Nitric oxide (NO), which plays a role in the posttranslational modification of proteins, exhibits tumoricidal activity. However, the mechanism remains largely unclear. We investigated whether the regulation of insulin receptor substrate (IRS)-1 protein expression and insulin/insulin-like growth factor (IGF) signaling by NO is involved in the proliferation and invasion of pancreatic cancer cells. NO donor inhibited insulin/IGF-I–stimulated phosphorylation of insulin receptor/IGF-I receptor, IRS-1, Akt/PKB, and glycogen synthase kinase-3β along with decreased expression of IRS-1 protein in MIAPaCa-2 cells, whereas NO donor enhanced the phosphorylation of extracellular signal-regulated kinase-1/2. In contrast, a selective inducible nitric oxide synthase inhibitor, 1400W, upregulated the expression of IRS-1 protein and the phosphorylation of IRS-1, Akt/PKB, and glycogen synthase kinase-3β, along with enhanced proliferation and invasion of Panc-1 cells expressing inducible nitric oxide synthase protein. NO donor induced IRS-1 protein reduction through increased ubiquitination and degradation. For the detection of the site responsible for NO-induced ubiquitination, IRS-1 deletion mutant genes were transfected and overexpressed in MIAPaCa-2 cells. The results indicate that the COOH terminus of the IRS-1 protein is required for NO donor–induced ubiquitination and protein degradation. Cells stably transfected with COOH-terminal deletion mutants of IRS-1 exhibited reduced IGF signaling and cell proliferation compared with vector alone–transfected cells, with no influence of NO on IGF signaling and invasion, although stable transfectants with full-length IRS-1 protein exhibited remarkable NO-induced reduction in IGF signaling, cell proliferation, and invasion. These findings indicate that NO inhibits the proliferation and invasion of pancreatic cancer cells, at least in part, through upregulation of IRS-1 protein degradation and resultant downregulation of the insulin/IGF-I-Akt pathway. Mol Cancer Res; 8(8); 1152–63. ©2010 AACR.

Insulin/insulin-like growth factor (IGF) signals play a key role in cancer proliferation and invasion (1-3). Insulin/IGF-I and IGF-II bind to insulin/IGF-I receptors and then phosphorylate the tyrosine of the cognate receptors. Insulin receptor substrate (IRS)-1, an adaptor protein, exists mainly in the cytosol and binds to phosphorylated insulin receptor and IGF-I receptor (IGF-IR), resulting in the phosphorylation and activation of IRS-1. IRS-1 transduces phosphatidylinositol-3 kinase (PI3K), which in turn activates further downstream components, including Akt/PKB and glycogen synthase kinase-3β (GSK-3β). Alternatively, phosphorylated and activated IRS-1 can also bind to another adaptor protein, Grb-2, which activates mitogen-activated protein kinase, another major insulin/IGF signaling cascade parallel to the PI3K-Akt/PKB pathway (4, 5). IRS-1 protein expression was detected in several types of cancer, including pancreatic cancer, breast cancer, and hepatic cell carcinoma (6, 7). Thus, insulin/IGF signal has been considered to play a major role not only in metabolic actions, including stimulation of glucose uptake and synthesis of glycogen and protein, but also in cancer viability including proliferation and invasion. IRS-1 is a key molecule in insulin/IGF signaling, which transduces a signal from the insulin receptor/IGF-IR to the PI3K and mitogen-activated protein kinase pathways (8). However, the mechanism of the regulation of IRS-1 expression and insulin/IGF signals in cancer cells remains unclear.

Recent studies have shown that nitric oxide (NO) plays a role in the posttranslational modification of proteins (9-12). NO is produced by three distinct genes: neuronal and endothelial nitric oxide synthases (nNOS and eNOS) and inducible nitric oxide synthases (iNOS; refs. 13, 14). In contrast to the activities of nNOS and eNOS that are tightly regulated by calcium-dependent calmodulin binding, iNOS does not require calcium ion or posttranslational modification for its activity. Therefore, iNOS expression is associated with prolonged, exaggerated NO generation up to > 1,000-fold compared with nNOS and eNOS (15, 16). Although iNOS expression is increased in macrophages and endothelial cells by various stimuli, including acute inflammation, recent studies revealed that iNOS is expressed even in normal conditions in many tissues, including skeletal muscle and cancer (17, 18). In the skeletal muscle of diabetic mice, NO was found to play a key role for insulin resistance (17, 19). Controversial results have been reported about the roles of NO in cancer. Recent articles reported that endogenous NO promotes oncogenesis and angiogenesis in various cancers (20, 21). In contrast, other studies have shown that NO inhibits cell proliferation and induces apoptosis in various cells including cancer cells, in vitro and in vivo (22-28). These studies suggest that NO can act either as a tumor suppressor or as a tumor enhancer depending on cell type and the level of NO in the cells. However, the molecular mechanism underlying the inhibitory effects of NO on cancer viability remains unclear.

