Skip to main content
Cell Proliferation logoLink to Cell Proliferation
. 2004 Nov 17;37(6):427–441. doi: 10.1111/j.1365-2184.2004.00324.x

Extracellular pressure stimulates colon cancer cell proliferation via a mechanism requiring PKC and tyrosine kinase signals

M F Walsh 1, R K‐Y Woo 1, R Gomez 1, M D Basson 1,
PMCID: PMC6495684  PMID: 15548175

Abstract

Abstract.  Pressure in colonic tumours may increase during constipation, obstruction or peri‐operatively. Pressure enhances colonocyte adhesion by a c‐Src‐ and actin‐cytoskeleton‐dependent PKC‐independent pathway. We hypothesized that pressure activates mitogenic signals.

Methods.  Malignant colonocytes on a collagen I matrix were subjected to 15 mmHg pressure. ERK, p38, c‐Src and Akt phosphorylation and PKCα redistribution were assessed by western blot after 30 min and PKC activation by ELISA. Cells were counted after 24 h and after inhibition of each signal, tyrosine phosphorylation or actin depolymerization.

Results.  Pressure time‐dependently increased SW620 and HCT‐116 cell counts on collagen or fibronectin (P < 0.01). Pressure increased the SW620 S‐phase fraction from 28 ± 1 to 47 ± 1% (P = 0.0002). Pressure activated p38, ERK, and c‐Src (P < 0.05 each) but not Akt/PKB. Pressure decreased cytosolic PKC activity, and translocated PKCα to a membrane fraction. Blockade of p38, ERK, c‐Src or PI‐3‐K or actin depolymerization did not inhibit pressure‐stimulated proliferation. However, global tyrosine kinase blockade (genistein) and PKC blockade (calphostin C) negated pressure‐induced proliferation.

Conclusions.  Extracellular pressure stimulates cell proliferation and activates several signals. However, the mitogenic effect of pressure requires only tyrosine kinase and PKCα activation. Pressure may modulate colon cancer growth and implantation by two distinct pathways, one stimulating proliferation and the other promoting adhesion.

INTRODUCTION

Physical forces, either within tissues, or applied externally, impact all tissues of the body and have been studied in some detail (Duncan & Turner 1995; Osol 1995; Chien et al. 1998; Sai et al. 1999). Mechanotransduction converts biophysical forces into cellular responses that influence gene expression, protein synthesis, proliferation and morphogenesis. The mechanisms coupling mechanical signals with biochemical events remain poorly understood. Responses to stretch, shear and deformation have been investigated in bone, vascular cells, cardiac fibroblasts and myocytes and mesangial cells, but less is known about the effects of pressure, and the distinction is important because different physical forces may have different effects (Han et al. 1998b; MacKenna et al. 1998; Ingram et al. 1999; Han et al. 2001; Moalli et al. 2001; Basson & Coppola 2002; Hosokawa et al. 2002; Zhang et al. 2003).

The normal gut experiences complex patterns of contractility and motility and changes in pressure during normal peristalsis. These internally generated pressures may help maintain normal gut cytoarchitecture. However, pressures elevated by diet or illness may adversely impact gut physiology (Brodribb et al. 1979). Luminal jejunal pressure may reach 50 mmHg in irritable bowel syndrome (Kellow & Phillips 1987). Abdominal pressures of 15 mmHg are common during insufflation for laparoscopy and surgical manipulation yields pressures exceeding 1500 mmHg (Dregelid & Svendsen 1988). Intra‐abdominal pressure also increases after surgery as a result of tissue oedema (Granger & Barrowman 1983). Such pressures may affect cancer dissemination (Basson et al. 2000; Thamilselvan & Basson 2004) and the function of intra‐abdominal organs (Walker & Criddle 2003).

Early concerns about cancer recurrence at surgical wounds after laparoscopic surgery (Pahlman 1997) prompted studies of the role of pressure in cancer cell adhesion and proliferation (Jacobi et al. 1998; Hewett et al. 1999; Jacobi et al. 2002; Dahn et al. 2003). Animal studies suggest that pressure increases may contribute to tumour growth after laparoscopic procedures (Bouvy et al. 1998). In mice, pneumoperitoneum enhanced growth of GW39 human colon cancer cells (Wu et al. 1997). Pressure (15 mmHg) applied to colon 26 cells increased liver metastasis regardless of insufflation gas (Gutt et al. 2000; Ishida et al. 2001), suggesting that even this modest pressure increase might influence malignant cell proliferation. The incidence of wound metastasis after laparoscopic and open surgery today is similar (Silecchia et al. 2002; Patankar et al. 2003), perhaps offset by immune depression after open surgery (Kawasaki et al. 2001), or application of substantial pressure to tumours during open surgical dissection (Dregelid & Svendsen 1988).

We have shown that a 30‐min exposure of non‐adherent primary human colon cancer and SW620 cells to 15 mmHg of extracellular pressure increases cell adhesion to both collagen I and endothelial cells via a FAK/c‐Src dependent mechanism that is also blocked by phalloidin treatment (Basson et al. 1992; Thamilselvan & Basson 2004). In the present study, we sought to determine whether longer exposure to increases of this magnitude affects proliferation in adherent cells. Previous in vitro studies of pressure‐mediated proliferation in other cells have considered much higher pressures. In mesangial cells, hydrostatic pressure induces proliferation in an intensity‐dependent fashion (Mattana & Singhal 1995; Kawata et al. 1998a). In the same cells, 40–80 mmHg pressures stimulated proliferation in a time‐ and pressure‐dependent fashion (Kato et al. 1999). Pressures of 105 mmHg increase proliferation in vascular smooth muscle (VSMC) and endothelial cells (Sumpio et al. 1994; Watase et al. 1997). In VSMC, pressure‐induced proliferation is inhibited by PKC, p38 and ERK inhibition as well as by the tyrosine kinase inhibitor, genistein (Tsuda et al. 2002). In fibroblasts, a mechanical load elicits both MAPK and NF‐κB activation (Chiquet et al. 2003). Li and Xu postulated that stretch of the VSMC membrane alters tyrosine receptor or G‐protein conformation, initiating signalling pathways normally used by growth factors (Hu et al. 1998; Li & Xu 2000) that stimulate PKC, MAPK, c‐fos and other gene expression (Li et al. 1999). Similarly, in rodent mesenteric arteries, increased pressure enhances c‐fos expression, an effect inhibited by genistein (Miriel et al. 1999), and requiring both c‐Src and ERK (Wesselman et al. 2001; Rice et al. 2002). Other investigators suggest that integrin aggregation per se may activate tyrosine kinase receptors in the absence of ligand (Miyamoto et al. 1996; Esposito et al. 2003), another possible mechanism for the transduction of external physical forces such as pressure.

We hypothesized that longer exposures to a physiologically relevant increase of 15 mmHg in pressure might stimulate colon cancer cell proliferation. We delineated the time course of this effect in human SW620 colon cancer cells and examined whether it could be duplicated in human HCT‐116 colon cancer cells. We further investigated whether pressure activated several intracellular signals known to be involved in proliferation in response to other forces or growth factors, particularly c‐Src, PI‐3‐K, MAPK, and PKC. We then assessed proliferation in the presence of specific signal inhibition, or the global tyrosine kinase inhibitor genistein or phalloidin, to block actin re‐organization.

