, we developed stable overexpression and lentiviral short hairpin RNA-mediated knockdown (KD) clones of Pak1 and
studied the changes in transforming properties of the cells. We also observed that Pak1 KD clones failed to form tumors in nude
mice. By adopting a quantitative PCR array-based approach, we identified fibronectin, a component of the extracellular matrix and a
mesenchymal marker, as a transcriptional target of Pak1 signaling. The underlying molecular mechanism of Pak1-mediated
transformation includes its nuclear import and recruitment to the fibronectin promoter via interaction with nuclear factor-kB
(NF-kB)–p65 complex. To our knowledge, this is the first study illustrating Pak1–NF-kB–p65-mediated fibronectin regulation as a
potent tumor-promoting mechanism in KRAS intact model.
Oncogene advance online publication, 24 February 2014; doi:10.1038/onc.2013.576
Keywords: Pak1; fibronectin; PDAC; NF-kB–p65; KRAS intact model
INTRODUCTION
Pancreatic cancer is the thirteenth most common type of cancer
worldwide. More than 95% of pancreatic cancers are ductal
adenocarcinomas and remain as one of the most devastating
malignancies.
1,2
Owing to the lack of symptoms during early
stages, most patients present with the disease at an advanced
stage during diagnosis, which is difficult to treat and manage.
Therefore, the 5-year survival rates are very low and are estimated
to be less than 3.5%.
3
In terms of clinically effective therapeutic
options, nothing much has changed significantly over the past
few years.
4,5
Our limited understanding of the pathobiology of
pancreatic ductal adenocarcinoma (PDAC) has been a barrier to
progress in developing suitable therapeutic strategies. Research
till date on PDAC has concentrated on understanding the basic
causative factors inducing tumorigenesis. Hence, there is an
increasing need to understand the novel signaling pathways that
contribute to the progression of PDAC, which might eventually
translate into potential therapeutic models of intervention.
It is accepted that key genes involved in pancreatic cancer
progression encode proteins that function as critical regulators of
signal–transduction networks that regulate cell proliferation,
differentiation and survival.
6
Studies have demonstrated that
several signaling cascades important for normal pancreatic
development are also dysregulated during the proliferation,
survival and metastasis of pancreatic ductal adenocarcinoma
cells.
7–9
Despite an in-depth research on various signaling
pathways involved in transforming properties that have been
implicated in the development of PDACs, treatments targeting
these pathways have proven to be ineffective. More than 90% of
pancreatic cancers carry oncogenic KRAS mutations at codon
12,
10,11
which cause a constitutive activation of the G protein RAS,
eventually leading to abnormal activation of various downstream
signaling pathways involving p21-activated kinase 1 (Pak1). Pak1
is a serine/threonine kinase that functions as a downstream
activator for various oncogenic signaling pathways. Pak1 is a well-
known regulator of cytoskeletal remodeling, cell proliferation, cell
survival and cell motility, all of which result in tumor formation
and cell invasiveness.
12,13
Alterations in Pak1 expression have
been detected in a wide variety of human tumors.
14
Pak1 was
shown to regulate cyclin D1 levels through the activation of
nuclear factor (NF)-kB and thus contributing to increased tumor
cell proliferation.
15
The hematopoietic-specific guanine nucleotide
exchange factor VAV1 reportedly contributes to tumorigenic
properties of pancreatic cancer cells through the regulation of
signaling axis involving Pak1.
16
The upstream regulators of Pak
family members, Rac1 and Cdc42, have been previously
1
Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, India;
2
Department of Human Genetics, Sri Ramachandra University, Chennai, India;
3
Department of Pathology, Sri Ramachandra University, Chennai, India;
4
Department of Plastic and Reconstructive Surgery, Government Medical College, Thiruvananthapuram,
India and
5
Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananth apuram, India. Correspondence: Dr SK Rayala, Department of Biotechnology, IIT Madras, Chennai 600036,
India or Dr G Venkatraman, Department of Human Genetics, Sri Ramachandra University, Porur, Chennai 600116, India or Dr AS Nair, Rajiv Gandhi Centre for Biotechnology
(RGCB), Thycaud Post, Poojappura, Thiruvananthapuram 695014, Kerala, India.
E-mail: rayala@iitm.ac.in or ganeshv@sriramachandra .edu.in or sasha@rgcb.res.in
6
These authors contributed equally to this work.
Received 10 June 2013; revised 3 December 2013; accepted 7 December 2013
Oncogene (2014), 110
&
2014 Macmillan Publishers Limited All rights reserved 0950-9232/14
www.nature.com/onc
implicated in pancreatic cancer initiation and progression.
17,18
Rac1 has been shown as a mediator of transforming growth
factor-b-1-induced effects on cell migration and Smad signaling in
pancreatic cancer cell lines.
19
Among the several important
therapeutic targets discovered so far for PDAC, all the receptor
tyrosine kinases, growth factor receptors, gastrin receptors and Src
kinases are known to activate Pak1 kinase.
20
Cox-2 and b-catenin
have been identified as downstream targets of Pak1 signaling.
21,22
Thus, Pak1 seems to be significantly involved in the multiple
signaling pathways dysregulated or hyperactivated in PDACs. This
study focuses on understanding the biology of Pak1 and its
contribution to the etiology of pancreatic cancer.
RESULTS
Clinical significance of Pak1
Pak1 expression is dysregulated in PDAC tissues and cell lines.In
order to study the role of Pak1 in PDAC, we initially analyzed the
expression of Pak1 protein in 14 paired samples of human PDAC
tumors and adjacent normal pancreatic tissue by western blotting,
and found that Pak1 levels are significantly upregulated in 9 out of
14 tumors as compared with that in normal tissue (Figure 1a). We
further analyzed the Pak1 mRNA levels in human PDAC tissue
samples (n ¼ 16 for PDAC) and normal pancreatic tissue (n ¼ 17 for
normal) by quantitative PCR (qPCR) and observed approximately
fivefold upregulation in the expression of Pak1 in PDAC tumors as
compared with the normal pancreatic tissue (Figure 1b). Further-
more, to evaluate the significance of our above findings, we
examined the expression levels of Pak1 in human tissue
microarray containing 60 cases of PDAC tissue samples with the
corresponding adjacent normal tissue by immunohistochemistry.
Although 70% of the PDAC tumors stained positive for Pak1, more
than 40% of the tumors exhibited intense staining for Pak1
(Table 1). The representative images of the stained tissue array are
shown in Figure 1c and Supplementary Figure S1. Next, in order to
choose the appropriate model system for our in vitro study, we
evaluated the expression of Pak1 protein in seven PDAC cell lines
and in the immortalized, nontumorigenic HPDE6 (E6/E7) cell line
by western blot and qPCR analysis. Results showed elevated
expression of Pak1 in all of the malignant cell lines compared with
the nontumorigenic control (Figures 1d and e). Together, these
results suggested that elevated Pak1 levels in PDAC tumors and
cell lines might contribute to the transforming properties of
pancreatic cells.
Transforming and tumorigenic properties of Pak1
The dysregulated expression of Pak1 in PDAC tissue samples
prompted us to examine its significance in the tumorigenic
properties of pancreatic tumor cells.
Generation of stable Pak1 overexpressing and Pak1 shRNA Knock-
down model cells. To explore the role of Pak1 as an upstream
determinant of oncogenic transformation in pancreatic cancers,
we generated stable clones of Pak1 overexpression in Mia Pa Ca 2
cell line. Stable overexpression of ectopic Pak1 expression was
confirmed by immunoprecipitation of T7-tagged Pak1. The
overexpression was also confirmed by running whole-cell lysates
and probing for Pak1 as well as immunofluorescence for Pak1
using confocal microscopy (Figure 2a and Supplementary Figure
S2). Similarly, we also developed stable lentiviral Pak1 short
hairpin RNA (shRNA) Knockdown clones in MDA Panc 28 cell line.
The downregulation of Pak1 was confirmed by western blot, qPCR
and immunofluorescence (Figure 3a and Supplementary Figure
S2). More than 90% knockdown (KD) in the levels of Pak1 was
observed in the Pak1 KD clones compared with the control
nontarget (NT) shRNA-expressing clone.
Figure 1. Pak1 is upregulated in PDAC. (a) Lysates from paired samples of human PDAC tumors and adjacent normal pancreatic tissue were
analyzed for Pak1 expression by western blotting. (b) mRNA levels of Pak1 in human PDAC and normal pancreatic tissue samples.
(c) Representative immuno-stained (against Pak1) paraffin-embedded tissue sections of paired normal and PDAC tumor tissue array.
(d) Western blot and (e) qPCR analysis of Pak1 expression in indicated PDAC cell lines and an immortalized normal pancreatic epithelial cell
line (HPDE6/E6E7). Vinculin/b-actin was used as loading control.