In this study, we show that NO induces proteasome-dependent IRS-1 protein degradation, and that the regulation of IRS-1 expression and insulin/IGF signaling by NO is involved in NO donor–mediated inhibition of the proliferative and invasive activities of pancreatic cancer cells. These data provide new insight into the molecular basis underlying the regulation of cancer viability by NO.

Materials

MG132, S-nitrosoglutathione (GSNO), S-nitroso-N-acetylpenicillamine (SNAP), and l-NAME were purchased from Calbiochem. A highly selective NO inhibitor, 1400W, was purchased from Cayman Chemical. Recombinant IGF-I was purchased from Peprotech. Insulin and anti-Flag antibody were purchased from Sigma. Anti–phospho-Tyr1135/1136 IGF-IRβ, anti–phospho-Ser473 Akt/PKB, anti–phospho-Ser9 GSK-3β, anti–phospho-extracellular signal-regulated kinase (Erk)-1/2, anti–Akt/PKB, anti–GSK-3β, and anti–Erk-1/2 antibodies were purchased from Cell Signaling Technology. Anti–IRS-1 antibody was purchased from Upstate Biotechnology. Anti-phosphotyrosine and anti-ubiquitin antibodies were purchased from Santa Cruz Biotechnology. Anti-iNOS antibody was purchased from Becton Dickinson. Geneticin (G-418) was purchased from Life Technologies, Inc.

Cell culture

MIAPaCa-2, Panc-1, MCF-7, MB 468, and HepG2 cells were obtained from the American Type Culture Collection and were maintained in DMEM supplemented with 10% fetal bovine serum (FBS) at 37°C under a humidified atmosphere of 5% CO2.

Nitrite assay

The concentration of nitrite was determined using a 2,3-diaminonaphthalene kit (Dojindo Laboratories; ref. 29). Briefly, MIAPaCa-2 cells and Panc-1 cells were incubated in phenol red–free DMEM. The medium was centrifuged at 1,000 × g for 15 minutes and the supernatant was placed on 96-well microplates. 2,3-Diaminonaphthalene was added to each well and incubated at room temperature for 15 minutes, and then stop solution was added. The fluorescence intensity of each well at 450 nm (excitation 360 nm) was measured with a fluorescence microplate reader. The concentrations of nitrite were calculated by reference to a standard curve of nitrite.

Constructs

cDNA construct of full-length human IRS-1 (hIRS) wild-type (WT) was produced by reverse transcription-PCR. ssDNA was used as a template, which was produced by reverse transcription using oligo-dT primer from mRNA isolated from HepG2 cells, and then PCR of the single-stranded hIRS-1 WT DNA was performed. For the cDNA construct of the full-length hIRS-1 WT gene, the sense primer was 5′-ATGGCGAGCCCTCCGGAGAGCGAT-3′ and the antisense primer was 5′-CTGACGGTCCTCTGGCTGCTTCTGGAAAC-3′. The PCR was begun with denaturation at 94°C for 5 minutes followed by 35 cycles of denaturation at 94°C for 45 seconds, annealing at 60°C for 1 minute, and extension at 72°C each for 3 minutes 30 seconds with a final extension at 72°C for 10 minutes. AccuPrime Pfx (Invitrogen) was used as the DNA polymerase.

For the production of deletion mutants (DM), PCR was performed using full-length hIRS-1 as a template. The primers used were as follows: DM1, 5′-ATGGCGAGCCCTCCGGAGAGCGAT-3′ (sense) and 5′-GTCCCCCACTGGTGACATGTTCA-3′ (antisense); DM2, 5′-ATGGCGAGCCCTCCGGAGAGCGAT-3′ (sense) and 5′-CTCATCACTCATGGCCCGCATGGC-3′ (antisense); and DM3, 5′-ATGTCCAACACCAGCAGCCCCTCCGACTG-3′ (sense) and 5′-CTGACGGTCCTCTGGCTGCTTCTGGAAAC-3′ (antisense). The PCR was begun with denaturation at 94°C for 5 minutes followed by 30 cycles of denaturation at 94°C for 45 seconds, annealing at 60°C for 1 minute, and extension at 72°C each for 3 minutes with a final extension at 72°C for 10 minutes. AccuPrime Pfx (Invitrogen) was used as the DNA polymerase.

