Abstract
The myofibroblast (MFB) has recently been identified as an important mediator of tumor necrosis factor-α (TNF-α)-associated colitis and cancer, but the mechanism(s) involved remains incompletely understood. Here, we show that treatment of 18Co cells, a model of human colonic MFBs, with TNF-α and lysophosphatidic acid (LPA) induced striking synergistic cyclooxygenase-2 (COX-2) protein expression and production of PGE2. This effect was prevented by the LPA1 receptor antagonist Ki16425, the Giα-specific inhibitor pertussis toxin, and by the preferential protein kinase (PK) C inhibitors GF109203X and Go6983. As a known downstream target of LPA and PKC, we tested whether PKD, recently implicated in the regulation of COX-2 expression in MFB, was involved in this response. TNF-α, while having no detectable effect on the activation of PKD when added alone, augmented PKD activation stimulated by LPA, as measured by PKD autophosphorylation at Ser910. LPA-induced PKD activation was also inhibited by Ki16425, pertussis toxin, GF109203X, and Go6983. Transfection of 18Co cells with short interfering RNA targeting PKD completely inhibited the synergistic increase in COX-2 protein, demonstrating a critical role of PKD in this response. Our results imply that cross talk between TNF-α and LPA results in the amplification of COX-2 protein expression via a conserved PKD-dependent signaling pathway that appears to involve the LPA1 receptor and the G protein Giα. PKD plays a critical role in the expression of COX-2 in human colonic MFBs and may contribute to an inflammatory microenvironment that promotes tumor growth.
Keywords: tumor necrosis factor, protein kinase D, colitis-associated cancer
myofibroblasts are a subpopulation of stromal cells located in the lamina propria of the gastrointestinal (GI) tract that interact with neighboring cells in a paracrine fashion through the secretion of cytokines, growth factors, and inflammatory mediators, including prostanoids (36, 62). Through these interactions, myofibroblasts regulate multiple biological processes, including epithelial proliferation and differentiation, apoptosis, migration, mucosal repair, and fibrosis (21, 36). They also participate in immune and inflammatory responses (39) and have been implicated in the underlying pathophysiology that characterizes both ulcerative colitis and colorectal cancer (16, 21, 36, 45).
Myofibroblasts are a major source of the inducible isoform of cyclooxygenase (COX-2) (2, 21, 24, 32, 43, 62), the rate-limiting enzyme in arachidonate metabolism that catalyzes the biosynthesis of prostaglandin (PG) H2, the precursor of prostanoids, including PGs and thromboxanes (25). Substantial evidence demonstrates that PGs play an important role in both inflammation and cancer in the GI tract (4, 16, 25, 45, 57a). Interestingly, increased expression of myofibroblasts, in parallel with elevated levels of COX-2, are seen in the stroma underlying colonic adenomas and carcinomas (1, 2, 5, 21, 43, 52, 58). Therefore, understanding the regulation of COX-2 expression in intestinal myofibroblasts may provide insights into the pathogenesis of inflammation-associated cancer in the gut. Despite their importance, the cell signaling pathways that regulate myofibroblast function, particularly in the setting of inflammation, remain incompletely understood.
TNF-α is a 17-kDa proinflammatory cytokine that plays a pivotal role in regulating the inflammatory signaling cascades characteristic of ulcerative colitis (3) and has been strongly implicated in colitis-associated cancer (17, 35). Binding of TNF-α to its receptors, TNF-α receptor 1 (TNFR1) and TNF-α receptor 2 (TNFR2), triggers the formation of a multiprotein complex (TNFR1-associated death domain protein, receptor interacting protein, TNFR-associated factor 2) that initiates downstream signaling via phosphorylation cascades that culminate in the activation of mitogen-activated protein kinases and the transcription factor NF-κB (reviewed in Ref. 53). Myofibroblasts have recently been identified as an important mediator of TNF-α-associated colitis, but the precise mechanism(s) involved remains incompletely understood (3). The ability of a single antibody targeted against TNF-α to induce clinical remission in patients with Crohn's disease and ulcerative colitis suggests that TNF-α is a central regulator of multiple inflammatory signaling pathways that could promote the development of colitis-associated cancer.
Lysophosphatidic acid (LPA) is a phospholipid that initiates signaling cascades involved in the inflammatory response and may be a potential target for cross talk with TNF-α. LPA is involved in diverse biological processes, including wound healing, cell migration and contraction, vascular remodeling, tumor progression, and cytokine and matrix metalloproteinase production (28). LPA-mediated signaling occurs via specific LPA receptors (LPA1–5) and at least three different G protein subfamilies (Gαq/11, Gi/o, G12/13). The main LPA-producing phospholipase is autotaxin, a member of the nucleotide pyrophosphatase/phosphodiesterase family of ectoenzymes, and a widely expressed autocrine motility factor that is identical to lysophospholipase D. Autotaxin is secreted by adipocytes (13) and connective tissue-type mast cells (49) and is present in the small and large intestine (13, 49), localized to the submucosal layer. Here, mast cells can provide locally bioactive LPA in high concentration (49), where it has effects on the overlying mucosa, and on surrounding mesenchymal cells (e.g., myofibroblasts) of the lamina propria. Autotaxin is overexpressed in many human cancers and has been associated with tumor progression and angiogenesis (28).
Protein kinase D (PKD) is the founding member of a new protein kinase family (38) that lies downstream of protein kinase C (PKC) isoforms in a phosphorylation cascade (18, 57, 67, 68), implicated in the regulation of a variety of biological responses, including inflammation (9, 62), oxidative stress (47, 48, 55, 56), cell proliferation (45, 46, 66), and carcinogenesis (7, 14, 20, 29, 34, 40). While little is known regarding the regulation and biological function of PKD in intestinal myofibroblasts, recent work in our laboratory has implicated PKD as an important mediator of TNF-α-associated COX-2 expression (62).
In the present study, we show synergistic effects between LPA and TNF-α in cultures of human colonic myofibroblasts (18Co cells), leading to dramatic COX-2 expression, PGE2 production, and PKC/PKD axis activation via the LPA1 receptor and the Giα protein subunit. The results presented here demonstrate unique cross talk interactions between TNF-α and Giα-mediated LPA signaling that result in striking amplification of COX-2 expression through a conserved PKD-dependent mechanism in colonic myofibroblasts.
MATERIALS AND METHODS
Cell culture.
18Co cells (CRL-1459) were purchased from American Type Culture Collection (Rockville, MD). These cells share many of the structural and functional characteristics of in situ colonic subepithelial myofibroblasts, including a reversible stellate morphology, α-smooth actin expression, and the presence of multiple cell surface receptors (51). 18Co cells provide a model to elucidate physiological and pathophysiological functions of intestinal subepithelial myofibroblasts and, accordingly, have been used extensively to study colonic myofibroblast function in a variety of settings (22, 33, 42). 18Co cells were maintained at 37°C in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum (FBS) in a humidified atmosphere containing 10% CO2 and 90% air. For experimental purposes, cells were plated in 35-mm dishes (1 × 105 cells/dish) and grown in DMEM containing 10% FBS for 5–7 days until confluent and used from passages 8–14.
Immunoblotting and detection of PKD and COX-2.