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
2
Oncogene (2014) 1 10 & 2014 Macmillan Publishers Limited
Pak1 regulates the transforming properties of pancreatic cancer
cells. Further, in order to establish the possible role of Pak1 in the
transforming properties of pancr eatic cells, we per formed
proliferation, colony formation, soft agar, migration and invasion
assays with the Pak1 overexpression and KD models of Mia Pa Ca
2 and MDA Panc 28, respectively. Proliferation a ssay results
showed that upon Wt-Pak1 overexpression in Mia Pa Ca 2 cell
line, the cell proliferation rate goes up significantly as compared
with the empty vector carrying cell line (Figure 2b). On the other
hand, MDA Panc 28 KD clones expressing lesser Pak1 than the NT
control clone exhibited decreased proliferation (Figure 3b). In the
col ony formation assay, we observed larger number of colonies
for med in Wt-Pak1 overexpression clone compared with the
empty vector control in Mia Pa Ca 2 cells (Figure 2c). On the other
hand, we observed fewer number of colo nies formed in Pak1
downregulated clones in MDA Panc 28 co mpared with the NT
shRNA-expressing clone (Figure 3 c). Further, t o assess the role of
Pak1 in promoting anchorage-independent growth, w e carried
out soft agar assay with both overexpression and KD models of
Mia Pa Ca 2 and MDA Panc 28, respectively. Results showed that
ectopic expressio n of Wt-Pak1 increased Mia Pa Ca 2 cell line’s
ability to form colonies in an anchorage-independent manner
compared with the empty vector clones (Fi gure 2d). Similarly,
stable KD of Pak1 in MDA Panc 28 reduced the number of
col onies formed in the soft agar as say (Figure 3d). Ne xt, to assess
the role of Pak1 o n the invasive and migratory behavior of
pancreatic cancer cells, we examined the clones using Boyden
chambers coated with or without matrigel, respectively. We
observed that stabl e overexpress ion of Pak1 in Mia Pa Ca 2
res ulted in a multiple-fold increase in the number of c ells
migrating as well as inv ading (Figures 2e and f). Likewise, there is
a multiple-fold decreas e in the number of cells migra ting and
invading towards the bo ttom well in Pak1 downregulated clones
in MDA Panc 28 compared with the NT shRNA -expressing clone
(Figures 3e and f). Similar results were obs erved with NT and
Pak1 KD clones made in Mia Pa Ca 2 (S upplementary Figure S3).
Consistent with these results, in vitro wound healing assay
showed that Pak1 overexpres sion lead s to improved wound
Table 1. Immunohistochemistry data of Pak1 expression in tissue samples
Negative Weak positive Intense staining
Adjacent normal 58.3% (35 of 60) 35% (21 of 60) 6.7% (4 of 60)
Pancreatic ductal adenocarcinoma 31.7% (19 of 60) 25% (15 of 60) 43.3% (26 of 60)
Figure 2. Pak1 regulates the transforming property of pancreatic cells. (a) Western blot analysis showing the stable overexpression of Pak1 in
Mia Pa Ca 2 cell line. Top panel shows immunoprecipitated T7-Pak1 from the clones, bottom two panels are direct lysates. Pak1 overexpression
accelerates (b) cell proliferation; (c) colony formation; (d) anchorage-independent soft agar growth; (e) migration; (f ) invasion and (g) wound
healing properties in stable Mia Pa Ca 2 clones. Each value represents the mean
±
s.e.m. *Po0.05, **Po0.005 compared with vector clones.
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
3
& 2014 Macmillan Publishers Limited Oncogene (2014) 1 10
healing and migration rates as compared with the control vector
carrying Mia Pa Ca 2 cell line (Figure 2g ). Correspondingly, stable
KD o f Pak1 in MDA Panc 28 resulted in a significant reduction in
the wound healing capacity (Figure 3g). All these results suggest
that Pak1 is the key effector node in controlling the downstream
signaling axis in PDAC.
Transforming potential of Pak1 in wild-type KRAS model.As
mutations in the KRAS oncogene account for more than 90% of
human pancreatic cancers, we intended to check whether Pak1
can transform pancreatic cells independent of mutant KRAS. To
this end, we transiently overexpressed Wt-Pak1 in BxPC 3 cell line
(having Wt-KRAS) and performed colony formation and ancho-
rage-independent soft agar assays with these cells. Results
showed that Pak1-transfected BxPC 3 cells showed significantly
increased ability to form colonies in both anchorage-dependent
and -independent manner compared with the empty vector
transfect (Figures 4a and b). Simultaneously, to prove the other
way, we developed stable lentiviral Pak1 shRNA-mediated KD
clones in BxPC 3 cell line. The downregulation of Pak1 was
confirmed by qPCR, western blot and immunofluorescence
(Figure 4c and Supplementary Figure S2). More than 95% KD in
the levels of Pak1 was observed in the Pak1 knockdown (KD1)
clone as compared with the control NT shRNA-expressing clone.
We performed colony formation, soft agar, migration and invasion
assays with these clones and results showed that stable KD of
Pak1 in BxPC 3 reduced the number of colonies formed in the
clonogenic and in soft agar assay (Figures 4d and e). In addition, a
significant reduction in the number of cells migrated and invaded
as compared with the BxPC 3 NT clone (Figures 4f and g). Taken
together, our results illustrate the potential role of Pak1 in
transforming pancreatic cells with intact KRAS (Wt).
Tumorigenic potential of Pak1 in nude mouse model. Consistent
with in vitro results, MDA Panc 28 shRNA Pak1 KD clones failed to
form tumors in nude mice, whereas the NT shRNA-expressing
clone formed palpable tumors (Figure 5a). These results showed
that Pak1 drives the tumorigenic property in pancreatic cancer
cells in vivo.
Molecular mechanism of Pak1-induced tumorigenesis
Fibronectin—a novel molecular target of Pak1. The evidence from
the above results suggested that Pak1 might have an important
role in the regulation of pancreatic cell’s invasive and metastatic
behavior. To identify the downstream molecular targets that might
be regulated through Pak1 signaling and also associated with the
tumorigenesis and metastatic potential of cells, we performed qPCR
expression profiling of 84 genes associated with tumorigenesis and
metastasis utilizing MDA Panc 28 NT shRNA-expressing clone NT
and Pak1 shRNA-expressing KD clone KD1 (Figure 5b). A complete
list of genes analyzed through the gene array and the genes that
are modulated by Pak1 activity are given as Supplementary Table 2.
The shortlisted genes that are significantly modulated by Pak1 are
given as Supplementary Table 3. As determined by this method, we
identified fibronectin as one of the potential targets that was
drastically downregulated in the Pak1 KD clone as compared with
NT clone. We further validated fibronectin in both overexpression
and KD models of Mia Pa Ca 2, MDA Panc 28 and BxPC 3,
respectively using individual qPCR assays and immunoblotting for
fibronectin. Results showed that fibronectin mRNA and protein
levels were significantly increased in Wt-Pak1 overexpressing Mia
Pa Ca 2 cell line as compared with the empty vector transfect
(Figure 5c). Similarly, there is a significant decrease in the
fibronectin mRNA and protein levels in the stable KD of Pak1 in
MDA Panc 28 and BxPC 3 clones as compared with the NT clones
Figure 3. Pak1 silencing decreases the transforming property of pancreatic cells. shRNA-mediated downregulation of Pak1 in stable MDA Panc
28 clone as confirmed by (a) Western blot and qPCR. Pak1 downregulation reduces (b) cell proliferation; (c) colony formation;
(d) anchorage-independent soft agar growth; (e) migration; (f ) invasion and (g) wound healing properties. Each value represents the
mean
±
s.e.m. *Po0.05, **Po0.005 compared with non-target clones.
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
4
Oncogene (2014) 1 10 & 2014 Macmillan Publishers Limited
(Figure 5c). In addition, we analyzed for the expression of
extracellular fibronectin (fibronectin EDA domain) by performing
western blotting using fibronectin EDA domain specific antibody
and results showed that there is a significant change in its
expression with the modulation of Pak1 (Supplementary Figure
S4A). We also assayed for the secretory fibronectin in the cell
culture conditioned media using ELISA kit and results showed a
significant change in its levels with the modulation of Pak1
(Supplementary Figure S4B).
Consistent with this, overexpression of Wt-Pak1 significantly
augmented fibronectin promoter activity in the Mia Pa Ca 2 cell line
(Figure 5d), whereas silencing of Pak1 significantly downregulated
fibronectin promoter activity in MDA Panc 28 cell line (Figure 5e).
Further, to determine whether kinase activity of Pak1 is required for
the regulation of fibronectin promoter, we used Wt-Pak1, Pak1-
T423E (kinase active) and Pak1-K299R (kinase dead) constructs and
performed fibronectin promoter luciferase assay in parental Mia Pa
Ca 2 cell line. Results showed that expression of Pak1-T423E led to
an increase in fibronectin promoter activity, whereas expression of
Pak1-K299R significantly decreased basal promoter activity
(Figure 6a). Interestingly, depletion of fibronectin using siRNA
significantly abated Pak1’s ability to form colonies in colony
formation assay and soft agar assay; it showed significant reduction
in the number of cells migrated and invaded as compared to the
control siRNA transfect in Wt-Pak1 stable overexpressing Mia Pa Ca
2 clone (Figures 6b–e). On the contrary, transient overexpression of
fibronectin cDNA in Pak1 KD clones showed increase in the
tumorigenic phenotype of Pak1 KD clones as evidenced by increase
in migration potential, soft agar and colony forming ability
(Supplementary Figure S5). Taken together, these results indicate
that Pak1 can promote tumorigenic properties in part through the
regulation of fibronectin promoter.