All PCR products were treated with Taq DNA polymerase and then subcloned into pCR2.1-TOPO vector (Invitrogen). Subcloned pCR2.1-TOPO vector was digested by EcoRI and then genes were inserted into pCMV Tag 4A vector (Stratagene), which is a mammalian expression vector, and then treated with EcoRI and CIAP. An illustration of the PCR products of full-length IRS-1, DM1, DM2, and DM3 is provided (see Fig. 2C).

FIGURE 1.

A and B, concentration of nitrite in medium. A, MIAPaCa-2 cells were cultured with GSNO (1 mmol/L) or SNAP (1 mmol/L) for 0, 6, and 24 h. The concentration of nitrite in supernatant was determined. *, P < 0.05, compared with control. B, Panc-1 cells were cultured with 1400W (100 μmol/L) for 0, 3, 6, and 24 h. The concentration of nitrite in supernatant was determined. *, P < 0.05, compared with 0 h. ++, P < 0.05, compared with control. C to E, regulation of insulin/IGF signal by NO donor in MIAPaCa-2 cells. MIAPaCa-2 cells, which were incubated in DMEM with 10% FBS overnight and grown to 80% confluence, were used. After incubation with SNAP (1 mmol/L) or GSNO (500 μmol/L) under serum starvation for 6 h, the cells were incubated with insulin (100 nmol/L) or IGF-I (25 nmol/L) for 5 min and then harvested. Cell lysates were subjected to immunoprecipitation (IP) associated with immunoblotting (IB) or immunoblotting. All experiments were repeated three times and the same results were obtained.

FIGURE 1.

A and B, concentration of nitrite in medium. A, MIAPaCa-2 cells were cultured with GSNO (1 mmol/L) or SNAP (1 mmol/L) for 0, 6, and 24 h. The concentration of nitrite in supernatant was determined. *, P < 0.05, compared with control. B, Panc-1 cells were cultured with 1400W (100 μmol/L) for 0, 3, 6, and 24 h. The concentration of nitrite in supernatant was determined. *, P < 0.05, compared with 0 h. ++, P < 0.05, compared with control. C to E, regulation of insulin/IGF signal by NO donor in MIAPaCa-2 cells. MIAPaCa-2 cells, which were incubated in DMEM with 10% FBS overnight and grown to 80% confluence, were used. After incubation with SNAP (1 mmol/L) or GSNO (500 μmol/L) under serum starvation for 6 h, the cells were incubated with insulin (100 nmol/L) or IGF-I (25 nmol/L) for 5 min and then harvested. Cell lysates were subjected to immunoprecipitation (IP) associated with immunoblotting (IB) or immunoblotting. All experiments were repeated three times and the same results were obtained.

Close modal

Cell transfection

MIAPaCa-2 cells were transfected with pCMV Tag 4A/IRS-1 (full-length), pCMV Tag 4A/IRS-1 DM1, pCMV Tag 4A/IRS-1 DM2 (as a dominant negative), pCMV Tag 4A/IRS-1 DM3, and pCMV Tag 4A vector alone using Lipofectamine 2000 (Invitrogen). Twenty-four hours after transfection, the cells were used for the assay of IRS-1 protein degradation and detection of ubiquitination. In addition, the cells transfected with protein expression vectors were incubated with G418 for the selection of transfected gene–expressing cells for 2 weeks and then cloned. The clone that highly expresses the transfected gene was selected and then used for the assays.

Cell lysis

Cell lysates were obtained as previously described (19). Briefly, cells were lysed with cell lysis buffer [50 mmol/L Tris-HCl (pH 7.6), 150 mmol/L NaCl, 10 mmol/L sodium fluoride, 2 mmol/L sodium vanadate, 1 mmol/L phenylmethylsulfonyl fluoride, 10 μg/mL aprotinin, 10 μg/mL leupeptin, 1 mmol/L DTT, and 1% NP40]. Following incubation on ice for 30 minutes, lysate samples were centrifuged at 13,000 × g for 30 minutes. Aliquots of the supernatant containing equal amounts of protein, determined using the Lowry assay, were subjected to immunoprecipitation followed by SDS-PAGE.