Confluent 18Co cells, treated with different agonists, antagonists, inhibitors, and short interfering RNA (siRNA), as indicated in the individual experiments, were lysed in 2× SDS-polyacrylamide gel electrophoresis (PAGE) sample buffer (20 mM Tris·HCl, pH 6.8, 6% SDS, 2 mM EDTA, 4% 2-mercaptoethanol, 10% glycerol) and boiled for 10 min. After SDS-PAGE, proteins were transferred to Immobilon-P membranes. The transfer was carried out at 100 V, 0.4 A at 4°C for 5 h using a Bio-Rad transfer apparatus. The transfer buffer consisted of 200 mM glycine, 25 mM Tris, 0.01% SDS, and 20% CH3OH. For detection of proteins, membranes were blocked using 5% nonfat dried milk in phosphate-buffered saline (pH 7.2) and then incubated for 2 h with the desired antibodies diluted in phosphate-buffered saline (pH 7.2) containing 3% nonfat dried milk. Primary antibodies bound to immunoreactive bands were visualized by enhanced chemiluminescence detection with horseradish peroxidase-conjugated anti-mouse or anti-rabbit antibodies (GE Healthcare, Piscataway, NJ). The phospho-PKD polyclonal antibody pSer910 (Millipore, Billerica, MA) detects PKD when it is phosphorylated on Ser910, shown in previous reports.
ELISA.
PGE2 was quantified from the supernatant of serum-starved, confluent 18Co cells following treatment conditions, according to EIA kit instructions (Prostaglandin E2 EIA kit, Cayman Chemical, Ann Arbor, MI). The collected supernatant was centrifuged at 5,000 g for 5 min to remove cell debris. Absorbance readings were set between 405 and 420 nm on a spectrophotometer.
PKD siRNA transfection.
The SMART pool PKD siRNA duplexes were purchased from Dharmacon (Lafayette, CO). The PKD siRNA pool was designed to target against the mRNA of human PKD (NM_002742) and consists of four selected siRNA oligonucleotides. The sequences were as follows: oligo 1, CGGCAAAUGUAGUGUAUUAUU; oligo 2, GAACCAACUUGCACAGAGAUU; oligo 3, GGUCUGAAUUACCAUAAGAUU; oligo 4, GGAGAUAGCCAUCCAGCAUUU. siCONTROL nontargeting siRNA no. 3 (D-001210-03-20) was used as the control. 18Co cells were plated at ∼70–80% confluency in a 12-well plate with DMEM supplemented with 10% FBS and 1% antibiotic/antimycotic at 37°C in a humidified atmosphere containing 10% CO2. After 24 h, each well was replaced with 400 μl DMEM + 10% FBS (no antibiotic). Added to this was a mixture containing the Mirus TKO-IT transfection agent and PKD siRNA or control nontargeting siRNA (total volume: 500 μl per well, total transfection agent: 4 μl per well, siRNA: 50 nM). After incubation for 72 h, cells were used for experiments and subsequently analyzed by Western blot.
Materials.
Bradykinin (BK) and the PKC inhibitor GF109203X were purchased from Sigma (St. Louis, MO). TNF-α was purchased from R&D Systems (Minneapolis, MN). COX-2 antibody was purchased from Cell Signaling Technology (Beverly, MA). The PKC inhibitor Go6983, pertussis toxin (PTx), SB-202190, and U-0126 were purchased from Calbiochem (La Jolla, CA). LPA was purchased from both Sigma (St. Louis, MO) and Cayman Chemical (Ann Arbor, MI). The LPA receptor inhibitor Ki16425 was purchased from Cayman Chemical (Ann Arbor, MI). PKD siRNA was purchased from Dharmacon (Lafayette, CO). α-Smooth muscle actin antibody was purchased from Abcam (Cambridge, MA).
RESULTS
LPA and TNF-α lead to synergistic COX-2 expression in 18Co cells.
To determine whether the proinflammatory mediators LPA and TNF-α regulate COX-2 expression in human colonic myofibroblasts, 18Co cells were treated with LPA or TNF-α, either alone or in combination, over 24 h, and the level of COX-2 protein expression was assessed by Western blot analysis. There was no detectable COX-2 protein in unstimulated 18Co cells. Treatment of 18Co cells with 10 μM LPA induced minimal COX-2 protein expression over the 24-h time period studied (Fig. 1A). Treatment with TNF-α (8 ng/ml) alone stimulated levels of COX-2 expression that were slightly higher than that in the LPA-treated cells. This effect reached a peak after 4 h, but was not detectable after 24 h. However, when myofibroblasts were treated with LPA following an overnight incubation with TNF-α, this led to a synergistic time-dependent increase in COX-2 protein expression that was evident after 2 h and was dramatically augmented between 4 and 8 h, followed by a decline over the 24-h time period studied (Fig. 1A).
Fig. 1.
Lysophosphatidic acid (LPA) and TNF-α lead to synergistic cyclooxygenase (COX)-2 expression and PGE2 production in 18Co cells. A: confluent 18Co cells were washed and equilibrated in serum-free media for 30 min, followed by treatment with 8 ng/ml TNF-α, 10 μM LPA, or both for various times (2, 4, 8, and 24 h, as indicated). Lysates of these cells were then analyzed by Western blot using a polyclonal antibody that detects COX-2 protein. Autoluminograms were quantified by densitometric scanning. The results shown are means ± SE, n = 3, and are expressed as percentage of the maximum level of COX-2 expression, which correlated with a 48-fold increase over control. Equal protein loading was verified using an antibody that detects ERK-2 and α-smooth muscle actin (α-SMA). B: cultures of confluent 18Co cells were incubated in serum-free medium with 10 μM LPA (medium shaded bars), 8 ng/ml TNF-α (TNF, light shaded bars), or both (TNF+LPA, dark shaded bars) for various times (2, 4, 8, and 24 h, as indicated), with untreated cells (open bar) serving as a control. PGE2 released into the medium was quantified by ELISA.
To determine whether the induction of COX-2 protein expression by LPA and TNF-α was associated with an increase in PGE2 production, 18Co cells were again treated with LPA and TNF-α, alone and in combination, over 24 h. Cell culture supernatant was collected at various times, and PGE2 levels were quantified utilizing an ELISA. As shown in Fig. 1B, treatment of 18Co cells with LPA stimulated low levels of PGE2 production, which correlated with the modest increase in COX-2 expression seen by Western blot analysis. Treatment with TNF-α over 24 h produced a greater effect compared with that in LPA-treated cells, although the effect was also modest. However, exposure of 18Co cells to TNF-α for 18 h followed by LPA induced a synergistic increase in the production of PGE2 that was evident after 8 h and was sustained over a 24-h time period. Thus the proinflammatory mediators LPA and TNF-α induced synergistic COX-2 protein expression that was paralleled by increases in the production of PGE2 in human colonic myofibroblasts.
Since the augmented expression of COX-2 protein was most apparent after 4 h, we utilized this early time period to perform an additional time course experiment to fully characterize the timing of COX-2 protein expression (Fig. 2A). 18Co cells were pretreated overnight with TNF-α, followed by treatment with LPA over 4 h, and COX-2 expression was analyzed by Western blot. Under these conditions, the upregulation of COX-2 was evident after 2 h, with a steady increase in protein expression between 2 and 4 h. Based on this experiment, 18Co cells were treated with LPA for 4 h for all subsequent experiments.