Pak1 regulates fibronectin via NF-kB–p65 pathway. The observa-
tion that active Pak1 (T423E) induced and kinase dead Pak1
(K299R) diminished fibronectin promoter activity prompted
us to investigate the mechanism of regulation of fibronectin
transcription by Pak1 in cancer cells. It was previously reported
that activated Pak1 gets localized to the nucleus.
23
In addition,
Pak1 has been shown in various cellular contexts to modulate
gene expression of nuclear factor of activated T cell,
phosphofructokinase-muscle isoform, tissue factor and tissue
Figure 4. Pak1 modulates the transforming property of KRAS wild-type pancreatic cells. Western blot analysis showing the transient
overexpression of Pak1 in BxPC 3 cell line. Pak1 overexpression accelerates (a) colony formation and (b) anchorage-independent soft agar
growth in transiently Pak1-overexpressed BxPC 3 cell line. Each value represents the mean
±
s.e.m. *Po0.05 compared with vector clones.
(c) shRNA-mediated downregulation of Pak1 in stable BxPC 3 cell line as confirmed by western blot and qPCR. Pak1 downregulation abrogates
(d) colony formation and (e) anchorage-independent soft agar growth, (f) invasion and (g) migration. Each value represents the mean
±
s.e.m.
*Po0.05, **Po0.005 compared with non-target clones.
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
5
& 2014 Macmillan Publishers Limited Oncogene (2014) 1 10
factor pathway inhibitor through the regulation of several
transcription factors, as it lacks transcriptional activity of its
own.
24
It was also shown that Pak1 has a role in the activation of
NF-kB.
25
Recently, it was reported that p65 subunit of NF-kB
activates fibronectin transcription.
26
Based on these, we
hypothesized that Pak1 might be regulating fibronectin via NF-
kB–p65. To check this possibility, we next performed a sequence
analysis of the fibronectin promoter using the ConSite prediction
software to identify the putative binding sites for transcriptional
factors that might be responsible for Pak1-mediated regulation of
fibronectin transcription. We found that NF-kB is one of the
potential transcriptional factors that bind to fibronectin promoter.
As Pak1’s role in activation of NF-kB is already established, we
hypothesized that this might be an appropriate candidate to study
Pak1-mediated regulation of fibronectin. To examine this
possibility, we initially checked for the levels of phospho-p65 in
the overexpression and KD models of Mia Pa Ca 2, MDA Panc 28
and BxPC 3, respectively by western blotting. Consistent with the
role of Pak1 in the activation of NF-kB as reported earlier, results
showed that there is a significant increase in the level of phospho-
p65 in the Wt-Pak1 overexpressing Mia Pa Ca 2 cell line as
compared with the empty vector transfect. Similarly, there is a
significant decrease in the phospho-p65 levels in the stable KD of
Pak1 in MDA Panc 28 and BxPC 3 clones as compared with the NT
clones (Figure 6f). There is no change in the levels of total p65.
Further, we observed a synergistic increase in the fibronectin
promoter activity upon co-transfection of p65 along with Wt-Pak1
(Figure 6g), whereas shRNA-mediated p65 depletion resulted in a
significant decrease in the Pak1-induced fibronectin promoter
activity (Figure 6h). These results clearly indicate that Pak1 could
regulate fibronectin via NF-kB–p65 pathway.
Pak1 recruitment onto the fibronectin promoter. Further, to gain a
deeper insight into the molecular mechanism involved in NF-kB-
mediated Pak1 regulation of fibronectin promoter, we focused on
the nuclear role of Pak1. To start with, we examined for the levels
of nuclear Pak1 in Pak1-overexpression clones of Mia Pa Ca 2 by
subcellular fractionation, followed by western blotting. Consistent
with the previous reports that active Pak1 translocates to the
nucleus, we observed that there is a significant increase in the
amount of Pak1 in the nuclear compartment in the Wt-Pak1-
overexpressing Mia Pa Ca 2 clone. We also observed an increase in
the nuclear levels of p65 in the Pak1 overexpression clones of Mia
Pa Ca 2 (Figure 7a). Consistent with this, confocal analysis for Pak1
in Pak1-overexpressing Mia Pa Ca 2 showed increased nuclear
localization of Pak1, whereas the KD clones of Pak1 in MDA Panc
28 and BxPC 3 showed decreased nuclear localization
(Supplementary Figure S2). Further, to establish that nuclear
Pak1 is essential for the activation of fibronectin promoter, we
used a Pak1 NLS mutant construct
23
and showed that this mutant
is not able to induce fibronectin promoter activity (Figure 7b).
From the above results, it was clear that kinase activity of Pak1 and
its nuclear localization are required for the activation of
fibronectin promoter via NF-kB–p65. Therefore, to establish the
molecular cooperation between these molecules in the regulation
of fibronectin promoter, we examined the recruitment of Pak1
onto the predicted 700 bp fibronectin core promoter region using
a chromatin immuno precipitation (ChIP)-based promoter walk
Figure 5. Fibronectin is a novel molecular target of Pak1. (a) Tumor xenograft images of MDA Panc 28/NT and MDA Panc 28/KD1 shRNA clones
implanted subcutaneously in nude mice. Western blot confirming Pak1 downregulation in MDA Panc 28 clones before injecting into mice.
Graph showing tumor growth in mouse xenografts. (b) Heat map of the human metastatic PCR array performed on MDA Panc 28 Pak1
knockdown clones. (c ) qPCR and western blot analysis for fibronectin levels in Pak1 overexpressing and knockdown clones. (d) Mia Pa Ca 2
cells were co-transfected with 0.5 mg of 1.2 k b pFN1-luciferase reporter and 0.5 mg of Pak1 or plasmid pcDNA 3. 1. After 24 h, the cells were
lysed, luciferase activity was measured (n ¼ 3) and normalized with b-galactosidase activity. Each value represents the mean
±
s.e.m.
**Po0.005 compared with vector control. (e) MDA Panc 28 cell lines were transfected with Pak1 siRNA/control siRNA. After 48 h of
transfection, the cells were transfected with 1.0 mg of 1.2 kb pFN1-luciferase reporter and with a pGL3-basic luciferase reporter. After 24 h, the
cells were lysed, luciferase activity was measured (n ¼ 3) and was normalized with b-galactosidase activity. Each value represents the
mean
±
s.e.m. **Po0.005, ***Po0.001, compared with control siRNA. Western blot analysis confirming Pak1 downregulation after siRNA
transfection of MDA Panc 28 cells.
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
6
Oncogene (2014) 1 10 & 2014 Macmillan Publishers Limited
with three different sets of primers covering the 700 bp region.
Results showed that Pak1 was recruited to the fibronectin
promoter at a region between 13 to 220 bp, which is about
200 bp region (Set 1; Figure 7ci). This was confirmed by
sequencing the PCR product of ChIP. We also observed that NF-
kB–p65 is also recruited to the same region as Pak1 (Figure 7cii).
Sequence analysis of this 200 bp (region 1) of the fibronectin
promoter using the ConSite prediction software revealed that NF-
kB binding consensus sequence GGGACAGCCC lies in the same
region. These results suggest that both Pak1 and NF-kB–p65
might be recruited to the same region onto the fibronectin
promoter. To confirm this, we performed a sequential double ChIP
analysis with Pak1 followed by p65 antibody and found that
indeed Pak1 and NF-kB–p65 are corecruited onto the region 1
(Figure 7ciii). To further demonstrate the importance of NF-kB–
p65 in Pak1-mediated fibronectin upregulation, we silenced NF-
kB–p65 expression using shRNA and examined for the recruitment
of Pak1 onto the region 1. Results showed that there is a
significant decrease in the amount of Pak1 recruited to the region
1 upon silencing NF-kB–p65 (Figure 7civ). Further, in order to
investigate the functional significance of this 200 bp region in the
transcriptional regulation of fibronectin by Pak1, we made a
fibronectin promoter construct deleting this 200 bp region where
Pak1–NF-kB–p65 are recruited, and performed reporter luciferase
assay. Results showed that Pak1 was not able to induce the
fibronectin promoter activity without the 200 bp region
(Figure 7d). Consistently, Pak1 did not show activation of
fibronectin promoter luciferase, when point mutations were made
in the NF-kB–p65 binding consensus site on fibronectin promoter
(from GGGACAGCCC to T GGACAGCCC) (Figure 7d). Next, to
examine the direct recruitment of Pak1 and NF-kB-p65 to the
fibronectin promoter, we performed electrophoretic mobility shift
Figure 6. Pak1 modulates fibronectin via NF-kB pathway. (a) Mia Pa Ca 2 cells were co-transfected with 0.5 mg of 1.2 kb pFN1 -luciferase
reporter and 0.5 mg of Pak1, or Pak1-T423E or Pak1-K299R or pCMV control vector. After 24 h, the cells were lysed, luciferase activity was
measured (n ¼ 3) and was normalized with b-galactosidase activity. Each value represents the mean
±
s.e.m. **Po0.005, compared with vector
control. (b) Western blot analysis confirming fibronectin downregulation after siRNA transfection of Mia Pa Ca 2 cells. Vector and Pak1
overexpressed Mia Pa Ca 2 cells transfected with FN1 siRNA/control siRNA were subjected to (b) colony formation, (c) anchorage-independent
soft agar, (d) migration and (e) invasion. Each value represents the mean
±
s.e.m. **Po0.005, ***Po0.001, compared with control siRNA (f ).
western blot showing the phosphorylation status of p65 in Pak1 modulated clones. (g) BxPC 3 cells were co-transfected with 0.5 mg of 1.2 kb
pFN1-luciferase reporter and 0.5 mg of Pak1 or p65 plasmid or both. After 24 h, the cells were lysed, luciferase activity was measured ( n ¼ 3)
and was normalized with b-galactosidase activity. Each value represents the mean
±
s.e.m. *Po0.05, **Po0.005, compared with vector control
(h). BxPC 3 cells were co-transfected with 0.5 mg of 1.2 kb pFN1-luciferase reporter and 0.5 mg of Pak1 and/or p65 shRNA plasmid/scramble
shRNA. After 24 h, the cells were lysed, luciferase activity was measured (n ¼ 3) and was normalized with b-galactosidase activity. Each value
represents the mean
±
s.e.m. ***Po0.001 compared with the scrambled shRNA treated.