Immunoprecipitation and immunoblotting

Immunoprecipitation was done by incubating the lysates with antibody at 4°C for 18 hours. The immune complexes were collected by incubation with protein A/G-agarose beads for 1.5 hours at 4°C, washed three times with wash buffer [50 mmol/L Tris-HCl (pH 7.6), 150 mmol/L NaCl, 10 mmol/L sodium fluoride, 2 mmol/L sodium vanadate, 1 mmol/L phenylmethylsulfonyl fluoride, 10 μg/mL aprotinin, 10 μg/mL leupeptin, 1 mmol/L DTT, and 0.1% NP40], and boiled in Laemmli sample buffer.

Cell lysates containing equal amounts of either protein or immunoprecipitates were subjected to 7.5% or 10% SDS-PAGE after the addition of Laemmli sample buffer and boiling for 5 minutes. After transferring electrophoretically onto nitrocellulose membrane (Bio-Rad), the membranes were blocked in 5% nonfat dried milk for 2 hours at room temperature and incubated with primary antibody for 2 hours at room temperature or overnight at 4°C. This was followed by incubation with secondary antibody conjugated with horseradish peroxidase for 1 hour at 4°C. Western blotting chemiluminescence luminol reagent (Perkin-Elmer) was used to visualize the blots. Bands of interest were scanned by using Power Look (UMAX Technologies) and were quantified by using NIH Image 1.62 software (NTIS).

Cell proliferation assay

To assess cell growth, MIAPaCa-2 and Panc-1 cells were seeded in 24-well plates (2 × 104 per well) in triplicates. The cells were preincubated for 24 hours in DMEM in the presence of 1% glutamine, 10% FBS, and antibiotics (1% penicillin and streptomycin sulfate) at 37°C under a humidified atmosphere of 5% CO2 and were then exposed to various reagents for 24, 48, and 72 hours. The cells were collected and counted after being stained with 0.4% trypan blue (Sigma-Aldrich).

Cell invasion assay

The in vitro invasive potential of MIAPaCa-2 cells and Panc-1 cells was determined using BioCoat Matrigel Invasion Chambers (Becton Dickinson). According to the manufacturer's instructions, the Matrigel was hydrated with 0.5 mL of DMEM (serum-free) and incubated for 2 hours in humidified tissue culture incubator at 37°C under a humidified atmosphere of 5% CO2. Evaluation of cell migration used control inserts with 8.0-μm pores in 24-well plates. After detaching the cells with 0.25% trypsin and counting, they were diluted to 1.0 × 105 cells/mL in medium containing 1% FBS. A total of 0.5 × 105 cells per well were placed on the top chamber of the insert above the medium containing chemoattractants, which was placed in the lower chamber. After incubation for 22 hours under a humidified atmosphere of 5% CO2, the cells on the upper surface of the membrane were removed with a cotton swab and the migrated cells on the lower side of the membrane were fixed and stained using Diff-Quick (American Scientific Products) and counted in 10 random fields at ×100 magnification (30).

Statistical analysis

Data were compared using one-way ANOVA followed by Fisher's protected least significant difference test. P < 0.05 was considered statistically significant. All values were expressed as mean ± SEM.

NO donors increase the concentration of nitrite (NO2−) and 1400W inhibits nitrite production in the medium in which cancer cells are cultured

NO donors added to the culture medium provided NO to cancer cells. Cancer cells expressing iNOS protein can produce NO in normal conditions. We determined the concentration of nitrite, a metabolite of NO, in the culture medium. Both GSNO and SNAP, NO donors, augmented the concentration of nitrite in the culture medium in which MIAPaCa-2 cells were cultured for 6 and 24 hours (Fig. 1A). The concentration of nitrite was elevated with time in the medium in which Panc-1 cells were cultured. 1400W, a highly selective iNOS inhibitor, inhibited nitrite production in the culture medium at 3 and 6 hours (Fig. 1B).