MATERIALS AND METHODS

Cell culture

SW620 cells were cultured at 37 °C in 5% CO2 in an equal mixture of Dulbecco's minimal essential medium (DMEM) and RPMI 1540 containing 5% foetal bovine serum (FBS, Gibco Life Technologies Inc., Rockville, MD, USA), 20 mm glutamine, 1 µm sodium pyruvate, 10 mm Hepes, 100 U/ml penicillin G, 100 µg/ml streptomycin (Sigma, St Louis, MO, USA) and 0.525 µg/ml transferrin (Roche Diagnostics, Indianapolis, IN, USA). The HCT‐116 cells were maintained in DMEM supplemented with 10% FBS. Both human colon cancer cell lines were obtained from ATCC (American Tissue Culture, Collection, Rockville, MD, USA).

Matrix pre‐coating

Six‐well plates were pre‐coated with collagen I or plasma fibronectin (12.5 µg/ml, Sigma) as previously described (Basson et al. 1992) and the wells were rinsed with sterile phosphate‐buffered saline prior to cell seeding.

Pressure application

Pressure was applied by placing the six‐well plates and cells within an airtight box equipped with inlet and outlet valves and a pressure gauge as previously described (Basson et al. 2000; Vouyouka et al. 2003; Thamilselvan & Basson 2004). The box was pre‐warmed to 37 °C; the temperature did not fluctuate more than ±2 °C for the duration of the experiment. The internal pressure was set at 15 mmHg and maintained within ±1.5 mmHg.

Proliferation assays

Uniform cell aliquots were dispensed into the pre‐coated wells and allowed to reach 70% confluence before being subjected to increased pressure (15 mmHg) for 24 h. At that time, the cells were detached with trypsin/EDTA, the trypsin was neutralized with medium containing 5% FBS and cell aliquots were counted in a Coulter Counter (Coulter Electronics, Hialeah, FL, USA). Control cells maintained in the same incubator but outside of the pressure box were treated similarly. When used, the following inhibitors, PD98059 (20 µm), SB203580 (20 µm), PP2 (20 µm), LY294002 (10 µm), genistein (10 µm), phalloidin (10 µm), or calphostin C (100 nm, light‐activated for 1 h) (all Calbiochem, La Jolla, CA, USA), were added to the cells 30 min before the application of pressure. Appropriate volumes of the dimethylsulfoxide (DMSO) or ethanol vehicle were added to the corresponding controls at the same time. Assays were normally performed on cells in DMEM/RPMI supplemented with 5% FBS. Basic proliferation assays in response to pressure were duplicated under serum‐free conditions to corroborate results obtained with flow cytometry.

Flow cytometry

To document proliferation, cells exposed to pressure for 24 h were detached with trypsin/EDTA and fixed in 70% ice‐cold ethanol. The cells were then stained with propidium iodide (5 µg/ml PBS, pH 7.4 containing 200 µg/ml DNase‐free Rnase A and 0.1% Triton X‐100). The cells were then analysed for transition to S‐phase by the Wayne State University School of Medicine Flow Cytometry Core Facility. Assays were performed on cells maintained in 5% FBS and repeated in cells rendered quiescent by serum starvation for 24 h prior to pressure exposure.

Signal activation assays

For signal activation studies, cells were exposed to 15 mmHg pressure for 30 min. The cells were then lysed on ice for 30 min in buffer containing 150 mm NaCl, 10 mm Tris, 1% TritonX‐100, 1% sodium deoxycholate, 0.1% sodium dodecyl sulfate (SDS), 1 mm EDTA, 1 mm phenylmethylsulphonyl fluoride, 1 mm Na3VO4, 50 mm NaF, 10 mm sodium pyrophosphate, 2 µg/ml aprotinin, 2 µg/ml leupeptin, pH 7.4. After centrifugation, supernatant protein concentrations were measured by the BCA protein method (Pierce, Rockford, IL, USA). Proteins were separated by SDS–polyacrylamide gel electrophoresis (PAGE) followed by transfer to Hybond ECL nitrocellulose and incubation with antibodies specific for the active (phosphorylated) forms of ERK (p44/42), p38, c‐Src, and Akt/PKB (Cell Signalling, Beverly, MA, USA; Santa Cruz Biotechnology, Santa Cruz, CA, USA; Transduction Laboratories, San Diego, CA, USA). Membranes were then stripped and re‐probed with antibodies to the total form of each molecule. After exposure to the appropriate second antibody coupled to horseradish peroxidase, bands were detected with enhanced chemiluminescence (ECL; Amersham Pharmacia Biotech, Piscataway, NJ, USA) and analysed with a Kodak Image Station 440CF (Perkin Elmer, Boston, MA, USA).

PKC activity and translocation

PKC activity was assayed in cytosolic fractions utilizing a synthetic PKC pseudosubstrate following the manufacturer's directions (Calbiochem). Briefly, control and test cells exposed to 15 mmHg for 30 min were lysed, sonicated on ice and centrifuged at 100 000 g for 60 min. Supernatant protein concentrations were equalized prior to assay for PKC activity. Cells treated with PMA (1 µm) served as a positive control. Calcium‐ and lipid‐sensitive PKC activity was quantified using a colourimetric ELISA based upon a monoclonal antibody to the phosphorylated form of the pseudosubstrate. For the translocation studies, soluble and particulate cell fractions were prepared according to the methods of Bissonnette (Bissonnette et al. 1994). After solubilization in extraction buffer (20 mm HEPES, pH 7.6, supplemented with 5 mm EGTA, 5 mm Na pyrophosphate, 1 mm MgCl2, 1 mm PMSF and 10 µm leupeptin), the samples were centrifuged at 100 000 g for 60 min and the soluble fraction was removed. The particulate fraction was re‐suspended in 20 mm HEPES, pH 7.6 containing 150 mm NaCl, 1 mm MgCl2, and 1% Triton X‐100. Equal protein aliquots of soluble and particulate fractions were separated via SDS–PAGE and transferred to nitrocellulose. Blots were probed with both a pan‐PKC antibody that recognizes all PKC isoforms and an antibody specific for PKCα (Calbiochem and Chemicon, Temecula, CA, USA). Protein loading was controlled for with GAPDH (Biodesign International, Saco, ME, USA, soluble fraction) and Na/K ATPase (Sigma, particulate fraction).

Statistical analysis

Results are expressed as mean ± SEM and differences between groups were evaluated using both unpaired and paired Student's t‐tests as warranted with statistical significance being set a priori at P < 0.05.