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
7
& 2014 Macmillan Publishers Limited Oncogene (2014) 1 10
assay using the oligonucleotides containing the NF-kB consensus
sequence GGGAGAGCCC, and binding of the Pak1 and NF-kB–p65
was analyzed. Results showed the formation of DNA–protein
complexes in the Wt consensus sequence, whereas there is no
binding in the mutant probe. In addition, the observed complex
was highly inhibited in the presence of Pak1 and p65 antibody
alone or together. Furthermore, electrophoretic mobility shift
assay using nuclear extracts from NF-kB–p65 shRNA showed a
significant decrease in the DNA–protein complex (Figure 7e).
Pak1 physically interacts with NF-kB–p65. It has been previously
reported that Pak1 activates NF-kB–p65, resulting in the
p65 phosphorylation followed by nuclear localization of NF-kb–
p65 complex. However, our above finding that Pak1 and
NF-kB–p65 are corecruited onto the fibronectin promoter raises
the possibility of physical interaction between these two proteins.
Results from in vitro pulldown and in vivo coimmunoprecipitation
experiments showed that indeed these two proteins are physically
associated with each other (Supplementary Figure S6).
Figure 7. Pak1 recruitment onto the fibronectin promoter. (a) Western blot showing increased p65 and Pak1 in the nuclear lysate of Pak1
clones indicating the nuclear import of p65 induced by Pak1 overexpression. PARP and a-tubulin were used as nuclear and cytoplasmic
loading controls. (b) BxPC 3 cells were co-transfected with 0.5 mg of 1.2 kb pFN1-luciferase reporter and 0.5 mg of Pak1 or NLS Pak1 mutant
plasmid or vector control. After 24 h, the cells were lysed, luciferase activity was measured (n ¼ 3) and activity was normalized with b-
galactosidase activity. Each value represents the mean
±
s.e.m. **Po0.005, compared with vector. (c) Schematic representation of the FN1
promoter, showing the regions analyzed. ChIP analysis showing the recruitment of (ci) PAK1 and (cii)p65 on the human FN1 promoter in Mia
Pa Ca 2 cells. (ciii) Double ChIP analysis of PAK1–p65 complex recruitment onto the FN1 chromatin region 1 (set 1) in Mia Pa Ca 2 cells. The first
ChIP was carried out with an anti-PAK1 antibody followed by second ChIP with anti-p65 antibody. (civ) ChIP analysis showing the reduction in
the recruitment of Pak1 to FN1 promoter after knockdown p65 using shRNA plasmid. p65 shRNA-mediated knockdown confirmed by western
blotting. Input represents around 1–10% of the total immunoprecipitated DNA. (d) Bx PC 3 cells were co-transfected with 0.5 mg of 1.2 kb
pFN1-luciferase reporter or 200 bp deletion of pFN1-luciferase reporter (pDel) or SDM mutant of pFN1-luciferase reporter (pSDM) and/or
0.5 mg of Pak1 plasmid. After 24 h, the cells were lysed, luciferase activity was measured (n ¼ 3) and was normalized with b-galactosidase
activity. Each value represents the mean
±
s.e.m. *Po0.05, **Po0.005, compared with vector. (e) Electrophoretic mobility shift assay (EMSA)
analysis of p65 and PAK1 binding to the human fibronectin (FN1) promoter using the wild-type biotin-labeled probe (*Probe), cold probe and
biotin-labeled mutant (Mut) oligonucleotides encompassing p65 consensus sites using Mia Pa Ca 2 nuclear lysates. # Indicates the specific
band of interest. EMSA using nuclear extracts from NF-kB–p65 shRNA showing a significant decrease in the DNA–protein complex.
(f) Schematic representation of Pak1 regulation of fibronectin via Pak1–NF-kB–p65 complex.
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
8
Oncogene (2014) 1 10 & 2014 Macmillan Publishers Limited
DISCUSSION
Previously, few of the upstream regulators of Pak1 pathway, such
as EGFR
27
and VAV1 protein,
16
have been shown to be involved
and dysregulated in PDAC. However, to our knowledge, this is the
first study to directly implicate the role of Pak1 in pancreatic
tumorigenesis. Recently, Pak1 has emerged as a potential
therapeutic target owing to its oncogenic role in several cancer
types.
28–31
Results from this study show that a majority of PDACs
and PDAC cell lines express high levels of Pak1, clearly indicating
that Pak1 has an important role in PDAC tumorigenesis. This was
supported and validated in vitro by Pak1 overexpression and Pak1
downregulated cell line models and in vivo by an animal model.
Importantly, our data from BxPC 3 cell line clearly demonstrated
that Pak1 modulates transforming properties in Wt-KRAS
pancreatic model also implying Pak1 as a potential oncogene in
KRAS intact tumorigenesis (which need to be explored further).
Further, for investigating the possible mechanism by which Pak1
could promote PDAC tumorigenesis, we utilized a qPCR array that
encoded several classes of protein factors including extracellular
matrix components, cell adhesion, cell cycle, cell growth and
proliferation, apoptosis, transcription factors and regulators and
other genes related to tumor metastasis. Among these, three
genes relevant to PDAC were analyzed, revealing that fibronectin
showed consistent and significant change, whereas Syk did not
show significant change and insulin like growth factor-1 (IGF-1)
was not detectable in the cell lysates (Supplementary Figure S7).
Hence, we established fibronectin as the potential transcriptional
target of Pak1. Fibronectin is an important component of the
extracellular matrix and has been shown to have a vital role in cell
migration, invasion and metastasis of tumors.
32,33
As Pak1 lacks
transcriptional activity of its own, we looked for intermediate
transcriptional factor that could mediate fibronectin transcription.
Previous studies have reported that p65 subunit of NF-kB has a
role in activating fibronectin transcription.
26
It is already reported
that Pak1 regulates cyclin D1 via NF-kB pathway.
14
In addition,
Pak1 signaling has been reported to play a crucial role in the
activation and nuclear translocation of p65 subunit of NF-kB.
25
Owing to the fact that the connection between Pak1 and NF-kB–
p65 is previously established and that there is a potent NF-kB
binding site on fibronectin promoter, we hypothesized that Pak1
might be regulating fibronectin via NF-kB. This was supported and
confirmed by promoter luciferase studies, Pak1–p65 interaction
studies, ChIP experiments and electrophoretic mobility shift assay
analysis. Further, our results from active-, kinase dead- and NLS
Pak1 mutants also showed that Pak1 kinase activity and its nuclear
localization are essential for induction of fibronectin transcription.
This clearly supports the previous observation that nuclear
localization of Pak1 has an important role in promoting
tumorigenic properties of cancer cells.
34
In summary, the study
illustrated that Pak1 interacts with and modulates the activity of
NF-kB–p65 subunit and together as a complex might move into
the nucleus where they bind with the fibronectin promoter and
enhance its transcription, thus promoting the invasion and
metastatic potential of the pancreatic cancer cells (Figure 7f).
Given the fact that Pak1 is a central player in growth factor
signaling and morphogenetic processes that control cell prolifera-
tion, migration, invasion and cytoskeleton organization and now
being recognized as a transcriptional regulator for several genes, it
would be worthwhile to explore Pak1 inhibitors that interfere with
Pak1 activity to deliver therapeutic benefits in several cancers.