NO influences insulin/IGF signals in MIAPaCa-2 cells

The stimulation of insulin or IGF-I resulted in remarkable tyrosine phosphorylation of insulin receptor, IGF-IR, and IRS-1; phosphorylation of Akt/PKB at Ser473 and GSK-3β at Ser9; and phosphorylation of Erk-1/2 in MIAPaCa-2 cells. SNAP, a NO donor, inhibited insulin-stimulated tyrosine phosphorylation of insulin receptor and IRS-1, phosphorylation of Akt/PKB at Ser473, and phosphorylation of GSK-3β at Ser9. In addition, SNAP inhibited IGF-I–stimulated tyrosine phosphorylation of IGF-IR and IRS-1, phosphorylation of Akt/PKB at Ser473, and phosphorylation of GSK-3β at Ser9. Furthermore, SNAP reduced IRS-1 protein expression, although this did not alter the expression of other proteins, including IGF-IR, Akt/PKB, GSK-3β, and Erk-1/2, in downstream of IGF signaling, as well as β-actin expression. On the other hand, SNAP induced the phosphorylation of Erk-1/2 without stimulation of insulin/IGF-I and enhanced insulin/IGF-I–stimulated phosphorylation of Erk-1/2; however, SNAP did not influence Erk-1/2 protein expression in MIAPaCa-2 cells (Fig. 1C and D). GSNO, a NO donor, inhibited IGF-I–induced phosphorylation of IGF-IR, IRS-1, and Akt/PKB, but enhanced the phosphorylation of Erk-1/2 as well as SNAP (Fig. 1E).

NO downregulates IRS-1 protein expression through proteasome-mediated degradation in MIAPaCa-2 cells

GSNO inhibited IRS-1 protein expression in MCF-7 as well as MIAPaCa-2 cells in a dose-dependent manner, but did not influence IRS-1 protein expression in MB 468 and Panc-1 cells, which exhibited less IRS-1 protein expression (Fig. 2A). The proteasome inhibitor MG132 completely reversed the reduction of IRS-1 protein expression by NO donors (GSNO and SNAP) in MIAPaCa-2 cells. Neither GSNO nor MG132 influenced GSK-3β and β-actin protein expression (Fig. 2B).

FIGURE 2.

NO donor–induced reduction of IRS-1 protein expression through proteasome-mediated degradation in MIAPaCa-2 cells. A, MB 468, MIAPaCa-2, Panc-1, and MCF-7 cells were incubated under various concentrations of GSNO in DMEM with 10% FBS for 24 h. After incubation, cells were harvested and cell lysates were subjected to immunoblotting for detection of IRS-1 protein expression. All experiments were repeated three times and the same results were obtained. B, MIAPaCa-2 cells were incubated in DMEM with 10% FBS overnight. Subsequently, MG132 (3.3 μmol/L), a proteasome inhibitor, was added 30 min before (a) GSNO (500 μmol/L) or (b) SNAP (1 mmol/L) administration, followed by incubation for 24 h. Cell lysates were subjected to immunoblotting for detection of IRS-1 protein expression. Data shown are the results of triplicate experiments. Bars, SEM. *, P < 0.05, compared with control; ++, P < 0.05, compared with GSNO only. C, schematic representation of the functional domains of hIRS-1 protein and the structural organization of the deletion mutants. PH and PTB indicate pleckstrin homology and phosphotyrosine binding regions, respectively. The various hIRS-1 deletion mutants contain a FLAG epitope (DYKDDDDK) at the COOH terminus. D, MIAPaCa-2 cells were transfected with full-length IRS-1, IRS-1 DM1, IRS-1 DM2, and IRS-1 DM3 using Lipofectamine 2000, followed by incubation with GSNO (500 μmol/L) for 24 h. Each overexpressed protein was detected by immunoblotting using anti-Flag antibody. E, MIAPaCa-2 cells were transfected with full-length IRS-1, IRS-1 DM1, IRS-1 DM2, and IRS-1 DM3. MIAPaCa-2 cells were incubated in DMEM with 10% FBS overnight, and then MG132 (3.3 μmol/L), a proteasome inhibitor, was added 30 min before SNAP (1 mmol/L) administration, followed by incubation for 4 h. For the detection of ubiquitination, cell lysates were subjected to immunoprecipitation with anti-Flag antibody, followed by immunoblotting with anti-ubiquitin. All experiments were repeated three times and the same results were obtained.

FIGURE 2.