RESULTS

15 mmHg pressure significantly increases cell proliferation

SW620 cell proliferation on collagen I is increased after 6 h of pressure. Using the pressure apparatus described above, we first determined the effects of increasing pressure by 15 mmHg over ambient pressure for 24 h on SW620 cells. As illustrated in Fig. 1(a), exposure to pressure increased cell number by 51 ± 7% (n = 14, P = 0.0001). Data shown represent absolute cell counts normalized to time = 0 (cell counts at the onset of the study) and are expressed as mean ± SEM. A time course study was performed to further delineate the pressure effect on proliferation. Cells were exposed to pressure for 0, 3, 4.5, 6, 12 and 24 h and then maintained under ambient pressure for the balance of a 24‐h period prior to trypsinization and cell counting. As shown in Figs 1(b), 3 h of pressure did not result in a subsequent significant increase in cell number (3.4 ± 3.0%). However, after 6 h of pressure, cell numbers at 24 h were significantly enhanced compared with cells exposed to ambient pressure. Furthermore, the magnitude of this increase was similar to that observed after either 12 or 24 h exposure to pressure. Figure 1(b) represents absolute cell counts two‐point normalized between t = 0 and t = 24 to adjust for differences in proliferation between studies. Thus, pressure‐mediated increases in cell number appeared duration‐dependent, requiring 6 h of exposure to reach maximal stimulation. Apoptosis, both early and late, accounted for only 1% of cells in either condition (data not shown). To document that the result of exposure of SW620 cells to pressure of 15 mmHg over ambient pressure cells for 24 h was proliferation, cells were assessed for S‐phase transition via flow cytometry following propidium iodide staining. When the cells were maintained in complete medium with 5% FBS, conditions for the original proliferation experiments, the percentage of cells in S‐phase increased from 39.5 ± 1.2 in control cells to 42.9 ± 2.2 in cells under pressure (P = 0.03, n = 6). When cells were rendered quiescent by serum‐starvation for 24 h prior to pressure, the difference was magnified. As shown in Fig. 1(c), 27.8 ± 1.0% of serum‐starved cells at ambient pressure were found to be in S‐phase compared with 46.6 ± 0.8% of serum‐starved cells subjected to pressure (P = 0.0002, n = 6). Cell count studies repeated in cells rendered quiescent showed proliferation of a similar magnitude to that observed in non‐quiescent cells (data not shown).

Figure 1.

Figure 1

Pressure stimulates time‐dependent SW620 cell proliferation. (a) 15 mmHg pressure application for 24 h results in a 51 ± 7% increase in cell number compared with the ambient pressure control at 24 h (n = 14, P = 0.0001). (b) Time course of the pressure effect. Cells were exposed to pressure for 0, 3, 4.5, 6, 12 or 24 h and then maintained at ambient pressure for the balance of the 24‐h period. Cell numbers are significantly (P < 0.05) increased over control values after 6 h of exposure (data shown are two‐point normalized between t = 0 and 24 h of exposure to pressure) and remain elevated for 24 h. (c) Flow cytometry in cells rendered quiescent by serum deprivation. The ratio of cells in S‐phase after 24 h of pressure was 46.6 ± 0.8% compared with 27.8 ± 0.1% in cells maintained at ambient pressure (P = 0.0002, n = 6). All data, mean ± SEM; t = 0, initial cell count; control, ambient pressure; pressure, 15 mmHg.

Pressure enhances proliferation on a different matrix and in a different cell line

We previously have reported that another physical force, cyclic strain, is mitogenic to cells on collagen but not to those fibronectin and that fibronectin inhibits strain‐stimulated signals (Zhang et al. 2003). To determine whether the pressure effect on proliferation was similarly inhibited by fibronectin, the experiments were repeated in SW620 cells plated on to plasma fibronectin. As seen in Fig. 2(a), a 24‐h exposure to 15 mmHg increased cell number by 29 ± 6% (P = 0.0013, n = 6). Results are normalized to cell counts at time = 0. Similar results were obtained in HCT‐116 colon cancer cells plated on to collagen I, in which cell number was increased by 27 ± 8% by pressure (P = 0.009, n = 6) (Fig. 2b). These results suggest that pressure‐mediated colon cancer cell proliferation is neither limited to a single cell line nor inhibited by fibronectin.

Figure 2.

Figure 2

Pressure‐stimulated colon cancer cell proliferation is independent of matrix and cell line. (a) In SW620 cells on a fibronectin matrix, 24 h of exposure to 15 mmHg increased pressure increased cell number by 29 ± 6% over ambient pressure controls at 24 h (P = 0.0013, n = 6). (b) Similarly, in HCT116 cells plated on collagen I, pressure enhanced cell number by 27 ± 8% (P = 0.009, n = 6). All data, mean ± SEM normalized to cell counts at t = 0.

15 mmHg pressure activates intracellular signals after 30 min

ERK, p38 and c‐Src are activated, but not Akt. We next examined several intracellular signals that have been implicated in either cellular proliferation or physical force effects in colonocytes or other cell systems. To determine whether activation of these signals preceded the observed proliferation, SW620 cells plated on collagen I were exposed to 15 mmHg pressure for 30 min. Cell lysates of control and experimental cells were analysed by western blot for their activated, phosphorylated forms. Results in the bar graphs in Fig. 3a were calculated as the ratio of phosphorylated to total band intensity for each signal and expressed as per cent of ambient control. Statistics were calculated on the initial raw data ratios before normalization to control values. Representative western immunoblots for the phosphorylated and total molecule are shown above the bars. After 30 min of pressure, ERK (p44/42) activity was higher than that of control by 33 ± 10% (P = 0.0035, n = 7), p38 activation was increased by 51 ± 18% (P = 0.0005, n = 5), and c‐Src phosphorylation was enhanced by 23 ± 7% (P = 0.05, n = 3). Akt, however, was not significantly activated by pressure (15 ± 5%, n = 6).

Figure 3.

Figure 3

Pressure induces early (30 min) signal activation. (a) Western blot assays of ERK, p38, c‐Src and Akt/PKB phosphorylation presented as the ratio of phosphorylated to total forms. Bars depict the results of densitometric analysis (mean ± SEM, n = 3–7, P < 0.05). Representative western blots are shown above the bars. ERK, p38, and c‐Src but not Akt/PKB, were activated by 30 min of pressure. (b) Cytosolic PKC activity, expressed as percentage of control, is significantly decreased by pressure (P = 0.026, n = 3). Activation by 1 µg/ml PMA was used as a positive control. DMSO‐treated cells were assayed as a vehicle control for the PMA. (c) Western blot analysis of PKCα subcellular distribution in response to pressure. Bars represent densitometric ratios of PKCα to GAPDH for the cytosolic fraction (Cyto) and to Na/K ATPase for the membrane fraction (Memb) (*P = 0.05 membrane pressure vs. membrane control, n = 3). A representative western blot is shown above the bars. Control, ambient; pressure, 15 mmHg.

PKC activity is lower in the cytosolic fraction while PKCα is increased in the membrane fraction of cells exposed to pressure

Activation of the various forms of PKC usually involves translocation of the enzyme from the cytosol to the membrane (Sylvester et al. 2001). We therefore measured calcium‐ and lipid‐dependent PKC activity in whole cell lysates and cytosolic fractions of control cells and cells subjected to 15 mmHg pressure for 30 min. Activity was compared with that obtained after exposure of the cells to 1 µg/ml PMA for 30 min. Data presented in Fig. 3(b) show per cent of control cell activity for both the pressure and PMA experiments. Pressure induced a 21 ± 5% decrease in overall PKC activity, smaller than the 70% decrease seen with PMA but nevertheless significant (P = 0.026, n = 3). To further demonstrate PKC activation, both the soluble (cytosolic) and particulate (membrane) fractions of cells exposed to pressure and their controls were assayed for PKC content. Western blotting with an anti‐PKC antibody that cross reacts with all PKC isozymes demonstrated significant pressure‐induced PKC translocation to the membrane fraction (data not shown). However, we chose to focus on PKCα because it is a calcium‐and lipid‐dependent isoform of the enzyme and previous work in our laboratory documented its activation in response to another physical force, repetitive deformation, in the Caco‐2 intestinal epithelial cell line (Han et al. 1998b). GAPDH and Na/K ATPase were used as controls for protein loading in the cytosolic and membrane fractions, respectively, because GAPDH is known to be a cytosolic enzyme and Na/K ATPase is a traditional membrane marker. Cytosolic PKCα content was not measurably changed by pressure, perhaps because it was much lower than that of the membrane fraction even in unstimulated cells (Fig. 3c) and any change would have been difficult to discern. However, membrane PKCα, calculated as the ratio of PKC to Na/K ATPase, was significantly higher in the membrane fraction of cells exposed to pressure (2.39 ± 0.24 vs. 1.58 ± 0.30, P = 0.05, n = 3), corroborating the PKC activity data and indicating that at least the α isoform of the PKC enzyme is translocated by pressure.