Fig. 2.
LPA-induced COX-2 protein expression is augmented by prior exposure to TNF-α. A: confluent 18Co cells were washed and equilibrated in serum-free media for 30 min, incubated with 8 ng/ml TNF-α for 18 h, and then stimulated with LPA (10 μM) for various times (0–4 h as indicated). Lysates were then analyzed by SDS-PAGE and Western blot using a polyclonal antibody that detects COX-2 protein. The results shown are means ± SE, n = 3, and are expressed as percentage of the maximum level of COX-2 expression, which correlated with a 4.3-fold increase over control. Equal protein loading was verified using an antibody that detects ERK-2. *Statistical significance (P < 0.05). B: 18Co cells were treated with TNF-α overnight at various concentrations (0–80 ng/ml), followed by treatment with LPA (10 μM) for 4 h. Lysates were analyzed by SDS-PAGE and Western blot using a polyclonal antibody that detects COX-2 protein. The results shown are means ± SE, n = 3, and are expressed as percentage of the maximum level of COX-2 expression, which correlated with a 24.4-fold increase over control. Equal protein loading was verified using an antibody that detects α-SMA. C: confluent 18Co cells were washed and equilibrated in serum-free media for 30 min, incubated with 8 ng/ml TNF-α for 18 h, and then stimulated with LPA for 4 h at varying concentrations (0–30 μM). Lysates were then analyzed by SDS-PAGE and Western blot using a polyclonal antibody that detects COX-2 protein. The results shown are means ± SE, n = 3, and are expressed as percentage of the maximum level of COX-2 expression, which correlated with a 4.5-fold increase over control. Equal protein loading was verified using an antibody that detects ERK-2. *Statistical significance (P < 0.05).
The effect of LPA and TNF-α on the synergistic expression of COX-2 was dose dependent, as seen in Fig. 2, B and C. In Fig. 2B, 18Co cells were treated overnight with TNF-α at varying concentrations (0–80 ng/ml), followed by treatment with LPA (10 μM) for 4 h. Increases in COX-2 protein expression were evident at a TNF-α concentration as low as 0.8 ng/ml, well within physiological levels, and persisted at a concentration of 80 ng/ml. Based on this data, subsequent experiments were performed using TNF-α at a concentration of 8 ng/ml, well within the range of concentrations used in other studies (12, 26, 41, 61). In Fig. 2C, 18Co cells were treated overnight with TNF-α, followed by treatment with LPA for 4 h at varying concentrations (0–30 μM). Treatment with LPA for 4 h following an overnight incubation with TNF-α led to increases in COX-2 expression that were evident at a concentration of 1 μM LPA and increased in a dose-dependent manner over the range of concentrations studied.
LPA-mediated PKD activation is augmented by overnight treatment with TNF-α.
Previous work in our laboratory identified PKD as a central mediator of COX-2 expression in human colonic myofibroblasts (62), and LPA has been shown to induce PKD activation in several cell types (8, 9, 64). To evaluate a possible role of PKD in the synergistic expression of COX-2 in colonic myofibroblasts in response to LPA and TNF-α, we first tested whether LPA induces PKD activation in these cells. Confluent 18Co cells were treated with LPA for various times and lysed. Activation of PKD was assessed by Western blot analysis of the cell lysates using site-specific primary antibodies that detect the phosphorylated state of Ser738 located in the activation loop of human PKD (18, 54) and the auto-phosphorylated state of PKD on Ser910 (63). Treatment of 18Co cells with LPA led to a rapid and transient activation of PKD, as judged by phosphorylation on Ser738 (data not shown) and Ser910 (Fig. 3). Autophosphorylation at Ser910 peaked at 15–30 min, with a steady decline over the 4-h time period studied.
Fig. 3.
LPA-induced protein kinase D (PKD) activation is augmented by prior cell treatment with TNF-α. A: 18Co cells were incubated in the presence or absence of TNF-α (8 ng/ml) for 18 h, followed by stimulation with LPA (10 μM) over various times (15, 30, 60, 120, 240 min, as indicated). The cells were analyzed by Western blotting using an antibody that detects the COOH-terminal autophosphorylation site of PKD (Ser910 in human PKD). Band intensity was analyzed by densitometric scanning of three independent experiments and is depicted in graphical form on the bottom and on the right, presented as means ± SE, and expressed as a percentage of the maximum level of phosphorylated Ser910, which correlated with a 5.6-fold increase over control. Equal protein loading was verified using an antibody that detects ERK-2. *Statistical significance (P < 0.05).
We next determined whether TNF-α enhanced the intensity and/or duration of LPA-mediated PKD activation in colonic myofibroblasts. Confluent 18Co cells were treated with TNF-α overnight, followed by treatment with LPA, and PKD activation was assessed by Western blot. In agreement with our laboratory's previous results (62), TNF-α alone did not induce any detectable increase in PKD phosphorylation in 18Co cells (data not shown). However, overnight pretreatment of 18Co cells with TNF-α enhanced LPA-mediated PKD activation at 15, 30, and 60 min (Fig. 3). These experiments demonstrate that TNF-α augments the intensity of LPA-mediated PKD activation in 18Co cells, while having no detectable effect on PKD activation in these cells when added alone.
PKD activation and synergistic COX-2 expression induced by LPA and TNF-α involves the LPA1 receptor and Giα.
The biological functions of LPA are mediated by a family of at least five different G protein-coupled receptors (GPCRs) (LPA1–5) (10). LPA1 was the first LPA receptor to be identified (28), and high mRNA levels are present in the colon and small intestine (27). The LPA1 receptor is also the most widely expressed LPA receptor in the human colon (10), highlighting its potential importance in LPA-mediated signaling in this organ. To determine which LPA receptor was involved in LPA-mediated PKD activation and synergistic COX-2 expression in the human colonic myofibroblast cell line 18Co, we utilized the inhibitory properties of 3-[4-(4-{[1-(2-chlorophenyl)ethoxl]carbonyl amino}-3-methyl-5-isoxazolyl) benzylsunfanyl] propanoic acid (Ki16425), an LPA receptor antagonist (LPA1 > LPA3 >>> LPA2), that is selective for LPA1 at the lowest concentrations. Utilizing these properties, confluent 18Co cells were treated with TNF-α overnight, followed by incubation with Ki16425 for 1 h at varying concentrations (0–5 μM), followed by stimulation with LPA for 4 h. PKD activation and COX-2 expression were assessed by Western blot. As shown in Fig. 4A and consistent with previous results (Fig. 1A), overnight treatment with TNF-α alone led to low levels of COX-2 expression, an effect that was augmented by the addition of LPA for 4 h. However, exposure to Ki16425 inhibited LPA-mediated COX-2 expression, beginning at the lowest concentration used (0.25 μM), implicating LPA1 as the main receptor involved in this response. The remaining low level of COX-2 expression following treatment with Ki16425 (0.25 μM) was equivalent to treatment with TNF-α alone, suggesting that Ki6425 had no effect on TNF-α-induced COX-2 expression. LPA-induced PKD activation was similarly inhibited to baseline levels, and this inhibition was maintained over the range of concentrations used. The profound inhibition by Ki6425 at the lowest concentrations used strongly suggested that LPA-mediated PKD activation and COX-2 expression involve signaling initiated through the LPA1 receptor, and not by the LPA receptors LPA2, LPA3, LPA4, and LPA5. These results also supported the hypothesis that PKD could mediate the cross talk between LPA and TNF-α in the synergistic expression of COX-2.