NO donor–induced reduction of IRS-1 protein expression through proteasome-mediated degradation in MIAPaCa-2 cells. A, MB 468, MIAPaCa-2, Panc-1, and MCF-7 cells were incubated under various concentrations of GSNO in DMEM with 10% FBS for 24 h. After incubation, cells were harvested and cell lysates were subjected to immunoblotting for detection of IRS-1 protein expression. All experiments were repeated three times and the same results were obtained. B, MIAPaCa-2 cells were incubated in DMEM with 10% FBS overnight. Subsequently, MG132 (3.3 μmol/L), a proteasome inhibitor, was added 30 min before (a) GSNO (500 μmol/L) or (b) SNAP (1 mmol/L) administration, followed by incubation for 24 h. Cell lysates were subjected to immunoblotting for detection of IRS-1 protein expression. Data shown are the results of triplicate experiments. Bars, SEM. *, P < 0.05, compared with control; ++, P < 0.05, compared with GSNO only. C, schematic representation of the functional domains of hIRS-1 protein and the structural organization of the deletion mutants. PH and PTB indicate pleckstrin homology and phosphotyrosine binding regions, respectively. The various hIRS-1 deletion mutants contain a FLAG epitope (DYKDDDDK) at the COOH terminus. D, MIAPaCa-2 cells were transfected with full-length IRS-1, IRS-1 DM1, IRS-1 DM2, and IRS-1 DM3 using Lipofectamine 2000, followed by incubation with GSNO (500 μmol/L) for 24 h. Each overexpressed protein was detected by immunoblotting using anti-Flag antibody. E, MIAPaCa-2 cells were transfected with full-length IRS-1, IRS-1 DM1, IRS-1 DM2, and IRS-1 DM3. MIAPaCa-2 cells were incubated in DMEM with 10% FBS overnight, and then MG132 (3.3 μmol/L), a proteasome inhibitor, was added 30 min before SNAP (1 mmol/L) administration, followed by incubation for 4 h. For the detection of ubiquitination, cell lysates were subjected to immunoprecipitation with anti-Flag antibody, followed by immunoblotting with anti-ubiquitin. All experiments were repeated three times and the same results were obtained.

Close modal

To further investigate IRS-1 protein degradation induced by NO donor, cDNA constructs of full-length IRS-1, IRS-1 DM1, IRS-1 DM2, and IRS-1 DM3 were produced and subcloned in mammalian expression vectors (Fig. 2C). MIAPaCa-2 cells were transfected with these expression vectors. GSNO reduced full-length IRS-1, IRS-1 DM1, and IRS-1 DM3 protein expression, although GSNO did not alter IRS-1 DM2 and β-actin protein expression (Fig. 2D). Ubiquitination of WT and mutated IRS-1 was detected by immunoprecipitation using anti-Flag antibody followed by immunoblotting with anti-ubiquitin. SNAP induced the ubiquitination of full-length IRS-1, IRS-1 DM1, and IRS-1 DM3, but did not induce the ubiquitination of IRS-1 DM2 (Fig. 2E). These results indicate that NO donor is capable of inducing ubiquitination at multiple sites in the COOH terminus of the IRS-1 protein.

iNOS protein is expressed in Panc-1 cells and 1400W upregulates IRS-1 protein expression and the IRS-1-Akt pathway in Panc-1 cells

iNOS protein was detected by immunoblotting in Panc-1 cells. IRS-1 protein expression was significantly increased by 1400W in a dose-dependent manner. However, Akt/PKB, β-actin, and Erk-1/2 protein expression was not altered in Panc-1 cells (Fig. 3A). l-NAME, a nonselective NOS inhibitor, increased IRS-1 expression in Panc-1 cells, but it was not remarkable compared with the increase by 1400W (Fig. 3B). GSNO inhibited IRS-1 protein expression, upregulated by 1400W in Panc-1 cells (Fig. 3C). On the other hand, 1400W did not alter IRS-1 protein expression in MCF-7, which expresses high level of the protein (Supplementary Figure). 1400W enhanced IGF-I–stimulated tyrosine phosphorylation of IRS-1, phosphorylation of Akt/PKB at Ser473, and phosphorylation of GSK-3β at Ser9 in Panc-1 cells. In contrast, 1400W did not alter IGF-I–stimulated phosphorylation of Erk-1/2 (Fig. 3D). These results indicate that endogenous NO produced by iNOS plays a role in insulin/IGF-I signaling.

FIGURE 3.