Pressure‐induced proliferation is inhibited by tyrosine kinase and PKC inhibition

We next sought to determine which of the signals shown to be activated by pressure might play a role in pressure‐stimulated SW620 cell proliferation. We used the following inhibitors: SB203580 (20 µm) for p38, PD98059 (20 µm) for p44/42, PP2 (20 µm) for c‐Src, LY294002 (10 µm) for PI‐3 kinase, and light‐activated calphostin C (100 nm) for PKC. We have previously demonstrated signal inhibition in SW620 by the SB, PD, LY and PP2 compounds at these doses (Walsh et al. 2003; Thamilselvan & Basson 2004; Thamilselvan et al. 2004) but inhibition of the requisite signal was further documented by western blot analysis in the current study (data not shown). As mechanical forces have been suggested to act directly on the cytoskeleton, we also used phalloidin (10 µm) to stabilize the actin cytoskeleton. In addition, we examined the effects of the less specific tyrosine kinase inhibitor, genistein (10 µm). Preliminary data indicated that the DMSO and ethanol vehicles, at the volumes used, did not affect cell proliferation. (The increase in pressure‐induced proliferation in these experiments, expressed as a percentage of respective control values was 44 ± 9% in untreated cells, 52 ± 11% in cells exposed to 0.1% DMSO, and 40 ± 10% in cells in 0.1% ethanol.) Therefore, results were not normalized and are presented as the number of cells exposed to the inhibitor both under ambient (control) pressure and 15 mmHg for 24 h. Of the inhibitors used, those that did not affect pressure‐induced cell proliferation are shown in Fig. 4(a). Pressure increased cell number by 44 ± 11% in LY‐treated cells (P = 0.003), 75 ± 17% in phalloidin‐treated cells (P = 0.0003), 55 ± 22% in PP2‐treated cells (P = 0.04), 73 ± 19% in SB‐treated cells (P = 0.015) and 32 ± 8% in PD‐treated cells (n = 4–7 for each). Thus, none of these agents blocked pressure‐stimulated SW620 cell proliferation. However, as shown in Fig. 4(b), exposure to either genistein or calphostin C prevented pressure‐induced cell proliferation. Whereas pressure enhanced cell number by 30 ± 9% in control cells (P = 0.04), the increase was limited to 11 ± 9% by genistein and 5 ± 3.5% by calphostin C (n = 5 each), not significantly different from control in either case, suggesting that pressure stimulates proliferation in SW620 cells by both PKC‐ and tyrosine‐kinase‐dependent mechanisms.

Figure 4.

Figure 4

Tyrosine kinase and PKC inhibition block pressure‐induced proliferation. (a) Pharmacological blockade of PI‐3‐kinase with 20 µm LY294002 (LY), actin depolymerization with 10 µm phalloidin (Ph), c‐Src blockade with 20 µm PP2 (PP2), p38 inhibition with 20 µm SB203580 (SB) and MEK inhibition with 20 µm PD98059 (PD) all failed to negate the pressure effect on proliferation. *P < 0.05 vs. respective control (ambient). Appropriate vehicle controls yielded similar pressure mediated increases in cell number. (b) Genistein (10 µm) and calphostin C (100 nm) abrogate pressure‐stimulated cell proliferation, limiting increases in cell number to 11 ± 9% with genistein and 5 ± 3% with calphostin C (n = 5, each).

DISCUSSION

We have previously reported that a 15 mmHg increase over ambient pressure increases colon cancer cell adhesion by a mechanism requiring FAK, c‐Src, and an intact actin‐cytoskeleton but is independent of PKC or ERK (Basson et al. 2000; Thamilselvan & Basson 2004). We now demonstrate that exposure to a 15 mmHg elevation in pressure stimulates colon cancer proliferation in a time‐dependent but matrix‐independent manner in adherent cells from two different colon cancer cell lines. Changes in proliferation (assessed by S‐phase fraction) is likely to explain this increase in cell number. Proliferation was preceded by pressure activation of p38, ERK, c‐Src and PKCα. However, only PKC blockade or global tyrosine kinase inhibition negated the pressure effect on proliferation.

Pressure enhancement of proliferation has been reported in mesangial (Mattana & Singhal 1995; 1998a, 1998b; Kato et al. 1999), vascular smooth muscle (Hishikawa et al. 1994; Watase et al. 1997; Tsuda et al. 2002) and endothelial cells (Sumpio et al. 1994). Because interest in these cells is related to hypertension, most of those studies were performed at the 40–120 mmHg pressures found in the vascular system, The same high pressures enhanced proliferation and DNA synthesis in a rat intestinal epithelial cell line (Hirokawa et al. 2001). In contrast, published studies have focused on the effects of pressures within the 15 mmHg range on colon cancer cells in vivo. A pneumoperitoneum yielding 15 mmHg pressures promoted the occurrence of liver metastasis in mice implanted with colon 26 cells (Ishida et al. 2001; Gutt et al. 2003). The mechanisms of this result, however, are unclear, and certainly may include the effects of pneumoperitoneal pressures on adhesion (Basson et al. 2000; Thamilselvan & Basson 2004), in addition to the outcome on proliferation suggested by our current observations. Pressures between 6 and 12 mmHg enhanced the proliferation of CX‐2 colon adenocarcinoma in vitro after 5 days but, unlike our results, proliferation was suppressed by pressure at earlier time points (Gutt et al. 2000). The disparity between the prior results and ours may reflect the somewhat higher pressure used in our study, an idiosyncracy of the CX‐2 cell line, or the effect of a more physiologic matrix protein substrate. The CX‐2 cells were cultured on tissue culture plastic, without a matrix substrate, perhaps eliminating the contribution of integrin engagement to the transduction of the pressure signals (Miyamoto et al. 1996; Esposito et al. 2003).

Of the signals studied, p38, ERK, c‐Src, and PKC were activated after 30 min of exposure to 15 mmHg pressure, but Akt/PKB was not. Many of these signals are activated in some, but not all, cells in response to pressure or other mitogenic stimuli such as growth factors (Hishikawa et al. 1994; Tsuda et al. 2002) (1998a, 1998b; Kato et al. 1999). Tyrosine kinase receptors activate PKC and PI‐3‐K in SW480 and Caco‐2 human colon cancer cells (Graness et al. 1998; Jasleen et al. 2000). In HT‐29 colon cancer cells, growth factors stimulate c‐Src, PI‐3‐K, Akt/PKB, ERK, and proliferation (Kim et al. 2002; Golubovskaya et al. 2003). Proliferative activity in five human colon cancer cell lines has been linked to c‐Src (Sekharam et al. 2003) but our previous results (Thamilselvan & Basson 2004) and those of others (Haier et al. 2002; Jones et al. 2002) suggest that c‐Src may be more important for pressure stimulation of cell adhesion than proliferation. In this study, c‐Src inhibition did not prevent the mitogenic effects of pressure. Similarly, although phalloidin blocks the stimulation of SW620 or primary human colon cancer cell adhesion by pressure (Thamilselvan & Basson 2004), phalloidin did not inhibit pressure‐induced proliferation, suggesting that the mitogenic effects of pressure in adherent cells may not require cytoskeletal input.