Fig. 4.
PKD activation and synergistic COX-2 expression involves the LPA1 receptor and Giα. A: 18Co cells were incubated for 18 h with TNF-α (8 ng/ml), then were pretreated with the LPA receptor inhibitor Ki16425 at various concentrations (0, 0.25, 0.5, 1.0, 5.0 μM) for 1 h, followed by treatment with LPA (10 μM) for 4 h. The cell extracts were analyzed by SDS-PAGE and Western blot using an antibody that detects PKD (Ser910) and a polyclonal antibody that detects COX-2 protein. Band intensity was analyzed by densitometric scanning of three independent experiments and is depicted in graphical form below, presented as means ± SE, and expressed as a percentage of the maximum level of COX-2 (which correlated with a 3.2-fold increase over control) and phosphorylated Ser910 (which correlated with a 4.4-fold increase over control). Equal protein loading was verified using an antibody that detects ERK-2. B: 18Co cells were incubated overnight for 18 h with TNF-α (8 ng/ml), then were pretreated with the Giα-specific inhibitor pertussis toxin (Ptx) at various concentrations (0, 1, 10, 50, 100 ng/ml, as indicated) for 2 h, followed by treatment with LPA (10 μM) for 4 h. The cells were lysed, and the extracts were analyzed by SDS-PAGE and Western blot using an antibody that detects the phosphorylated state of PKD (Ser910), and a polyclonal antibody that detects COX-2 protein. Band intensity was analyzed by densitometric scanning of three independent experiments and is depicted in graphical form below, presented as means ± SE, and expressed as a percentage of the maximum level of COX-2 (which correlated with a 2-fold increase over control) and phosphorylated Ser910 (which correlated with a 3.9-fold increase over control). Equal protein loading was verified using an antibody that detects ERK-2. C: 18Co cells were incubated overnight for 18 h with TNF-α (8 ng/ml), then were pretreated with the Giα-specific inhibitor Ptx at various concentrations (0, 1, 10, 50, 100 ng/ml, as indicated) for 2 h, followed by treatment with bradykinin (BK; 50 nM) for 4 h. The cells were lysed, and the extracts were analyzed by SDS-PAGE and Western blot using an antibody that detects the phosphorylated state of PKD (Ser910), and a polyclonal antibody that detects COX-2 protein. Band intensity was analyzed by densitometric scanning of three independent experiments and is depicted in graphical form, presented as means ± SE, and expressed as a percentage of the maximum level of COX-2 (which correlated with a 10.2-fold increase over control) and phosphorylated Ser910 (which correlated with a 10.3-fold increase over control). Equal protein loading was verified using an antibody that detects α-SMA.
Following LPA binding to one of its cognate receptors (LPA1–5), downstream signals are propagated via one of several G protein subunits (Gαq/11, Gi/o, G12/13). The LPA1 receptor is capable of coupling with the Giα, G12/13, and Gαq family of G proteins (10, 31). To determine which G protein subunit may be involved in the LPA-mediated activation of PKD and the expression of COX-2, we used the Giα-specific inhibitor PTx. PTx is an A/B type toxin that, upon entry into a cell, catalyzes the ADP-ribosylation of the α-subunit of G proteins, locking Giα in an inactive state on the cell surface. 18Co cells were treated overnight with TNF-α, followed by LPA for 4 h, with or without a 2-h pretreatment with PTx, at various concentrations (0–50 ng/ml). As shown in Fig. 4B, PTx inhibited both the activation of PKD and the synergistic increase in COX-2 expression following treatment with TNF-α and LPA in a dose-dependent fashion. These experiments demonstrate that LPA-mediated PKD activation and COX-2 expression signal predominantly through Giα and further support the hypothesis that PKD may play a critical role in the synergistic expression of COX-2 in colonic myofibroblasts.
Previous work in our laboratory demonstrated that the proinflammatory mediator BK also induced PKD activation and synergistic COX-2 expression in response to TNF-α in 18Co cells (62). However, BK-induced PKD activation and COX-2 expression signals predominantly through Gαq. To confirm the specificity of PTx on the inhibition of Giα, and to distinguish the upstream signaling mechanisms initiated by BK and LPA, 18Co cells were treated overnight with TNF-α, followed by BK for 4 h, with or without a 2-h pretreatment with PTx, at various concentrations (0–50 ng/ml). In stark contrast to its effect on LPA, the Giα-specific inhibitor PTx failed to inhibit BK-mediated COX-2 expression and BK-mediated PKD phosphorylation at all concentrations used (Fig. 4C). These data further supports the predominant role of Giα, and not other G proteins, in LPA-mediated COX-2 expression and PKD activation.
LPA-mediated PKD activation and synergistic COX-2 expression involves PKC.
Isoforms of the PKC family are major downstream targets of LPA-mediated signaling in other cell systems (8). Furthermore, both the LPA1 receptor and Giα propagate signaling cascades that can result in the activation of phospholipase C and its downstream target PKC (10). Therefore, we sought to determine the role of PKC, and possibly its downstream target PKD, in the synergistic expression of COX-2 in response to LPA and TNF-α in colonic myofibroblasts. Cultures of 18Co cells were incubated overnight with TNF-α, then were pretreated for 60 min with the preferential PKC inhibitors GF109203X (also known as bisindolylmaleimide I) at 5 μM or Go6983 (2.5 μM), before stimulation with LPA for 4 h. Cell treatment with either Go6983 or GF109203X completely inhibited both the augmented activation of PKD and the synergistic expression of COX-2 induced by LPA and TNF-α (Fig. 5A). In a parallel fashion, Go6983 and GF109203X also blocked LPA and TNF-α-mediated PGE2 production, as quantified by ELISA (Fig. 5B). These results suggest that PKC isoforms play an upstream role in both LPA-mediated PKD activation and in the synergistic enhancement of COX-2 expression and PGE2 production mediated by TNF-α and LPA.
Fig. 5.
LPA-induced PKD activation and synergistic COX-2 expression requires PKC activity. A: cultures of 18Co cells were incubated for 18 h with TNF-α (8 ng/ml), then were pretreated for 1 h with the preferential PKC inhibitors GF109203X (GF1, 5 μM) or Go6983 (Go, 2.5 μM), followed by treatment with LPA (10 μM) for 4 h. Cells were lysed, and cell extracts were analyzed by SDS-PAGE and Western blot using anti-COX-2 antibody and an antibody that detects the phosphorylated state of PKD (Ser910). Similar results were obtained in at least three independent experiments for each condition. The results shown are means ± SE and are expressed as percentage of the maximum level of COX-2 expression (which correlated with a 5.2-fold increase above control) and as a percentage of the maximum level of phosphorylated Ser910 (which correlated with a 4.5-fold increase above control). Equal protein loading was verified using an antibody that detects ERK-2. *Statistical significance (P < 0.05). B: cultures of 18Co cells were incubated for 18 h with TNF-α (8 ng/ml), then were pretreated for 1 h with the PKC inhibitors GF1 (5 μM) or Go (2.5 μM), followed by treatment with LPA (10 μM) for 4 h. PGE2 released into the supernatant was quantified by ELISA. *Statistical significance (P < 0.05). TL, TNF-α plus LPA.