MATERIALS AND METHODS
Antibodies and reagents
Pak1, phospho-p65 and PARP antibody were obtained from Cell Signaling
Technology (Beverly, MA, USA), Vinculin and b-actin from Sigma-Aldrich
(St Louis, MO, USA), T7 from Bethyl Laboratories (Montgomery, TX, USA),
p65 and a-tubulin from Santa Cruz Biotechnology (Dallas, TX, USA), and
fibronectin—total and EDA (IST-9) from BD Biosciences (Bedford, MA, USA)
and Abcam (Cambridge, MA, USA) respectively. Fibronectin human ELISA
kit was purchased from Abcam. siRNA: Pak1 was obtained from Cell
Signaling Technology, FN1 from SA Biosciences (Valencia, CA, USA) and
control siRNA from Santa Cruz Biotechnology. Plasmids: pCMV6-Myc-Pak1,
pCMV6-Myc-Pak1-T423E and pCMV6-Myc-Pak1-K299R were obtained from
Addgene, Cambridge, MA, USA deposited by Dr Jonathan Chernoff, Fox
Chase Cancer Centre, Philadelphia. p65 plasmid, anti-p65 shRNA and
scramble shRNA plasmid were obtained from Dr K Boume
´
diene, University
of Caen Lower Normandy, France, pFN1 (1.2 Kb) Luc Promoter (pGL3–FN1
Luc) from Dr Eric S White, University of Michigan Medical School, USA. All
the vectors (pcDNA3.1-A,B,C and pcDNA6-V5-HisA,B,C) were purchased
from Invitrogen (Carlsbad, CA, USA). pcDNA3.1-C-T7-Pak1, pcDNA6-V5-
Pak1, and pGEX-GST-Pak1 were cloned from pCMV6-myc-Pak1. Pak1 NLS
mutant was made by site-directed mutagenesis using conditions and
primers as described earlier.
23
pRC-FN1-Wt and pRC-FN1-EDA domain was
kindly gifted by Dr Andre
´
s F Muro, ICGEB, Trieste, Italy. All the recombinant
plasmids and lentiviral particles used in this manuscript have the clearance
and approval of DBT—Institutional Biosafety Committee (IBSC) of IIT
Madras. Additional primer details are provided in Supplementary Table 1.
Cell lines and tissues
The PDAC cell lines Mia Pa Ca 2 and Panc 1 were procured from NCCS,
India. Capan 1 was kindly gifted by Dr Subhash C. Chauhan (Cancer Biology
Research Center, Sanford Research, USA); Capan 2 by Prof Ilona Silins
(Institute of Environmental Medicine, Karolinska Institutet, Sweden); MDA
Panc 28 by Dr Marsha Fraizer (University of Texas, M. D. Anderson Cancer
Center, USA); MDA Panc 48 by Dr Paul J Chiao (University of Texas, M. D.
Anderson Cancer Center, USA); and BxPC 3 by Dr Uddalak Bharadwaj
(Baylor College of Medicine, USA). Human immortalized ductal epithelial
cells HPV16 E6/E7 (HPDE6/E6E7) was purchased from Applied Biological
Materials (Richmond, BC, Canada). Human pancreatic tissue samples were
collected and the study was approved by the Institutional Human Ethics
Committee.
Generation of stable clones
Stable Pak1 overexpression clones were generated upon transfection of
pcDNA3.1-T7-Pak1 plasmid and corresponding empty vector in Mia Pa Ca
2, followed by selection with G418 (MP Biomedicals, Santa Ana, CA, USA).
Pak1 KD clones were generated by transducing MDA Panc 28, Mia Pa Ca 2
and BxPC 3 with MISSION lentiviral particles coding for shRNA sequence
against PAK1 and NT shRNA as the negative control (Sigma-Aldrich) and
were selected for stable integration using puromycin (MP Biomedicals).
Immunohistochemistry
Tissue microarrays containing 60 tumors and adjacent normal tissue were
purchased from US Biomax (Rockville, MD, USA) and ISU ABXIS AccuMax
(San Diego, CA, USA). Pak1 expression was evaluated using an indirect
immunoperoxidase procedure (ABC-Elite, Vector Laboratories, Burlingame,
CA, USA). Staining results were scored by the pathologist. Severity of
cytoplasmic staining of tumor cells and rate of staining were graded semi-
quantitatively as follows: 0 ¼ o1%, 1 þ¼1–10%, 2 þ¼11–50%,
3 þ¼51–80%, 4 þ¼480% of tumor cells stained. Based on the score,
Pak1 expression was categorized as negative (no cytoplasmic staining in
tumor cells, 0,1 þ ), weak (low staining, 2 þ ), and high (diffuse moderate to
strong staining in tumor cells, 3 þ and 4 þ ).
Cell migration and invasion assays
Cells (5 10
4
) were suspended in serum-free media and added to the
upper chamber of an insert (Biocoat-BD Biosciences), and the insert was
placed in a 24-well plate containing media supplemented with 10% fetal
bovine serum. The migrated/invaded cells were stained and scored.
Anchorage-independent colony formation assay
Cells were suspended in complete media containing 0.5% agarose. Cells
were overlaid onto a bottom layer of solidified 0.8% agarose in media at
cell concentrations of 5 10
3
cells per well and incubated for 2 weeks,
stained and counted.
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
9
& 2014 Macmillan Publishers Limited Oncogene (2014) 1 10
Generation of SDM and deletion construct of fibronectin promoter
Using pGL3-FN (1.2 Kb)-luc plasmid as template, mutations were made in
the NF-kB consensus site within the region 318 to 13 by mutating
G-T in the position 97 using QuikChange kit (Stratagene, La Jolla, CA,
USA). The deletion construct excluding the Pak1 binding region ( 318 to
78) was made with NEB Q5 polymerase, using specific primers, followed
by ligation of the digested fragments. Additional details are provided in
Supplementary methods.
Animal studies
Mice (nu/nu) that were 6 weeks old were obtained from the Animal
Breeding Facility, Reliance Life Sciences, India, after obtaining ethical
clearance. Mice were divided into two groups and injected subcutaneously
on both the sides using a 25-gauge needle with MDA Panc 28 stable Pak1
KD clones versus the NT clones. The growth of xenografts was monitored.
PCR array studies
Human tumor metastasis PCR array for 84 genes involved in tumorigenesis
and metastasis was purchased from SA Biosciences. Target genes whose
expression was differentially regulated (at least fivefold difference) in the
clones were selected. Additional details are provided in Supplementary
methods and Supplementary Tables 2 and 3.
Statistical analysis
Data are expressed as the mean
±
s.e.m. and analyzed by Student’s t-test
using SigmaPlot (SYSTAT software Inc., Chicago, IL, USA). The P-value,
Po0.05 is considered significant.
CONFLICT OF INTEREST
The authors declare no conflict of interest.
ACKNOWLEDGEMENTS
We thank Silpa for help in mutagenesis and cloning and Swarnalatha for help with
Pak1 lentiviral clones. Thanks to Sekar Sathiya, Rohan Prasad, Hemadev and the
entire team of the Centre for Toxicology and Developmental Research (CEFT), Sri
Ramachandra University, Chennai, for help with animal experiments. We thank the
Department of Biotechnology (DBT), Government of India for the financial support to
SKR (grant no.: BT/PR13559/Med/30/283/2010) and Indian Institute of Technology
Madras (IITM) for all other facilities.
REFERENCES
1 World Cancer Research Fund/American Institute for Cancer Research. Continuous
Update Project Summary. Food, Nutrition, Physical Activity, and the Prevention of
Pancreatic Cancer 2012.
2 Hidalgo M, Von Hoff DD. Translational therapeutic opportunities in ductal
adenocarcinoma of the pancreas. Clin Cancer Res 2012; 18: 4249–4256.
3 Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010; 60:
277–300.
4 Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet
2011; 378: 607–620.
5 Maitra A, Hurban RH. Pancreatic cancer. Annu Rev Pathol 2008; 3: 157–188.
6 Costello E, Greenhalf W, Neoptolemos JP. New biomarkers and targets in pan-
creatic cancer and their application to treatment. Nat Rev Gastroenterol Hepatol
2012; 9: 435–444.
7 Yeh JJ, Der CJ. Targeting signal transduction in pancreatic cancer treatment.
Expert Opin Ther Targets 2007; 11: 673–694.
8 Krautz C, Ru¨ ckert F, Saeger HD, Pilarsky C, Gru¨ tzmann R. An update on molecular
research of pancreatic adenocarcinoma. Anticancer Agents Med Chem 2011; 11:
411–417.
9 Falasca M, Selvaggi F, Buus R, Sulpizio S, Edling CE. Targeting phosphoinositide
3-kinase pathways in pancreatic cancer--from molecular signalling to clinical trials.
Anticancer Agents Med Chem 2011; 11: 455–463.
10 Almoguera C, Shibata D, Forrester K, Martin J, Arnheim N, Perucho M. Most
human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell
1988; 53: 549–554.
11 Morris 4th JP, Wang SC, Hebrok M. KRAS, Hedgehog, Wnt and the twisted
developmental biology of pancreatic ductal adenocarcinoma. Nat Rev Cancer
2010; 10: 683–695.
12 Bokoch GM. Biology of the p21-activated kinases. Annu Rev Biochem 2003; 72:
743–781.
13 Kumar R, Gururaj AE, Barnes CJ. p21-activated kinases in cancer. Nat Rev Cancer
2006; 6: 459–471.
14 Arias-Romero LE, Chernoff J. A tale of two Paks. Biol Cell 2008; 100:
97–108.
15 Balasenthil S, Sahin AA, Barnes CJ, Wang RA, Pestell RG, Vadlamudi RK et al.
p21-activated kinase-1 signaling mediates cyclin D1 expression in mammary
epithelial and cancer cells. J Biol Chem 2004; 279: 1422–1428.
16 Fernandez-Zapico ME, Gonzalez-Paz NC, Weiss E, Savoy DN, Molina JR, Fonseca R
et al. Ectopic expression of VAV1 reveals an unexpected role in pancreatic cancer
tumorigenesis. Cancer Cell 2005; 7: 39–49.