In non‐adherent SW620 cells, pressure activates p38 but not ERK (Thamilselvan & Basson 2004). Pressure stimulates both ERK and p38 in adherent cells, but inhibition of these MAPK components did not block proliferation. ERK activation has been associated with proliferation (Shapiro et al. 1996). However, ERK activity may need to exceed a specific threshold to stimulate proliferation (Kuwada & Li 2000). In addition, and consistent with our results, MAPK activation is not necessarily related to proliferation in other cells (Xu et al. 2000). Strain stimulates endothelial ERK and PKC, but only PKC inhibition prevents proliferation (Ikeda et al. 1999). Similarly, collagen expression in mesangial cells required both ERK and PKC. Interestingly, the PKC‐δ, ‐ɛ and ‐ζ effects were ERK‐dependent but those mediated via PKC‐α and ‐β were ERK‐independent (Hua et al. 2001). We found similar dissociation between ERK and PKC‐α effects.

Although pressure activated MAPK, c‐Src, and PKC, only genistein and calphostin C negated the mitogenic effect of pressure. The genistein data suggest that some receptor or intracellular tyrosine kinase is necessary. Genistein blocks pressure‐stimulated smooth muscle and mesangial cell proliferation and the mitogenic effects of GLP‐2 and strain in Caco‐2 cells (Han et al. 1998b; 1998a, 1998b; Tsuda et al. 2002). However, in VSMC both ERK and p38 are required (Tsuda et al. 2002), whereas Caco‐2 mitogenicity is sensitive to PI‐3‐K and MAPK blockade (Han et al. 1998b; Jasleen et al. 2000). In contrast, the genistein‐sensitive mitogenic pressure effect in SW620 cells was independent of c‐Src or the MAPK. Genistein may be inhibiting tyrosine kinase activation of a number of growth factor receptors, such as those of EGF or IGF‐1, intermediary or docking proteins such as Shc and Grb2, or the JAK/STAT pathway, among others. Each of these has been implicated in cell proliferation but their role(s) in the mitogenic effect of pressure awaits further study.

Here, exposure to 15 mmHg pressure for 30 min stimulated PKC‐α translocation to the membrane and PKC inhibition by calphostin C blocked the ensuing proliferation. Early mitogenic signals are commonly associated with delayed effects, because either the signals themselves or their downstream consequences persist. Proliferation and PKC activation have been closely linked in colonocytes, and we have previously reported that a rapid biphasic PKC activation over 1–5 min was associated with increased Caco‐2 cell number 24 h later in response to cyclic strain (Cadoret et al. 1998; 1998a, 1998b; Davidson et al. 2000; Cerda et al. 2001). The PKC family of serine/threonine kinases includes conventional lipid‐ and calcium‐sensitive PKCs (α, β and γ), novel lipid‐sensitive calcium‐independent PKCs (δ, ɛ, η and θ), and atypical PKCs (Mellor & Parker 1998; Toker 1998). PKC isoform abundance varies with proliferation, differentiation and apoptosis in normal colon (Cesaro et al. 2001). Human, mouse and rat colonocytes, both normal and malignant, express the α, β, δ, ɛ and ζ isoforms, with lower expression in tumour cells (Kahl‐Rainer et al. 1994; Perletti et al. 1996; 1998a, 1998b; Verstovsek et al. 1998). Each isoform may subserve a different role in colon cancer proliferation (1998, 1999; Weller et al. 1999) although their exact function is controversial (Scaglione‐Sewell et al. 1998). PKC‐α supports proliferation, a role also documented for PKC‐ɛ and ‐β in HT‐29 and HD3 and Caco‐2 cells (Cadoret et al. 1998; Weller et al. 1999; Davidson et al. 2000; Cesaro et al. 2001). PKC‐α was linked to ERK in bile‐acid‐treated AA/C1 adenoma cells (McMillan et al. 2003), but not in HCT‐116 cells (Qiao et al. 2000). PKC action appeared independent of ERK in our cells.

In conclusion, our data demonstrate that adherent SW620 cells proliferate more rapidly in response to a modest increase in pressure. In particular, the mitogenic effects of pressure require PKC, are independent of Src and are not blocked by phalloidin, precisely the reverse of the effects of pressure on adhesion. Thus, the effects of pressure on colon cancer cell proliferation are mediated by pathways different from the mitogenic effects of strain or the adhesion‐promoting effects of pressure on colon cancer cells. However, both greater adhesion and enhanced proliferation could facilitate colon cancer implantation and growth. Tumour progression in intact organisms is a highly complex process, but these results suggest that forces such as pressure and strain, acting through different pathways, may promote colon cancer progression.

ACKNOWLEDGEMENTS

Supported in part by NIH RO1 DK60771 (MDB).