Synergistic COX-2 expression induced by LPA and TNF-α involves PKD.
The stimulatory effects produced by TNF-α and LPA on COX-2 expression and PKD activation and the inhibitory effects on both responses by Ki16425, PTx, and the preferential PKC inhibitors GF109203X and Go6983 are all consistent with the hypothesis that PKD plays a critical role in mediating synergistic COX-2 expression in response to sequential cell exposure to TNF-α and LPA. To test this hypothesis directly, we determined whether siRNA-mediated knockdown of PKD protein in 18Co cells inhibits the synergistic expression of COX-2 in response to these proinflammatory mediators. After determining the optimal concentration of PKD siRNA (50 nM), conditions for transfection and incubation time (72 h) in 18Co cells, lysates of cells treated with targeting or nontargeting PKD siRNAs were analyzed by Western blotting to assess the level of PKD. As shown in Fig. 6A, siRNA targeting PKD produced a robust depletion of PKD protein in 18Co cells.
Fig. 6.
Synergistic COX-2 expression induced by LPA and TNF-α is mediated by PKD. A: 18Co cells were transfected with either nontargeting (NT) short interfering RNA (siRNA) or with PKD siRNA [targeting (T)] at 50 nM in the presence of 4 μl Mirus TKO-IT transfection agent for 72 h. The level of PKD protein was analyzed by Western blotting using the anti-PKD C-20 antibody. Antibody against ERK-2 was used to verify equal protein loading. Band intensity was analyzed by densitometric scanning and is presented as means ± SE, n = 3, and expressed as a percentage of the maximum level of PKD [which correlated with a 6.1-fold increase above control (Con)]. Equal protein loading was verified using an antibody that detects ERK-2. *Statistical significance (P < 0.05). B: 18Co cells were transfected with 50 nM siRNA T PKD or with a NT sequence also at 50 nM, as described in A, followed by incubation with TNF-α for 18 h and then stimulation with 10 μM LPA for 4 h. Cell lysates were analyzed by Western blotting using anti-COX-2 antibody. Antibody against ERK-2 was used to verify equal protein loading. Band intensity was analyzed by densitometric scanning and is presented as means ± SE, n = 3, and expressed as a percentage of the maximum level of COX-2 (which correlated with a 3-fold increase above Con). *Statistical significance (P < 0.05).
Having demonstrated that PKD protein can be strikingly depleted in 18Co cells via transfection with PKD siRNA, we next determined the role of PKD in mediating synergistic COX-2 expression in response to LPA and TNF-α. As shown in Fig. 6B, knockdown of PKD in 18Co cells completely prevented the synergistic increase in COX-2 protein induced by treatment with LPA and TNF-α. The results show that PKD plays a critical role in mediating synergistic expression of COX-2 protein induced by LPA and TNF-α in 18Co myofibroblasts.
DISCUSSION
Dynamic interactions that occur between the GI epithelium and its underlying stroma regulate important biological functions in both normal and pathological conditions. In the setting of chronic inflammation, a large body of evidence supports a critical role of the stromal microenvironment on the epithelial transformation to an invasive phenotype and the development of cancer. The mechanism(s) underlying colitis-associated cancer is initiated, in part, by the local production of proinflammatory mediators that are released by multiple cell populations, including myofibroblasts. Cellular responses to the inflammatory microenvironment lead to complex interactions between neighboring cells that induce epithelial genomic instability, leading to proliferative and antiapoptotic properties, tumor invasion, and metastasis (50).
Mounting evidence supports a causal association between LPA-mediated signaling and cancer progression (28), particularly in the context of colitis-associated cancer (23). The importance of LPA in the development and progression of cancer was supported by the discovery that autotaxin, an ectoenzyme known to promote tumor invasion and metastasis, acts by producing bioactive LPA extracellularly in the tumor microenvironment (27). LPA has been linked to ovarian, prostate, gastric, and colorectal cancer (10, 23, 44, 65), as well as to the production of COX-2 and PG secretion (23, 30) by stromal cells of the GI tract (59).
The present study identifies a possible mechanism for the association between LPA-mediated signaling, inflammation, and cancer development. We found synergistic effects between LPA and TNF-α, resulting in the amplification of COX-2 expression and PGE2 production through a conserved PKD-dependent signaling mechanism that appears to predominantly involve the LPA1 receptor and the Giα protein subunit, leading to augmented COX-2 expression in human colonic myofibroblasts.
The biological activity of LPA is tightly controlled by its production and degradation, but is also regulated by cross-talk signaling between other proinflammatory mediators, such as TNF-α. Evidence for cross talk between TNF-α and GPCR-mediated signaling exists (6, 15, 19, 62). Our laboratory previously identified signaling interactions between TNF-α and the specific GPCR BK, which also led to augmented COX-2 expression in colonic myofibroblasts through a PKD-dependent process (62). However, BK-mediated signaling involved a completely different cell surface receptor (the BK B2 receptor) and downstream G protein target (Gαq). Given the central role that TNF-α plays in GI enteropathies, such as Crohn's disease and ulcerative colitis, we speculate that a mechanism of action could involve cross talk between other inflammatory signaling pathways that lead to exaggerated signaling and an amplification of physiological responses. The present data confirm that TNF-α is capable of interacting with multiple proinflammatory GPCRs in human colonic myofibroblasts, despite the fact that they signal through unique cell surface membrane receptors and through alternative G protein families.
The synergistic expression of COX-2 by TNF-α and LPA appears to involve a signaling cascade initiated, in part, by the LPA1 receptor and the G protein Giα, which, in turn, activates phospholipase C, PKC, and PKD. This pathway implies that other molecules downstream of PKD may also be involved. In other cell types, including Swiss 3T3 fibroblasts, a prominent mechanism by which PKD mediates GPCR-induced signaling is by increasing the duration of MEK/ERK/90-kDa ribosomal S6 kinase activation (46), which is also one of the major pathways leading to COX-2 transcriptional activation (1, 45, 60). It is plausible that the ERKs play a role in relaying the downstream PKD signal. Also, despite the linear nature of the signaling cascade we have identified, we cannot rule out overlapping contributions by the different LPA receptors (LPA1–5) and their downstream G protein targets: Gαq/11, Gi/o, and G12/13 (64). This is supported by the fact that the putative LPA3-receptor-specific agonist (2S)-3-[(hydroxymercaptophosphinyl)oxy]-2-methoxypropyl ester, 9Z-octadecenoic acid, triethyl ammonium salt independently induced COX-2 expression following overnight treatment with TNF-α, though not through either PKD or Giα (data not shown). These findings confirm the redundancy in function between the different LPA receptors but through completely different signaling pathways, potentially providing a mechanism to maintain homeostasis, despite the loss of an upstream receptor or signaling protein.