17 Crnogorac-Jurcevic T, Efthimiou E, Capelli P, Blaveri E, Baron A, Terris B et al. Gene
expression profiles of pancreatic cancer and stromal desmoplasia. Oncogene
2001;
20: 7437–7446.
18 Kimmelman AC, Hezel AF, Aguirre AJ, Zheng H, Paik JH, Ying H et al. Genomic
alterations link Rho family of GTPases to the highly invasive phenotype of
pancreas cancer. Proc Natl Acad Sci USA 2008; 105: 19372–19377.
19 Ungefroren H, Groth S, Sebens S, Lehnert H, Gieseler F, Fa
¨
ndrich F. Differential
roles of Smad2 and Smad3 in the regulation of TGF-b1-mediated growth inhi-
bition and cell migration in pancreatic ductal adenocarcinoma cells: control by
Rac1. Mol Cancer 2011; 10: 67.
20 Menard RE, Mattingly RR. Cell surface receptors activate p21-activated kinase 1 via
multiple Ras and PI3-kinase-dependent pathways. Cell Signal 2003; 15:
1099–1109.
21 Wu R, Abramson AL, Symons MH, Steinberg BM. Pak1 and Pak2 are activated in
recurrent respiratory papillomas, contributing to one pathway of Rac1-mediated
COX-2 expression. Int J Cancer 2010; 127: 2230–2237.
22 He H, Shulkes A, Baldwin GS. PAK1 interacts with beta-catenin and is required for
the regulation of the beta-catenin signalling pathway by gastrins. Biochim Biophys
Acta 2008; 1783: 1943–1954.
23 Singh RR, Song C, Yang Z, Kumar R. Nuclear localization and chromatin targets of
p21-activated kinase 1. J Biol Chem 2005; 280: 18130–18137.
24 Sa
´
nchez-Solana B, Motwani M, Li DQ, Eswaran J, Kumar R. p21-activated kinase-1
signaling regulates transcription of tissue factor and tissue factor pathway
inhibitor. J Biol Chem 2012; 287: 39291–39302.
25 Frost JA, Swantek JL, Stippec S, Yin MJ, Gaynor R, Cobb MH. Stimulation of NF-kB
activity by multiple signaling pathways requires PAK1. J Biol Chem 2000; 275:
19693–19699.
26 Stanisavljevic J, Porta-de-la-Riva M, Batlle R, de Herreros AG, Baulida J. The p65
subunit of NF-kB and PARP1 assist Snail1 in activating fibronectin transcription.
J Cell Sci 2011; 124: 4161–4171.
27 Navas C, Herna
´
ndez-Porras I, Schuhmacher AJ, Sibilia M, Guerra C, Barbacid M.
EGF receptor signaling is essential for k-ras oncogene-driven pancreatic ductal
adenocarcinoma. Cancer Cell 2012; 22: 318–330.
28 Kichina JV, Goc A, Al-Husein B, Somanath PR, Kandel ES. PAK1 as a therapeutic
target. Expert Opin Ther Targets 2010; 14: 703–725.
29 Ong CC, Jubb AM, Zhou W, Haverty PM, Harris AL, Belvin M et al. p21-activ ated
kinase 1: PAK’ed with potential. Oncotarget 2011; 2: 491–496.
30 Ong CC, Jubb AM, Haverty PM, Zhou W, Tran V, Truong T et al. Targeting p21-
activated kinase 1 (PAK1) to induce apoptosis of tumor cells. Proc Natl Acad Sci
USA 2011; 108: 7177–7182.
31 Ye DZ, Field J. PAK signaling in cancer. Cell Logist 2012; 2: 105–116.
32 Linder S, Castan
˜
os-Velez E, von Rosen A, Biberfeld P. Immunohistochemical
expression of extracellular matrix proteins and adhesion molecules in pancreatic
carcinoma. Hepatogastroenterology 2001; 48: 1321–1327.
33 Mao Y, Schwarzbauer JE. Stimulatory effects of a three-dimensional micro-
environment on cell-mediated fibronectin fibrillogenesis. JCellSci2005; 118 :
4427–4436.
34 Holm C, Rayala S, Jirstro
¨
m K, Stål O, Kumar R, Landberg G. Association between
Pak1 expression and subcellular localization and tamoxifen resistance in breast
cancer patients. J Natl Cancer Inst 2006; 98: 671–680.
Supplementary Information accompanies this paper on the Oncogene website (http://www.nature.com/onc)
Pak1 signaling regulates fibronectin transcription
S Jagadeeshan et al
10
Oncogene (2014) 1 10 & 2014 Macmillan Publishers Limited
... However, the clinical and functional relevance of PAK1 and associated signaling remains undefined in myxofibrosarcoma. Compared with RSF1, PAK1, the prototypical member of p21-activating kinase family, is relatively feasible as a direct therapeutic target, since PAK1 functions as a cytosolic molecular hub converging upstream signals mediated by RhoGTPases to integrate various mitogenic and morphogenetic inputs and transcriptionally co-regulate expression of target genes upon nuclear relocation [12][13][14][15][16][17][18][19]. ...
... Apart from validating the canonical pro-proliferative and pro-metastatic functions of PAK1, we have characterized a hitherto undescribed PAK1/STAT5B/ CSF2 regulatory axis in myxofibrosarcomas, wherein the pro-angiogenic attribute of PAK1 offers a viable molecular target to inform risk stratification and therapeutic strategies. Although PAK1-associated oncogenesis is thought to classically depend on the interaction of PAK1 with RhoGTPases in the cytosol [13,17,19,34,35], PAK1 nuclear entry has emerged as a crucial mechanism to regulate expression of myriad downstream cancer-associated genes [12,13,18,19,36]. In our cell-based models, concomitant tyrosyl and threonine phosphorylation expedited the nuclear entry of activated PAK1, through which PAK1 interacted with nuclear STAT5B at the STAT5 binding site to co-transactivate the CSF2 promoter. ...
Article
Myxofibrosarcoma is genetically complex without established nonsurgical therapies. In public datasets, PAK1 was recurrently gained with mRNA upregulation. Using myxofibrosarcoma cells, we explored the oncogenic underpinning of PAK1 with genetic manipulation and a pan-PAK inhibitor (PF3758309). Myxofibrosarcoma specimens were analyzed for the levels of PAK1, phospho-PAKT423, CSF2 and microvascular density (MVD) and those of PAK1 gene and mRNA. PAK1-expressing xenografts were assessed for the effects of PF3758309 and CSF2 silencing. Besides pro-proliferative and pro-migrator/pro-invasive attributes, PAK1 strongly enhanced angiogenesis in vitro, which, not phenocopied by PAK2-4, was identified as CSF2-mediated using antibody arrays. PAK1 underwent phosphorylation at tyrosines153,201,285 and threonine423 to facilitate nuclear entry, whereby nuclear PAK1 bound STAT5B to co-transactivate the CSF2 promoter, increasing CSF2 secretion needed for angiogenesis. Angiogenesis driven by PAK1-upregulated CSF2 was negated by CSF2 silencing, anti-CSF2, and PF3758309. Clinically, overexpressed whole-cell phospho-PAKT423, related to PAK1 amplification, was associated with increased grades, stages, and PAK1 mRNA, higher MVD, and CSF2 overexpression. Overexpressed whole-cell phospho-PAKT423 and CSF2 independently portended shorter metastasis-free survival and disease-specific survival, respectively. In vivo, both CSF2 silencing and PF3758309 suppressed PAK1-driven tumor proliferation and angiogenesis. Conclusively, the nuclear entry of overexpressed/activated PAK1 endows myxofibrosarcomas with pro-angiogenic function, highlighting the vulnerable PAK1/STAT5B/CSF2 regulatory axis.
... The involvement of PAK1 in PDA tumorigenesis is characterised by its intricate and multifaceted nature. PAK1 interacts with the NFκB-p65 complex, thereby enhancing the transcriptional expression of fibronectin, causing cells to transform, and promoting the invasive epithelial-mesenchymal transition (EMT) [58]. PAK1 influences the production of VEGF, a growth factor that supports tumour invasion and metastasis [59,60]. ...
Article
Full-text available
Angiogenesis has been associated with numbers of solid tumours. Anti-angiogenesis drugs starve tumours of nutrients and oxygen but also make it difficult for a chemo reagent to distribute into a tumour, leading to aggressive tumour growth. Anti-angiogenesis drugs do not appear to improve the overall survival rate of pancreatic cancer. Vessel normalisation is merging as one of the new approaches for halting tumour progression by facilitating the tumour infiltration of immune cells and the delivery of chemo reagents. Targeting p21-activated kinases (PAKs) in cancer has been shown to inhibit cancer cell growth and improve the efficacy of chemotherapy. Inhibition of PAK enhances anti-tumour immunity and stimulates the efficacy of immune checkpoint blockades. Inhibition of PAK also improves Car-T immunotherapy by reprogramming the vascular microenvironment. This review summarizes current research on PAK’s role in tumour vasculature and therapeutical response, with a focus on pancreatic cancer.