REFERENCES

  1. Basson MD, Modlin IM, Madri JA (1992) Human enterocyte (Caco‐2) migration is modulated in vitro by extracellular matrix composition and epidermal growth factor. J. Clin. Invest. 90, 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Basson MD, Yu CF, Herden‐Kirchoff O, Ellermeier M, Sanders MA, Merrell RC, Sumpio BE (2000) Effects of increased ambient pressure on colon cancer cell adhesion. J. Cell Biochem. 78, 47. [DOI] [PubMed] [Google Scholar]
  3. Basson MD, Coppola CP (2002) Repetitive deformation and pressure activate small bowel and colonic mucosal tyrosine kinase activity in vivo . Metabolism 51, 1525. [DOI] [PubMed] [Google Scholar]
  4. Bissonnette M, Tien XY, Niedziela SM, Hartmann SC, Frawley BP Jr, Roy HK, Sitrin MD, Perlman RL, Brasitus TA (1994) 1,25(OH)2 vitamin D3 activates PKC‐α in Caco‐2 cells: a mechanism to limit secosteroid‐induced rise in [Ca2+]i. Am. J. Physiol. 267, G465. [DOI] [PubMed] [Google Scholar]
  5. Bouvy ND, Giuffrida MC, Tseng LN, Steyerberg EW, Marquet RL, Jeekel H, Bonjer HJ (1998) Effects of carbon dioxide pneumoperitoneum, air pneumoperitoneum, and gasless laparoscopy on body weight and tumor growth. Arch. Surg. 133, 652. [DOI] [PubMed] [Google Scholar]
  6. Brodribb AJ, Condon RE, Cowles V, Decosse JJ (1979) Effect of dietary fiber on intraluminal pressure and myoelectrical activity of left colon in monkeys. Gastroenterology 77, 70. [PubMed] [Google Scholar]
  7. Cadoret A, Baron‐Delage S, Bertrand F, Kornprost M, Groyer A, Gespach C, Capeau J, Cherqui G (1998) Oncogene‐induced up‐regulation of Caco‐2 cell proliferation involves IGF‐II gene activation through a protein kinase C‐mediated pathway. Oncogene 17, 877. [DOI] [PubMed] [Google Scholar]
  8. Cerda SR, Bissonnette M, Scaglione‐Sewell B, Lyons MR, Khare S, Mustafi R, Brasitus TA (2001) PKC‐δ inhibits anchorage‐dependent and ‐independent growth, enhances differentiation, and increases apoptosis in CaCo‐2 cells. Gastroenterology 120, 1700. [DOI] [PubMed] [Google Scholar]
  9. Cesaro P, Raiteri E, Demoz M, Castino R, Baccino FM, Bonelli G, Isidoro C (2001) Expression of protein kinase C β1 confers resistance to TNFα‐ and paclitaxel‐induced apoptosis in HT‐29 colon carcinoma cells. Int. J. Cancer. 93, 179. [DOI] [PubMed] [Google Scholar]
  10. Chien S, Li S, Shyy YJ (1998) Effects of mechanical forces on signal transduction and gene expression in endothelial cells. Hypertension 31, 162. [DOI] [PubMed] [Google Scholar]
  11. Chiquet M, Renedo AS, Huber F, Fluck M (2003) How do fibroblasts translate mechanical signals into changes in extracellular matrix production? Matrix Biol. 22, 73. [DOI] [PubMed] [Google Scholar]
  12. Dahn S, Schwalbach P, Wohleke F, Benner A, Kuntz C (2003) Influence of different gases used for laparoscopy (helium, carbon dioxide, room air, xenon) on tumor volume, proliferation, and apoptosis. Surg. Endosc. [DOI] [PubMed]
  13. Davidson LA, Brown RE, Chang WC, Morris JS, Wang N, Carrol RJ, Turner ND, Lupton JR, Chapkin RS (2000) Morphodensitometric analysis of protein kinase C β(II) expression in rat colon: modulation by diet and relation to in situ cell proliferation and apoptosis. Carcinogenesis 21, 1513. [PubMed] [Google Scholar]
  14. Dregelid E, Svendsen E (1988) Endothelial cell injury in human saphenous veins after manipulation and tweezer grasping. J. Cardiovasc. Surg. (Torino). 29, 464. [PubMed] [Google Scholar]
  15. Duncan RL, Turner CH (1995) Mechanotransduction and the functional response of bone to mechanical strain. Calcif. Tissue Int. 57, 344. [DOI] [PubMed] [Google Scholar]
  16. Esposito F, Chirico G, Gesualdi NM, Posadas I, Ammendola R, Russo T, Cirino G, Cimino F (2003) Protein kinase B activation by reactive oxygen species is independent of tyrosine kinase receptor phosphorylation and requires SRC activity. J. Biol. Chem. 278, 20828. [DOI] [PubMed] [Google Scholar]
  17. Golubovskaya VM, Gross S, Kaur AS, Wilson RI, Xu LH, Yang XH, Cance WG (2003) Simultaneous inhibition of focal adhesion kinase and SRC enhances detachment and apoptosis in colon cancer cell lines. Mol. Cancer Res. 1, 755. [PubMed] [Google Scholar]
  18. Graness A, Adomeit A, Heinze R, Wetzker R, Liebmann C (1998) A novel mitogenic signaling pathway of bradykinin in the human colon carcinoma cell line SW‐480 involves sequential activation of a Gq/11 protein, phosphatidylinositol 3‐kinase β, and protein kinase Cepsilon. J. Biol. Chem. 273, 32016. [DOI] [PubMed] [Google Scholar]
  19. Granger DN, Barrowman JA (1983) Microcirculation of the alimentary tract. II. Pathophysiology of edema. Gastroenterology 84, 1035. [PubMed] [Google Scholar]
  20. Gutt CN, Kim ZG, Gessmann T, Lorenz M, Paolucci V (2000) Hepatic tumor spread of colorectal cancer in a laparoscopic animal model. Surg. Endosc. 14, 448. [DOI] [PubMed] [Google Scholar]
  21. Gutt CN, Gessmann T, Schemmer P, Mehrabi A, Schmandra T, Kim ZG (2003) The impact of carbon dioxide and helium insufflation on experimental liver metastases, macrophages, and cell adhesion molecules. Surg. Endosc. 17, 1628. [DOI] [PubMed] [Google Scholar]
  22. Haier J, Gallick GE, Nicolson GL (2002) Src protein kinase pp60c‐src influences adhesion stabilization of HT‐29 colon carcinoma cells to extracellular matrix components under dynamic conditions of laminar flow. J. Exp. Ther. Oncol. 2, 237. [DOI] [PubMed] [Google Scholar]
  23. Han O, Sumpio BE, Basson MD (1998a) Mechanical strain rapidly redistributes tyrosine phosphorylated proteins in human intestinal Caco‐2 cells. Biochem. Biophys. Res. Commun. 250, 668. [DOI] [PubMed] [Google Scholar]
  24. Han O, Li GD, Sumpio BE, Basson MD (1998b) Strain induces Caco‐2 intestinal epithelial proliferation and differentiation via PKC and tyrosine kinase signals. Am. J. Physiol. 275, G534. [DOI] [PubMed] [Google Scholar]
  25. Han O, Takei T, Basson M, Sumpio BE (2001) Translocation of PKC isoforms in bovine aortic smooth muscle cells exposed to strain. J. Cell Biochem. 80, 367. [DOI] [PubMed] [Google Scholar]
  26. Hewett PJ, Texler ML, Anderson D, King G, Chatterton BE (1999) In vivo real‐time analysis of intraperitoneal radiolabeled tumor cell movement during laparoscopy. Dis. Colon Rectum 42, 868. [DOI] [PubMed] [Google Scholar]
  27. Hirokawa M, Miura S, Kishikawa H, Yoshida H, Nakamizo H, Higuchi H, Nakatsumi RC, Suzuki H, Saito H, Ishii H (2001) Loading of mechanical pressure activates mitogen‐activated protein kinase and early immediate gene in intestinal epithelial cells. Dig. Dis. Sci. 46, 1993. [DOI] [PubMed] [Google Scholar]