While the exact mechanism of interaction between TNF-α and these various GPCRs remains unclear, to date, the common thread appears to be the involvement of PKD. Despite the differences in the initiating signaling events, interactions with TNF-α still led to the synergistic expression of COX-2 through a conserved pattern of activation involving PKC and PKD. The present study supports the notion that PKD plays a critical role in the regulation of COX-2 in human colonic myofibroblasts. Given the current limitations (i.e., side effects) with existing COX-2 inhibitors in the clinical treatment of patients with colonic adenomas or a genetic predisposition for colorectal cancer, the present findings support a role for PKD as a novel, and potentially more effective, target for COX-2 inhibition and cancer prevention.
GRANTS
This work was supported by National Institutes of Health Grants R0-1DK55003, R0-1DK56930, R21CA137292, and P30-DK41301 (to E. Rozengurt), and 1KO8DK085136-01A1 (to J. Yoo).
DISCLOSURES
No conflicts of interest, financial or otherwise, are declared by the author(s).
REFERENCES
- 1. Adegboyega PA, Mifflin RC, Di Mari JF, Saada JI, Powell DW. Immunohistochemical study of myofibroblasts in normal colonic mucosa, hyperplastic polyps, and adenomatous colorectal polyps. Arch Pathol Lab Med 126: 829–836, 2002 [DOI] [PubMed] [Google Scholar]
- 2. Adegboyega PA, Ololade O, Saada J, Mifflin R, DiMari JF, Powell DW. Subepithelial myofibroblasts express cyclooxygenase-2 in colorectal tubular adenomas. Clin Cancer Res 10: 5870–5879, 2004 [DOI] [PubMed] [Google Scholar]
- 3. Armaka M, Apostolaki M, Jacques P, Kontoyiannis DL, Elewaut D, Kollias G. Mesenchymal cell targeting by TNF as a common pathogenic principle in chronic inflammatory joint and intestinal diseases. J Exp Med 205: 331–337, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Castellone MD, Teramoto H, Williams BO, Druey KM, Gutkind JS. Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-β-catenin signaling axis. Science 310: 1504–1510, 2005 [DOI] [PubMed] [Google Scholar]
- 5. Chapple KS, Cartwright EJ, Hawcroft G, Tisbury A, Bonifer C, Scott N, Windsor ACJ, Guillou PJ, Markham AF, Coletta PL, Hull MA. Localization of cyclooxygenase-2 in human sporadic colorectal adenomas. Am J Pathol 156: 545–553, 2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Chen H, Tliba O, Van Besien CR, Panettieri RA, Jr, Amrani Y. TNF-α modulates murine tracheal rings responsiveness to G-protein-coupled receptor agonists and KCl. J Appl Physiol 95: 864–872; discussion 863, 2003 [DOI] [PubMed] [Google Scholar]
- 7. Chen J, Deng F, Singh SV, Wang QJ. Protein kinase D3 (PKD3) contributes to prostate cancer cell growth and survival through a PKC epsilon/PKD3 pathway downstream of Akt and ERK 1/2. Cancer Res 68: 3844–3853, 2008 [DOI] [PubMed] [Google Scholar]
- 8. Chiu T, Rozengurt E. PKD in intestinal epithelial cells: rapid activation by phorbol esters, LPA, and angiotensin through PKC. Am J Physiol Cell Physiol 280: C929–C942, 2001 [DOI] [PubMed] [Google Scholar]
- 9. Chiu TT, Leung WY, Moyer MP, Strieter RM, Rozengurt E. Protein kinase D2 mediates lysophosphatidic acid-induced interleukin 8 production in nontransformed human colonic epithelial cells through NF-κB. Am J Physiol Cell Physiol 292: C767–C777, 2007 [DOI] [PubMed] [Google Scholar]
- 10. Choi JW, Herr DR, Noguchi K, Yung YC, Lee CW, Mutoh T, Lin ME, Teo ST, Park KE, Mosley AN, Chun J. LPA receptors: subtypes and biological actions. Annu Rev Pharmacol Toxicol 50: 157–186, 2010 [DOI] [PubMed] [Google Scholar]
- 12. Edelblum KL, Goettel JA, Koyama T, McElroy SJ, Yan F, Polk DB. TNFR1 promotes tumor necrosis factor-mediated mouse colon epithelial cell survival through RAF activation of NF-kappa B. J Biol Chem 283: 29485–29494, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Giganti A, Rodriguez M, Fould B, Moulharat N, Coge F, Chomarat P, Galizzi JP, Valet P, Saulnier-Blache JS, Boutin JA, Ferry G. Murine and human autotaxin alpha, beta, and gamma isoforms: gene organization, tissue distribution, and biochemical characterization. J Biol Chem 283: 7776–7789, 2008 [DOI] [PubMed] [Google Scholar]
- 14. Guha S, Rey O, Rozengurt E. Neurotensin induces protein kinase C-dependent protein kinase D activation and DNA synthesis in human pancreatic carcinoma cell line PANC-1. Cancer Res 62: 1632–1640, 2002 [PubMed] [Google Scholar]
- 15. Huang CD, Ammit AJ, Tliba O, Kuo HP, Penn RB, Panettieri RA, Jr, Amrani Y. G-protein-coupled receptor agonists differentially regulate basal or tumor necrosis factor-alpha-stimulated activation of interleukin-6 and RANTES in human airway smooth muscle cells. J Biomed Sci 12: 763–776, 2005 [DOI] [PubMed] [Google Scholar]
- 16. Inatomi O, Andoh A, Yagi Y, Ogawa A, Hata K, Shiomi H, Tani T, Takayanagi A, Shimizu N, Fujiyama Y. Matrix metalloproteinase-3 secretion from human pancreatic periacinar myofibroblasts in response to inflammatory mediators. Pancreas 34: 126–132, 2007 [DOI] [PubMed] [Google Scholar]
- 17. Itzkowitz SH, Yio X. Inflammation and cancer. IV. Colorectal cancer in inflammatory bowel disease: the role of inflammation. Am J Physiol Gastrointest Liver Physiol 287: G7–G17, 2004 [DOI] [PubMed] [Google Scholar]
- 18. Jacamo R, Sinnett-Smith J, Rey O, Waldron RT, Rozengurt E. Sequential protein kinase C (PKC)-dependent and PKC-independent protein kinase D catalytic activation via Gq-coupled receptors: differential regulation of activation loop Ser(744) and Ser(748) phosphorylation. J Biol Chem 283: 12877–12887, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Khoa ND, Postow M, Danielsson J, Cronstein BN. Tumor necrosis factor-alpha prevents desensitization of Galphas-coupled receptors by regulating GRK2 association with the plasma membrane. Mol Pharmacol 69: 1311–1319, 2006 [DOI] [PubMed] [Google Scholar]