... Several pathways have been identified to be involved in the transcriptional activation of fibronectin, such as the binding of the p21-activated kinase 1/NF-κB complex with the fibronectin promoter and the activation of MAPK in pancreatic cancer and hepatocellular carcinoma, respectively. 22,23 Moreover, EGF significantly induces the expression of fibronectin in triple-negative breast cancer cells. 24 The depletion of PDK1 or cyclooxygenase-2 also inhibited EGF-induced fibronectin expression and HNSCC cell metastasis. ...
Article
Full-text available
There is a higher expression level of epidermal growth factor receptor (EGFR) in up to 90% of advanced head and neck squamous cell carcinoma (HNSCC) tissue than in normal surrounding tissues. However, the role of RNA‐binding proteins (RBPs) in EGFR‐associated metastasis of HNSCC remains unclear. In this study, we reveal that RBPs, specifically nucleolin (NCL) and heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNPA2B1), correlated with the mesenchymal phenotype of HNSCC. The depletion of RBPs significantly attenuated EGF‐induced HNSCC metastasis. Intriguingly, the EGF‐induced EMT markers, such as fibronectin, were regulated by RBPs through the ERK and NF‐κB pathway, followed by the enhancement of mRNA stability of fibronectin through the 5′ untranslated region (5′‐UTR) of the gene. The upregulation of fibronectin triggered the integrin signaling activation to enhance tumor cells’ attachment to endothelial cells and increase endothelial permeability. In addition, the concurrence of EGFR and RBPs or EGFR and fibronectin was associated with overall survival and disease‐free survival of HNSCC. The in vivo study showed that depletion of NCL, hnRNPA2B1, and fibronectin significantly inhibited EGF‐promoted extravasation of tumor cells into lung tissues. The depletion of fibronectin or treatment with integrin inhibitors dramatically attenuated EGF‐induced HNSCC metastatic nodules in the lung. Our data suggest that the RBPs/fibronectin axis is essential for EGF‐induced tumor‐endothelial cell interactions to enhance HNSCC cell metastasis.
... Similarly, p21activated kinase 1 (Pak1) acts as an oncogene enhancing tumor formation by binding to fibronectin and interacting with the NF-κB-p65 complex in the Kras intact model. 370 Furthermore, NF-κB also modulates inflammatory macrophages through the direct regulation of growth and differentiation factor 15 (GDF-15)/macrophage inhibitory cytokine 1 (MIC-1), which is highly expressed in PC, 371 serving as a promoter of early carcinogenesis. Together, these observations support the critical roles of NF-κB in pancreatic carcinogenesis (Fig. 7). ...
Article
Full-text available
Research on obesity- and diabetes mellitus (DM)-related carcinogenesis has expanded exponentially since these two diseases were recognized as important risk factors for cancers. The growing interest in this area is prominently actuated by the increasing obesity and DM prevalence, which is partially responsible for the slight but constant increase in pancreatic cancer (PC) occurrence. PC is a highly lethal malignancy characterized by its insidious symptoms, delayed diagnosis, and devastating prognosis. The intricate process of obesity and DM promoting pancreatic carcinogenesis involves their local impact on the pancreas and concurrent whole-body systemic changes that are suitable for cancer initiation. The main mechanisms involved in this process include the excessive accumulation of various nutrients and metabolites promoting carcinogenesis directly while also aggravating mutagenic and carcinogenic metabolic disorders by affecting multiple pathways. Detrimental alterations in gastrointestinal and sex hormone levels and microbiome dysfunction further compromise immunometabolic regulation and contribute to the establishment of an immunosuppressive tumor microenvironment (TME) for carcinogenesis, which can be exacerbated by several crucial pathophysiological processes and TME components, such as autophagy, endoplasmic reticulum stress, oxidative stress, epithelial-mesenchymal transition, and exosome secretion. This review provides a comprehensive and critical analysis of the immunometabolic mechanisms of obesity- and DM-related pancreatic carcinogenesis and dissects how metabolic disorders impair anticancer immunity and influence pathophysiological processes to favor cancer initiation.
... CXCL ligands bind to specific G protein-coupled 7-transmembrane receptors (GPCRs), namely CXCRs, in order to exert downstream effects 13 . PAK1 is an effector protein downstream to a GPCR and facilitates many pro-tumour effects, such as metastasis and chemoresistance 238,[240][241][242][243] . 3.4.3A ...
Thesis
Full-text available
Chemoresistance is the major contributor to the low survival of pancreatic cancer (PC). PC progression is a complex process reliant on interactions between tumor and tumor microenvironment (TME). A family of structurally similar inflammatory chemokines, namely CXC ligands (CXCLs), were recently discovered to play important roles in various cancer types, including PC. This thesis aimed to investigate the role of CXCL5 in chemoresistance of PC. In both human and mice PC cell lines tested, CXCL5 expression was dramatically upregulated. The expressions of CXCL5, CXCL10 and selected CSC genes were various in gemcitabine resistant cell lines, and gemcitabine treated cells. However, in mouse xenografted tumor samples, which was generated from a patient-derived cell line, gemcitabine alone or in combination with other chemotherapeutic reagents led to increased CXCL5 protein level while CXCL10 level remained unchanged. These results suggested that expression of CXCL5 may be stimulated upon administration of gemcitabine or other chemotherapeutic reagents. Therefore, CXCL5 has a role in chemoresistance and clinical importance in PC; however, the mechanisms involved deserves a careful investigation. To determine whether CXCL5 mediates chemoresistance in PC, CXCL5 expression in MiaPaCa-2 cells was knocked down by shRNA. To determine whether CXCL5-mediated chemoresistance in vitro, two chemotherapeutic drugs, were used to treat a negative control (NC) and CXCL5 knockdown (KD) clones. In the cell proliferation assays, CXCL5 was found to mediate the resistance to gemcitabine and 5-fluouracil (5-FU). Mice carrying xenografted tumors inoculated by either NC or CXCL5 KD cells were treated with gemcitabine. CXCL5 KD suppressed tumor growth and enhanced the inhibitory effect of gemcitabine by decreasing proliferation and promoting apoptosis. These results indicated that knockdown of CXCL5 sensitized PC cells in response to gemcitabine and 5-FU, suggesting that CXCL5 mediates chemoresistance in PC. Finally, the global proteomic analysis showed CXCL5 knockdown resulted in significant changes in expression of several proteins. Each of these proteins had a distinct biological function in cancer as determined with KEGG pathway analysis and NCBI. From the phospho-proteomic analysis, CXCL5 knockdown induced significant changes of certain phosphorylated proteins. Cross-referencing with the database of NCBI clearly identified the biological functions of these proteins. Although experimental and clinical validation are necessary, CXCL5 serves as a pivotal molecular target in overcoming chemoresistance and eliminating PC tumors in clinical practices. In summary, these studies have revealed that CXCL5 plays an important role in chemoresistance and activates several intracellular pathways that contribute to resistance to therapeutic treatments and PC progression. Therefore, CXCL5 could serve as a potential molecular target in reversing chemoresistance in pancreatic cancer.
... PAK1 plays a functional role in the Met receptor tyrosine kinase-induced pancreatic adenocarcinoma growth and metastasis [84]. PAK1 modulates pancreatic cancer cell transformation as well as an invasive EMT phenotype via the NF-κB/p65/fibronectin pathway [83]. PAK4 interacts with p85α, which is a subunit of PI3K, and promotes PDAC cell motility by positively stimulating the AKT activity that is downstream of HGF [139]. ...
Article
Full-text available
Gastrointestinal tumors are the most common tumors, and they are leading cause of cancer deaths worldwide, but their mechanisms are still unclear, which need to be clarified to discover therapeutic targets. p21-activating kinase (PAK), a serine/threonine kinase that is downstream of Rho GTPase, plays an important role in cellular signaling networks. According to the structural characteristics and activation mechanisms of them, PAKs are divided into two groups, both of which are involved in the biological processes that are critical to cells, including proliferation, migration, survival, transformation and metabolism. The biological functions of PAKs depend on a large number of interacting proteins and the signaling pathways they participate in. The role of PAKs in tumors is manifested in their abnormality and the consequential changes in the signaling pathways. Once they are overexpressed or overactivated, PAKs lead to tumorigenesis or a malignant phenotype, especially in tumor invasion and metastasis. Recently, the involvement of PAKs in cellular plasticity, stemness and the tumor microenvironment have attracted attention. Here, we summarize the biological characteristics and key signaling pathways of PAKs, and further analyze their mechanisms in gastrointestinal tumors and others, which will reveal new therapeutic targets and a theoretical basis for the clinical treatment of gastrointestinal cancer.
... However, recent studies refined the variety of stromal cellular components such as CAF, and their abilities to potentially support or restrain PDAC progression 1,5,6,36,37 . This knowledge highlighted the necessity to precisely identify and target stromal cues and mechanisms that underlie the pro-tumoral intercellular communication between CAF and pancreatic cancer cells 24,41 . These results strongly suggest a transactivation mechanism of EGFR, that has been previously reported as integrinand FAK-related in PDAC 26,42 . ...