  28. Hishikawa K, Nakaki T, Marumo T, Hayashi M, Suzuki H, Kato R, Saruta T (1994) Pressure promotes DNA synthesis in rat cultured vascular smooth muscle cells. J. Clin. Invest. 93, 1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Hosokawa H, Aiuchi S, Kambe T, Hagiwara Y, Kubo T (2002) Mechanical stretch‐induced mitogen‐activated protein kinase activation is mediated via angiotensin and endothelin systems in vascular smooth muscle cells. Biol. Pharm. Bull. 25, 1588. [DOI] [PubMed] [Google Scholar]
  30. Hu Y, Bock G, Wick G, Xu Q (1998) Activation of PDGF receptor α in vascular smooth muscle cells by mechanical stress. FASEB J. 12, 1135. [DOI] [PubMed] [Google Scholar]
  31. Hua H, Goldberg HJ, Fantus IG, Whiteside CI (2001) High glucose‐enhanced mesangial cell extracellular signal‐regulated protein kinase activation and α1 (IV) collagen expression in response to endothelin‐1: role of specific protein kinase C isozymes. Diabetes 50, 2376. [DOI] [PubMed] [Google Scholar]
  32. Ikeda M, Takei T, Mills I, Kito H, Sumpio BE (1999) Extracellular signal‐regulated kinases 1 and 2 activation in endothelial cells exposed to cyclic strain. Am. J. Physiol. 276, H614. [DOI] [PubMed] [Google Scholar]
  33. Ingram AJ, Ly H, Thai K, Kang M, Scholey JW (1999) Activation of mesangial cell signaling cascades in response to mechanical strain. Kidney Int. 55, 476. [DOI] [PubMed] [Google Scholar]
  34. Ishida H, Idezuki Y, Yokoyama M, Nakada H, Odaka A, Murata N, Fujioka M, Hashimoto D (2001) Liver metastasis following pneumoperitoneum with different gases in a mouse model. Surg. Endosc. 15, 189. [DOI] [PubMed] [Google Scholar]
  35. Jacobi CA, Wenger FA, Ordemann J, Gutt C, Sabat R, Muller JM (1998) Experimental study of the effect of intra‐abdominal pressure during laparoscopy on tumour growth and port site metastasis. Br. J. Surg. 85, 1419. [DOI] [PubMed] [Google Scholar]
  36. Jacobi CA, Bonjer HJ, Puttick MI, O'Sullivan R, Lee SW, Schwalbach P, Tomita H, Kim ZG, Hewett P, Wittich P, Fleshman JW, Paraskeva P, Gessman T, Neuhaus SJ, Wildbrett P, Reymond MA, Gutt C, Whelan R (2002) Oncologic implications of laparoscopic and open surgery. Surg. Endosc. 16, 441. [DOI] [PubMed] [Google Scholar]
  37. Jasleen J, Shimoda N, Shen ER, Tavakkolizadeh A, Whang EE, Jacobs DO, Zinner MJ, Ashley SW (2000) Signaling mechanisms of glucagon‐like peptide 2‐induced intestinal epithelial cell proliferation. J. Surg. Res. 90, 13. [DOI] [PubMed] [Google Scholar]
  38. Jones RJ, Avizienyte E, Wyke AW, Owens DW, Brunton VG, Frame MC (2002) Elevated c‐Src is linked to altered cell‐matrix adhesion rather than proliferation in KM12C human colorectal cancer cells. Br. J. Cancer 87, 1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Kahl‐Rainer P, Karner‐Hanusch J, Weiss W, Marian B (1994) Five of six protein kinase C isoenzymes present in normal mucosa show reduced protein levels during tumor development in the human colon. Carcinogenesis 15, 779. [DOI] [PubMed] [Google Scholar]
  40. Kato H, Osajima A, Uezono Y, Okazaki M, Tsuda Y, Tanaka H, Oishi Y, Izumi F, Nakashima Y (1999) Involvement of PDGF in pressure‐induced mesangial cell proliferation through PKC and tyrosine kinase pathways. Am. J. Physiol. 277, F105. [DOI] [PubMed] [Google Scholar]
  41. Kawasaki T, Ogata M, Kawasaki C, Tomihisa T, Okamoto K, Shigematsu A (2001) Surgical stress induces endotoxin hyporesponsiveness and an early decrease of monocyte mCD14 and HLA‐DR expression during surgery. Anesth. Analg. 92, 1322. [DOI] [PubMed] [Google Scholar]
  42. Kawata Y, Mizukami Y, Fujii Z, Sakumura T, Yoshida K, Matsuzaki M (1998a) Applied pressure enhances cell proliferation through mitogen‐activated protein kinase activation in mesangial cells. J. Biol. Chem. 273, 16905. [DOI] [PubMed] [Google Scholar]
  43. Kawata Y, Fujii Z, Sakumura T, Kitano M, Suzuki N, Matsuzaki M (1998b) High pressure conditions promote the proliferation of rat cultured mesangial cells in vitro . Biochim. Biophys. Acta 1401, 195. [DOI] [PubMed] [Google Scholar]
  44. Kellow JE, Phillips SF (1987) Altered small bowel motility in irritable bowel syndrome is correlated with symptoms. Gastroenterology 92, 1885. [DOI] [PubMed] [Google Scholar]
  45. Kim ZG, Mehl C, Lorenz M, Gutt CN (2002) Impact of laparoscopic CO2‐insufflation on tumor‐associated molecules in cultured colorectal cancer cells. Surg. Endosc. 16, 1182. [DOI] [PubMed] [Google Scholar]
  46. Kuwada SK, Li X (2000) Integrin α5/β1 mediates fibronectin‐dependent epithelial cell proliferation through epidermal growth factor receptor activation. Mol. Biol. Cell 11, 2485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Li C, Hu Y, Mayr M, Xu Q (1999) Cyclic strain stress‐induced mitogen‐activated protein kinase (MAPK) phosphatase 1 expression in vascular smooth muscle cells is regulated by Ras/Rac‐MAPK pathways. J. Biol. Chem. 274, 25273. [DOI] [PubMed] [Google Scholar]
  48. Li C, Xu Q (2000) Mechanical stress‐initiated signal transductions in vascular smooth muscle cells. Cell Signal. 12, 435. [DOI] [PubMed] [Google Scholar]
  49. MacKenna DA, Dolfi F, Vuori K, Ruoslahti E (1998) Extracellular signal‐regulated kinase and c‐Jun NH2‐terminal kinase activation by mechanical stretch is integrin‐dependent and matrix‐specific in rat cardiac fibroblasts. J. Clin. Invest. 101, 301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Mattana J, Singhal PC (1995) Applied pressure modulates mesangial cell proliferation and matrix synthesis. Am. J. Hypertens. 8, 1112. [DOI] [PubMed] [Google Scholar]
  51. McMillan L, Butcher SK, Pongracz J, Lord JM (2003) Opposing effects of butyrate and bile acids on apoptosis of human colon adenoma cells: differential activation of PKC and MAP kinases. Br. J. Cancer 88, 748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Mellor H, Parker PJ (1998) The extended protein kinase C superfamily. Biochem. J. 332, 281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Miriel VA, Allen SP, Schriver SD, Prewitt RL (1999) Genistein inhibits pressure‐induced expression of c‐fos in isolated mesenteric arteries. Hypertension 34, 132. [DOI] [PubMed] [Google Scholar]
  54. Miyamoto S, Teramoto H, Gutkind JS, Yamada KM (1996) Integrins can collaborate with growth factors for phosphorylation of receptor tyrosine kinases and MAP kinase activation: roles of integrin aggregation and occupancy of receptors. J. Cell Biol. 135, 1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Moalli MR, Wang S, Caldwell NJ, Patil PV, Maynard CR (2001) Mechanical stimulation induces pp125(FAK) and pp60(src) activity in an in vivo model of trabecular bone formation. J. Appl. Physiol. 91, 912. [DOI] [PubMed] [Google Scholar]
  56. Osol G (1995) Mechanotransduction by vascular smooth muscle. J. Vasc. Res. 32, 275. [DOI] [PubMed] [Google Scholar]
  57. Pahlman L (1997) The problem of port‐site metastases after laparoscopic cancer surgery. Ann. Med. 29, 477. [DOI] [PubMed] [Google Scholar]