- 20. Kisfalvi K, Rey O, Young SH, Sinnett-Smith J, Rozengurt E. Insulin potentiates Ca2+ signaling and phosphatidylinositol 4,5-bisphosphate hydrolysis induced by Gq protein-coupled receptor agonists through an mTOR-dependent pathway. Endocrinology 148: 3246–3257, 2007 [DOI] [PubMed] [Google Scholar]
- 21. Konstantinopoulos PA, Vandoros GP, Karamouzis MV, Gkermpesi M, Sotiropoulou-Bonikou G, Papavassiliou AG. EGF-R is expressed and AP-1 and NF-kappa B are activated in stromal myofibroblasts surrounding colon adenocarcinomas paralleling expression of COX-2 and VEGF. Cell Oncol 29: 477–482, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Kruidenier L, MacDonald TT, Collins JE, Pender SL, Sanderson IR. Myofibroblast matrix metalloproteinases activate the neutrophil chemoattractant CXCL7 from intestinal epithelial cells. Gastroenterology 130: 127–136, 2006 [DOI] [PubMed] [Google Scholar]
- 23. Lin S, Wang D, Iyer S, Ghaleb AM, Shim H, Yang VW, Chun J, Yun CC. The absence of LPA2 attenuates tumor formation in an experimental model of colitis-associated cancer. Gastroenterology 136: 1711–1720, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Mahida YR, Beltinger J, Makh S, Goke M, Gray T, Podolsky DK, Hawkey CJ. Adult human colonic subepithelial myofibroblasts express extracellular matrix proteins and cyclooxygenase-1 and -2. Am J Physiol Gastrointest Liver Physiol 273: G1341–G1348, 1997 [DOI] [PubMed] [Google Scholar]
- 25. Marnett LJ, DuBois RN. COX-2: a target for colon cancer prevention. Annu Rev Pharmacol Toxicol 42: 55–80, 2002 [DOI] [PubMed] [Google Scholar]
- 26. McElroy SJ, Frey MR, Yan F, Edelblum KL, Goettel JA, John S, Polk DB. Tumor necrosis factor inhibits ligand-stimulated EGF receptor activation through a TNF receptor 1-dependent mechanism. Am J Physiol Gastrointest Liver Physiol 295: G285–G293, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Mills GB, Moolenaar WH. The emerging role of lysophosphatidic acid in cancer. Nat Rev Cancer 3: 582–591, 2003 [DOI] [PubMed] [Google Scholar]
- 28. Moolenaar WH, van Meeteren LA, Giepmans BN. The ins and outs of lysophosphatidic acid signaling. Bioessays 26: 870–881, 2004 [DOI] [PubMed] [Google Scholar]
- 29. Mottet D, Bellahcene A, Pirotte S, Waltregny D, Deroanne C, Lamour V, Lidereau R, Castronovo V. Histone deacetylase 7 silencing alters endothelial cell migration, a key step in angiogenesis. Circ Res 101: 1237–1246, 2007 [DOI] [PubMed] [Google Scholar]
- 30. Nochi H, Tomura H, Tobo M, Tanaka N, Sato K, Shinozaki T, Kobayashi T, Takagishi K, Ohta H, Okajima F, Tamoto K. Stimulatory role of lysophosphatidic acid in cyclooxygenase-2 induction by synovial fluid of patients with rheumatoid arthritis in fibroblast-like synovial cells. J Immunol 181: 5111–5119, 2008 [DOI] [PubMed] [Google Scholar]
- 31. Noguchi K, Herr D, Mutoh T, Chun J. Lysophosphatidic acid (LPA) and its receptors. Curr Opin Pharmacol 9: 15–23, 2009 [DOI] [PubMed] [Google Scholar]
- 32. Ohama T, Okada M, Murata T, Brautigan DL, Hori M, Ozaki H. Sphingosine-1-phosphate enhances IL-1β-induced COX-2 expression in mouse intestinal subepithelial myofibroblasts. Am J Physiol Gastrointest Liver Physiol 295: G766–G775, 2008 [DOI] [PubMed] [Google Scholar]
- 33. Pacheco II, MacLeod RJ. CaSR stimulates secretion of Wnt5a from colonic myofibroblasts to stimulate CDX2 and sucrase-isomaltase using Ror2 on intestinal epithelia. Am J Physiol Gastrointest Liver Physiol 295: G748–G759, 2008 [DOI] [PubMed] [Google Scholar]
- 34. Paolucci L, Rozengurt E. Protein kinase D in small cell lung cancer cells: rapid activation through protein kinase C. Cancer Res 59: 572–577, 1999 [PubMed] [Google Scholar]
- 35. Popivanova BK, Kitamura K, Wu Y, Kondo T, Kagaya T, Kaneko S, Oshima M, Fujii C, Mukaida N. Blocking TNF-alpha in mice reduces colorectal carcinogenesis associated with chronic colitis. J Clin Invest 118: 560–570, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Powell DW, Adegboyega PA, Di Mari JF, Mifflin RC. Epithelial cells and their neighbors. I. Role of intestinal myofibroblasts in development, repair, and cancer. Am J Physiol Gastrointest Liver Physiol 289: G2–G7, 2005 [DOI] [PubMed] [Google Scholar]
- 38. Rozengurt E, Rey O, Waldron RT. Protein kinase D signaling. J Biol Chem 280: 13205–13208, 2005 [DOI] [PubMed] [Google Scholar]
- 39. Saada JI, Pinchuk IV, Barrera CA, Adegboyega PA, Suarez G, Mifflin RC, Di Mari JF, Reyes VE, Powell DW. Subepithelial myofibroblasts are novel nonprofessional APCs in the human colonic mucosa. J Immunol 177: 5968–5979, 2006 [DOI] [PubMed] [Google Scholar]
- 40. Scholz RP, Regner J, Theil A, Erlmann P, Holeiter G, Jahne R, Schmid S, Hausser A, Olayioye MA. DLC1 interacts with 14–3-3 proteins to inhibit RhoGAP activity and block nucleocytoplasmic shuttling. J Cell Sci 122: 92–102, 2009 [DOI] [PubMed] [Google Scholar]
- 41. Segers VF, Van Riet I, Andries LJ, Lemmens K, Demolder MJ, De Becker AJ, Kockx MM, De Keulenaer GW. Mesenchymal stem cell adhesion to cardiac microvascular endothelium: activators and mechanisms. Am J Physiol Heart Circ Physiol 290: H1370–H1377, 2006 [DOI] [PubMed] [Google Scholar]
- 42. Shao J, Sheng GG, Mifflin RC, Powell DW, Sheng H. Roles of myofibroblasts in prostaglandin E2-stimulated intestinal epithelial proliferation and angiogenesis. Cancer Res 66: 846–855, 2006 [DOI] [PubMed] [Google Scholar]
- 43. Shattuck-Brandt RL, Varilek GW, Radhika A, Yang F, Washington MK, DuBois RN. Cyclooxygenase 2 expression is increased in the stroma of colon carcinomas from IL-10(−/−) mice. Gastroenterology 118: 337–345, 2000 [DOI] [PubMed] [Google Scholar]
- 44. Shida D, Watanabe T, Aoki J, Hama K, Kitayama J, Sonoda H, Kishi Y, Yamaguchi H, Sasaki S, Sako A, Konishi T, Arai H, Nagawa H. Aberrant expression of lysophosphatidic acid (LPA) receptors in human colorectal cancer. Lab Invest 84: 1352–1362, 2004 [DOI] [PubMed] [Google Scholar]