Article
Full-text available
Objective Intercellular communication within pancreatic ductal adenocarcinoma (PDAC) dramatically contributes to metastatic processes. The underlying mechanisms are poorly understood, resulting in a lack of targeted therapy to counteract stromal-induced cancer cell aggressiveness. Here, we investigated whether ion channels, which remain understudied in cancer biology, contribute to intercellular communication in PDAC. Design We evaluated the effects of conditioned media from patient-derived cancer-associated fibroblasts (CAFs) on electrical features of pancreatic cancer cells (PCC). The molecular mechanisms were deciphered using a combination of electrophysiology, bioinformatics, molecular and biochemistry techniques in cell lines and human samples. An orthotropic mouse model where CAF and PCC were co-injected was used to evaluate tumour growth and metastasis dissemination. Pharmacological studies were carried out in the Pdx1-Cre, Ink4a fl/fl LSL - Kras G12D (KIC pdx1 ) mouse model. Results We report that the K ⁺ channel SK2 expressed in PCC is stimulated by CAF-secreted cues (8.84 vs 2.49 pA/pF) promoting the phosphorylation of the channel through an integrin–epidermal growth factor receptor (EGFR)–AKT (Protein kinase B) axis. SK2 stimulation sets a positive feedback on the signalling pathway, increasing invasiveness in vitro (threefold) and metastasis formation in vivo. The CAF-dependent formation of the signalling hub associating SK2 and AKT requires the sigma-1 receptor chaperone. The pharmacological targeting of Sig-1R abolished CAF-induced activation of SK2, reduced tumour progression and extended the overall survival in mice (11.7 weeks vs 9.5 weeks). Conclusion We establish a new paradigm in which an ion channel shifts the activation level of a signalling pathway in response to stromal cues, opening a new therapeutic window targeting the formation of ion channel-dependent signalling hubs.
Article
Pancreatic cancer is a lethal malignancy for which there is currently no effective treatment strategy. We previously reported that p21-activated kinase 1 (PAK1) is aberrantly expressed in pancreatic cancer patients and that targeted inhibition of PAK1 significantly suppressed pancreatic cancer progression in vitro and in vivo. In this study, we identified the drug azeliragon as a novel inhibitor of PAK1. Cell experiments revealed that azeliragon abolished PAK1 activation and promoted apoptosis in pancreatic cancer cells. Azeliragon was also found to significantly inhibit tumor growth in a pancreatic cancer xenograft model; when combined with afuresertib, an oral pan-AKT kinase inhibitor, azeliragon exhibited a strong synergistic effect against pancreatic cancer cells. Interestingly, afuresertib enhanced the antitumor efficacy of azeliragon in a xenograft mouse model. Collectively, our findings revealed previously unreported aspects of the drug azeliragon, and identified a novel combination strategy for the treatment of pancreatic cancer patients.
Article
Full-text available
Transformation of a normal cell to a cancer cell is caused by mutations in genes that regulate proliferation, apoptosis, and invasion. Small GTPases such as Ras, Rho, Rac and Cdc42 orchestrate many of the signals that are required for malignant transformation. The p21-activated kinases (PAKs) are effectors of Rac and Cdc42. PAKs are a family of serine/threonine protein kinases comprised of six isoforms (PAK1-6), and they play important roles in cytoskeletal dynamics, cell survival and proliferation. They act as key signal transducers in several cancer signaling pathways, including Ras, Raf, NFκB, Akt, Bad and p53. Although PAKs are not mutated in cancers, they are overexpressed, hyperactivated or amplified in several human tumors and their role in cell transformation make them attractive therapeutic targets. This review discusses the evidence that PAK is important for cell transformation and some key signaling pathways it regulates. This review primarily discusses Group I PAKs (PAK1, PAK2 and PAK3) as Group II PAKs (PAK4, PAK5 and PAK6) are discussed elsewhere in this issue (by Minden).
Article
Full-text available
Tissue factor (TF) is a cell-surface glycoprotein responsible for initiating the coagulation cascade. Besides its role in homeostasis, studies have shown the implication of TF in embryonic development, cancer related events, and inflammation via coagulation-dependent and -independent (signaling) mechanisms. Tissue factor pathway inhibitor (TFPI) plays an important role in regulating TF-initiated blood coagulation. Therefore, transcriptional regulation of TF expression and its physiologic inhibitor TFPI, would allow us to understand the critical step that control many different processes. From a gene profiling study aimed to identify differentially regulated genes between wild type (WT) and p21-activated kinase 1-null (PAK1-KO) mouse embryonic fibroblasts (MEFs), we found TF and TFPI are differentially expressed in the PAK1-KO MEFs in comparison to wild-type MEFs. Based on these findings, we further investigated in the present study the transcriptional regulation of TF and TFPI by PAK1, a serine/threonine kinase. We found that PAK1/c-Jun complex stimulates the transcription of TF and consequently, its procoagulant activity. Moreover, PAK1 negatively regulates the expression of TFPI and thus, additionally, contributes to increased TF activity. For the first time, this study implicates PAK1 in coagulation processes, through its dual transcriptional regulation of TF and its inhibitor.
Article
Full-text available
Pancreatic ductal adenocarcinoma (PDA) remains a devastating disease with nearly equal incidence and mortality rates. Over the past few decades, a litany of randomized clinical trials has failed to improve the outcome of this disease. More recently, the combination chemotherapy regimen FOLFIRINOX has shown improvement in overall survival over the single agent gemcitabine, and nab-paclitaxel (an albumin-coated formulation of paclitaxel) in combination with gemcitabine has shown promising results in phase II studies. Despite limited impact on patient care as of yet, the molecular and biologic understanding of PDA has advanced substantially. This includes understanding the genomic complexity of the disease, the potential importance of the tumor microenvironment, the metabolic adaptation of PDA cells to obtain nutrients in a hypoxic environment, and the role of pancreatic cancer stem cells. These fundamental discoveries are starting to be translated into clinical studies. In this overview, we discuss the implications of biologic understanding of PDA in clinical research and provide insights for future development of novel approaches and agents in this disease.
Article
Pancreatic cancer is a highly malignant tumor, and is difficult to be diagnosed in its early stage. microRNAs (miRNAs) are small single-stranded RNA molecules of 19-23 nucleotides and have been recognized as a post-transcriptional negative regulator of the expression of target genes, thereby affecting the development, differentiation and apoptosis of tumor cells. Aberrant miRNAs expression patterns have been described in recent studies in tumor tissue, cell line, precancerous tissue and plasma of pancreatic cancer, which suggest that miRNAs might have some value in mechanism research on the genesis and progress, diagnosis, treatment and prognostic assessment of pancreatic cancer. This article reviewed recent progress in related field.
Article
The past two decades have witnessed an explosion in Our understanding of pancreatic cancer, and it is now clear that pancreatic cancer is a disease of inherited (germ-line) and somatic gene mutations. The genes mutated in pancreatic cancer include KRAS2, p16/CDKN2A, TP53, and SMAD4/DPC4, and these are accompanied by a substantial compendium of genomic and transcriptomic alterations that facilitate cell cycle deregulation, cell survival, invasion, and metastases. Pancreatic cancers do not arise de novo, and three distinct precursor lesions have been identified. Experimental models of pancreatic cancer have been developed in genetically engineered mice, which recapitulate the multistep progression of the cognate human disease. Although the putative cell of origin for pancreatic cancer remains elusive, minor populations of cells with stem-like properties have been identified that appear responsible for tumor initiation, metastases, and resistance of Pancreatic cancer to conventional therapies.
Article
p21-activated kinases (PAKs) were the first identified mammalian members of a growing family of Ste20-like serine-threonine protein kinases. In this study, we show that PAK1 can be stimulated by carbachol, lysophosphatidic acid (LPA), epidermal growth factor (EGF), and phorbol 12-myristate 13-acetate (PMA) by multiple independent and overlapping pathways. Dominant-negative Ras, Rac, and Cdc42 inhibited PAK1 activation by all of these agonists, while active Rac1 and Cdc42 were sufficient to maximally activate PAK1 in the absence of any treatment. Active Ras induced only a weak activation of PAK1 that could be potentiated by muscarinic receptor stimulation. Studies using inhibitors of the EGF receptor tyrosine kinase, phosphatidylinositol 3-kinase (PI3-kinase) and protein kinase C (PKC) revealed that all of the cell surface agonists could activate PAK1 through pathways independent of PKC, that EGF stimulated a PI3-kinase dependent pathway to stimulate PAK1, and that muscarinic receptor stimulation of PAK1 was predominantly mediated through this EGF-R-dependent mechanism. Activation of PAK1 by LPA was independent of PI3-kinase and the EGF receptor, but was inhibited by dominant-negative RhoA. These results identify multiple Ras-dependent pathways to activation of PAK1.
Article
Clinical evidence indicates that mutation/activation of EGF receptors (EGFRs) is mutually exclusive with the presence of K-RAS oncogenes in lung and colon tumors. We have validated these observations using genetically engineered mouse models. However, development of pancreatic ductal adenocarcinomas driven by K-Ras oncogenes are totally dependent on EGFR signaling. Similar results were obtained using human pancreatic tumor cell lines. EGFRs were also essential even in the context of pancreatic injury and absence of p16Ink4a/p19Arf. Only loss of p53 made pancreatic tumors independent of EGFR signaling. Additional inhibition of PI3K and STAT3 effectively prevented proliferation of explants derived from these p53-defective pancreatic tumors. These findings may provide the bases for more rational approaches to treat pancreatic tumors in the clinic.