  58. Patankar SK, Larach SW, Ferrara A, Williamson PR, Gallagher JT, Dejesus S, Narayanan S (2003) Prospective comparison of laparoscopic vs. open resections for colorectal adenocarcinoma over a ten‐year period. Dis. Colon Rectum. 46, 601. [DOI] [PubMed] [Google Scholar]
  59. Perletti GP, Folini M, Lin HC, Mischak H, Piccinini F, Tashjian AH Jr (1996) Overexpression of protein kinase C∈ is oncogenic in rat colonic epithelial cells. Oncogene 12, 847. [PubMed] [Google Scholar]
  60. Perletti GP, Concari P, Brusaferri S, Marras E, Piccinini F, Tashjian AH Jr (1998) Protein kinase Cɛ is oncogenic in colon epithelial cells by interaction with the ras signal transduction pathway. Oncogene 16, 3345. [DOI] [PubMed] [Google Scholar]
  61. Perletti GP, Marras E, Concari P, Piccinini F, Tashjian AH Jr (1999) PKCδ acts as a growth and tumor suppressor in rat colonic epithelial cells. Oncogene 18, 1251. [DOI] [PubMed] [Google Scholar]
  62. Qiao D, Chen W, Stratagoules ED, Martinez JD (2000) Bile acid‐induced activation of activator protein‐1 requires both extracellular signal‐regulated kinase and protein kinase C signaling. J. Biol. Chem. 275, 15090. [DOI] [PubMed] [Google Scholar]
  63. Rice DC, Dobrian AD, Schriver SD, Prewitt RL (2002) Src autophosphorylation is an early event in pressure‐mediated signaling pathways in isolated resistance arteries. Hypertension 39, 502. [DOI] [PubMed] [Google Scholar]
  64. Sai X, Naruse K, Sokabe M (1999) Activation of pp60 (src) is critical for stretch‐induced orienting response in fibroblasts. J. Cell Sci. 112, 1365. [DOI] [PubMed] [Google Scholar]
  65. Scaglione‐Sewell B, Abraham C, Bissonnette M, Skarosi SF, Hart J, Davidson NO, Wali RK, Davis BH, Sitrin M, Brasitus TA (1998) Decreased PKC‐α expression increases cellular proliferation, decreases differentiation, and enhances the transformed phenotype of CaCo‐2 cells. Cancer Res. 58, 1074. [PubMed] [Google Scholar]
  66. Sekharam M, Nasir A, Kaiser HE, Coppola D (2003) Insulin‐like growth factor 1 receptor activates c‐SRC and modifies transformation and motility of colon cancer in vitro . Anticancer Res. 23, 1517. [PubMed] [Google Scholar]
  67. Shapiro PS, Evans JN, Davis RJ, Posada JA (1996) The seven‐transmembrane‐spanning receptors for endothelin and thrombin cause proliferation of airway smooth muscle cells and activation of the extracellular regulated kinase and c‐Jun NH2‐terminal kinase groups of mitogen‐activated protein kinases. J. Biol. Chem. 271, 5750. [DOI] [PubMed] [Google Scholar]
  68. Silecchia G, Perrotta N, Giraudo G, Salval M, Parini U, Feliciotti F, Lezoche E, Morino M, Melotti G, Carlini M, Rosato P, Basso N (2002) Abdominal wall recurrences after colorectal resection for cancer: results of the Italian registry of laparoscopic colorectal surgery. Dis. Colon Rectum. 45, 1172. [DOI] [PubMed] [Google Scholar]
  69. Sumpio BE, Widmann MD, Ricotta J, Awolesi MA, Watase M (1994) Increased ambient pressure stimulates proliferation and morphologic changes in cultured endothelial cells. J. Cell Physiol. 158, 133. [DOI] [PubMed] [Google Scholar]
  70. Sylvester PW, McIntyre BS, Gapor A, Briski KP (2001) Vitamin E inhibition of normal mammary epithelial cell growth is associated with a reduction in protein kinase C(α) activation. Cell Prolif. 34, 347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Thamilselvan V, Basson MD (2004) Pressure activates colon cancer cell adhesion by inside‐out focal adhesion complex and cytoskeletal signaling. Gastroenterology 126, 8. [DOI] [PubMed] [Google Scholar]
  72. Thamilselvan V, Nunkumar N, Basson MD (2004) Pressure stimulates colon cancer cell adhesion by a pathway involving Src, PI3 Kinase, Akt, and FAK. Gastroenterology 126, A–395. [Google Scholar]
  73. Toker A (1998) Signaling through protein kinase C. Front. Biosci. 3, D1134. [DOI] [PubMed] [Google Scholar]
  74. Tsuda Y, Okazaki M, Uezono Y, Osajima A, Kato H, Okuda H, Oishi Y, Yahiro A, Nakashima Y (2002) Activation of extracellular signal‐regulated kinases is essential for pressure‐induced proliferation of vascular smooth muscle cells. Eur. J. Pharmacol. 446, 15. [DOI] [PubMed] [Google Scholar]
  75. Verstovsek G, Byrd A, Frey MR, Petrelli NJ, Black JD (1998) Colonocyte differentiation is associated with increased expression and altered distribution of protein kinase C isozymes. Gastroenterology 115, 75. [DOI] [PubMed] [Google Scholar]
  76. Vouyouka AG, Salib SS, Cala S, Marsh JD, Basson MD (2003) Chronic high pressure potentiates the antiproliferative effect and abolishes contractile phenotypic changes caused by endothelial cells in cocultured smooth muscle cells. J. Surg. Res. 110, 344. [DOI] [PubMed] [Google Scholar]
  77. Walker J, Criddle LM (2003) Pathophysiology and management of abdominal compartment syndrome. Am. J. Crit. Care. 12, 367–371. [PubMed] [Google Scholar]
  78. Walsh MF, Thamilselvan V, Grotelueschen R, Farhana L, Basson M (2003) Absence of adhesion triggers differential FAK and SAPKp38 signals in SW620 human colon cancer cells that may inhibit adhesiveness and lead to cell death. Cell Physiol. Biochem. 13, 135. [DOI] [PubMed] [Google Scholar]
  79. Watase M, Awolesi MA, Ricotta J, Sumpio BE (1997) Effect of pressure on cultured smooth muscle cells. Life Sci. 61, 987. [DOI] [PubMed] [Google Scholar]
  80. Weller SG, Klein IK, Penington RC, Karnes WE Jr (1999) Distinct protein kinase C isozymes signal mitogenesis and apoptosis in human colon cancer cells. Gastroenterology 117, 848. [DOI] [PubMed] [Google Scholar]
  81. Wesselman JP, Dobrian AD, Schriver SD, Prewitt RL (2001) Src tyrosine kinases and extracellular signal‐regulated kinase 1/2 mitogen‐activated protein kinases mediate pressure‐induced c‐fos expression in cannulated rat mesenteric small arteries. Hypertension 37, 955. [DOI] [PubMed] [Google Scholar]
  82. Wu JS, Brasfield EB, Guo LW, Ruiz M, Connett JM, Philpott GW, Jones J, Fleshman JW (1997) Implantation of colon cancer at trocar sites is increased by low pressure pneumoperitoneum. Surgery 122, 1. [DOI] [PubMed] [Google Scholar]
  83. Xu Q, Schett G, Li C, Hu Y, Wick G (2000) Mechanical stress‐induced heat shock protein 70 expression in vascular smooth muscle cells is regulated by Rac and Ras small G proteins but not mitogen‐activated protein kinases. Circ. Res. 86, 1122. [DOI] [PubMed] [Google Scholar]
  84. Zhang J, Li W, Sanders MA, Sumpio BE, Panja A, Basson MD (2003) Regulation of the intestinal epithelial response to cyclic strain by extracellular matrix proteins. FASEB J. 17, 926. [DOI] [PubMed] [Google Scholar]

Articles from Cell Proliferation are provided here courtesy of Wiley

RESOURCES