- 45. Sinnett-Smith J, Jacamo R, Kui R, Wang YM, Young SH, Rey O, Waldron RT, Rozengurt E. Protein kinase D mediates mitogenic signaling by Gq-coupled receptors through protein kinase C-independent regulation of activation loop Ser744 and Ser748 phosphorylation. J Biol Chem 284: 13434–13445, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Sinnett-Smith J, Zhukova E, Hsieh N, Jiang X, Rozengurt E. Protein kinase D potentiates DNA synthesis induced by Gq-coupled receptors by increasing the duration of ERK signaling in Swiss 3T3 cells. J Biol Chem 279: 16883–16893, 2004 [DOI] [PubMed] [Google Scholar]
- 47. Storz P, Doppler H, Toker A. Protein kinase C delta selectively regulates protein kinase D-dependent activation of NF-kappa B in oxidative stress signaling. Mol Cell Biol 24: 2614–2626, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Storz P, Toker A. Protein kinase D mediates a stress-induced NF-kappa B activation and survival pathway. EMBO J 22: 109–120, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Tanaka T, Kohno H, Suzuki R, Yamada Y, Sugie S, Mori H. A novel inflammation-related mouse colon carcinogenesis model induced by azoxymethane and dextran sodium sulfate. Cancer Sci 94: 965–973, 2003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Terzic J, Grivennikov S, Karin E, Karin M. Inflammation and colon cancer. Gastroenterology 138: 2101–2114, 2010 [DOI] [PubMed] [Google Scholar]
- 51. Valentich JD, Popov V, Saada JI, Powell DW. Phenotypic characterization of an intestinal subepithelial myofibroblast cell line. Am J Physiol Cell Physiol 272: C1513–C1524, 1997 [DOI] [PubMed] [Google Scholar]
- 52. Vandoros GP, Konstantinopoulos PA, Sotiropoulou-Bonikou G, Kominea A, Papachristou GI, Karamouzis MV, Gkermpesi M, Varakis I, Papavassiliou AG. PPAR-gamma is expressed and NF-kB pathway is activated and correlates positively with COX-2 expression in stromal myofibroblasts surrounding colon adenocarcinomas. J Cancer Res Clin Oncol 132: 76–84, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Wajant H, Pfizenmaier K, Scheurich P. Tumor necrosis factor signaling. Cell Death Differ 10: 45–65, 2003 [DOI] [PubMed] [Google Scholar]
- 54. Waldron RT, Rey O, Iglesias T, Tugal T, Cantrell D, Rozengurt E. Activation loop Ser744 and Ser748 in protein kinase D are transphosphorylated in vivo. J Biol Chem 276: 32606–32615, 2001 [DOI] [PubMed] [Google Scholar]
- 55. Waldron RT, Rey O, Zhukova E, Rozengurt E. Oxidative stress induces protein kinase C-mediated activation loop phosphorylation and nuclear redistribution of protein kinase D. J Biol Chem 279: 27482–27493, 2004 [DOI] [PubMed] [Google Scholar]
- 56. Waldron RT, Rozengurt E. Oxidative stress induces protein kinase D activation in intact cells. Involvement of Src and dependence on protein kinase C. J Biol Chem 275: 17114–17121, 2000 [DOI] [PubMed] [Google Scholar]
- 57. Waldron RT, Rozengurt E. Protein kinase C phosphorylates protein kinase D activation loop Ser744 and Ser748 and releases autoinhibition by the pleckstrin homology domain. J Biol Chem 278: 154–163, 2003 [DOI] [PubMed] [Google Scholar]
- 57a. Wang D, Dubois RN. Pro-inflammatory prostaglandins and progression of colorectal cancer. Cancer Lett 267: 197–203, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Wendum D, Comperat E, Boelle PY, Parc R, Masliah J, Trugnan G, Flejou JF. Cytoplasmic phospholipase A2 alpha overexpression in stromal cells is correlated with angiogenesis in human colorectal cancer. Mod Pathol 18: 212–220, 2005 [DOI] [PubMed] [Google Scholar]
- 59. Woclawek-Potocka I, Kondraciuk K, Skarzynski DJ. Lysophosphatidic acid stimulates prostaglandin E2 production in cultured stromal endometrial cells through LPA1 receptor. Exp Biol Med (Maywood) 234: 986–993, 2009 [DOI] [PubMed] [Google Scholar]
- 60. Wu JM, Xu Y, Skill NJ, Sheng H, Zhao Z, Yu M, Saxena R, Maluccio MA. Autotaxin expression and its connection with the TNF-alpha-NF-kappaB axis in human hepatocellular carcinoma. Mol Cancer 9: 71, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Yamaoka T, Yan F, Cao H, Hobbs SS, Dise RS, Tong W, Polk DB. Transactivation of EGF receptor and ErbB2 protects intestinal epithelial cells from TNF-induced apoptosis. Proc Natl Acad Sci U S A 105: 11772–11777, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Yoo J, Chung C, Slice L, Sinnett-Smith J, Rozengurt E. Protein kinase D mediates synergistic expression of COX-2 induced by TNF-α and bradykinin in human colonic myofibroblasts. Am J Physiol Cell Physiol 297: C1576–C1587, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Yoo J, Nichols A, Song JC, Mammen J, Calvo I, Worrell RT, Cuppoletti J, Matlin K, Matthews JB. Bryostatin-1 attenuates TNF-induced epithelial barrier dysfunction: role of novel PKC isozymes. Am J Physiol Gastrointest Liver Physiol 284: G703–G712, 2003 [DOI] [PubMed] [Google Scholar]
- 64. Yuan J, Slice LW, Gu J, Rozengurt E. Cooperation of Gq, Gi, and G12/13 in protein kinase D activation and phosphorylation induced by lysophosphatidic acid. J Biol Chem 278: 4882–4891, 2003 [DOI] [PubMed] [Google Scholar]
- 65. Zhang H, Bialkowska A, Rusovici R, Chanchevalap S, Shim H, Katz JP, Yang VW, Yun CC. Lysophosphatidic acid facilitates proliferation of colon cancer cells via induction of Kruppel-like factor 5. J Biol Chem 282: 15541–15549, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Zhukova E, Sinnett-Smith J, Rozengurt E. Protein kinase D potentiates DNA synthesis and cell proliferation induced by bombesin, vasopressin, or phorbol esters in Swiss 3T3 cells. J Biol Chem 276: 40298–40305, 2001 [DOI] [PubMed] [Google Scholar]
- 67. Zugaza JL, Sinnett-Smith J, Van Lint J, Rozengurt E. Protein kinase D (PKD) activation in intact cells through a protein kinase C-dependent signal transduction pathway. EMBO J 15: 6220–6230, 1996 [PMC free article] [PubMed] [Google Scholar]
- 68. Zugaza JL, Waldron RT, Sinnett-Smith J, Rozengurt E. Bombesin, vasopressin, endothelin, bradykinin, and platelet-derived growth factor rapidly activate protein kinase D through a protein kinase C-dependent signal transduction pathway. J Biol Chem 272: 23952–23960, 1997 [DOI] [PubMed] [Google Scholar]