1400W, a highly selective iNOS inhibitor, upregulates IRS-1 protein expression and insulin/IGF signal in Panc-1 cells, which express iNOS protein. A, Panc-1 cells were incubated with various concentrations of 1400W for 24 h. Cell lysates were subjected to immunoblotting for the detection of expression of several proteins. Data shown are the results of triplicate of experiments. Bars, SEM. *, P < 0.05, compared with control. B, Panc-1 cells were incubated with various concentrations of l-NAME for 24 h. Cell lysates were subjected to immunoblotting for the detection of expression of several proteins. All experiments were repeated three times and the same results were obtained. C, Panc-1 cells were incubated with 1400W (100 μmol/L) and GSNO (500 μmol/L) for 24 h. Cell lysates were subjected to immunoblotting for the detection of expression of several proteins. All experiments were repeated three times and the same results were obtained. D, Panc-1 cells were incubated in DMEM with 10% FBS overnight and grown to 80% confluence. After incubation with 1400W (100 μmol/L) under serum starvation for 6 h, Panc-1 cells were incubated with IGF-I (25 nmol/L) for 5 min and then harvested. For the detection of IRS-1 phosphorylation, cell lysates were subjected to immunoprecipitation with anti–IRS-1 antibody followed by immunoblotting with anti-phosphotyrosine (PY) antibody. Then, cell lysates were directly subjected to immunoblotting. Data shown are the results of triplicate experiments. Bars, SEM. *, P < 0.05, compared with IGF-I only.

FIGURE 3.

1400W, a highly selective iNOS inhibitor, upregulates IRS-1 protein expression and insulin/IGF signal in Panc-1 cells, which express iNOS protein. A, Panc-1 cells were incubated with various concentrations of 1400W for 24 h. Cell lysates were subjected to immunoblotting for the detection of expression of several proteins. Data shown are the results of triplicate of experiments. Bars, SEM. *, P < 0.05, compared with control. B, Panc-1 cells were incubated with various concentrations of l-NAME for 24 h. Cell lysates were subjected to immunoblotting for the detection of expression of several proteins. All experiments were repeated three times and the same results were obtained. C, Panc-1 cells were incubated with 1400W (100 μmol/L) and GSNO (500 μmol/L) for 24 h. Cell lysates were subjected to immunoblotting for the detection of expression of several proteins. All experiments were repeated three times and the same results were obtained. D, Panc-1 cells were incubated in DMEM with 10% FBS overnight and grown to 80% confluence. After incubation with 1400W (100 μmol/L) under serum starvation for 6 h, Panc-1 cells were incubated with IGF-I (25 nmol/L) for 5 min and then harvested. For the detection of IRS-1 phosphorylation, cell lysates were subjected to immunoprecipitation with anti–IRS-1 antibody followed by immunoblotting with anti-phosphotyrosine (PY) antibody. Then, cell lysates were directly subjected to immunoblotting. Data shown are the results of triplicate experiments. Bars, SEM. *, P < 0.05, compared with IGF-I only.

Close modal

NO inhibits the proliferation of cancer cell lines and IRS-1 protein expression is associated with cancercell proliferation

Cell proliferation assay revealed that 100 and 500 μmol/L GSNO inhibited the proliferation of MIAPaCa-2 and MCF-7 cells in culture medium containing 10% FBS (Fig. 4A). Mammalian cell expression vectors, pCMV Tag 4/IRS-1 (full-length), pCMV Tag 4/IRS-1 DM2, and pCMV Tag 4A vector alone, were transfected into MIAPaCa-2 and incubated with G418 for the selection of protein-expressing cells for more than 14 days. Subsequently, the cells expressing high levels of full-length IRS-1 protein or IRS-1 DM2 protein were cloned. Phosphorylation of Akt/PKB at Ser473 and of GSK-3β at Ser9 by stimulation of IGF-I were reduced in IRS-1 DM2–transfected cells in comparison with vector alone– or full-length IRS-1–transfected cells. SNAP failed to reduce the phosphorylation of Akt/PKB at Ser473 and GSK-3β at Ser9 by stimulation of IGF-I in IRS-1 DM2–transfected cells, but reduced their phosphorylation in vector alone– or full-length IRS-1–transfected cells. No difference was observed between these cell lines in terms of Akt/PKB and GSK-3β protein expression (Fig. 4B).

FIGURE 4.

NO donor inhibits proliferation through downregulation of the IGF-Akt pathway in MIAPaCa-2 cells. A, MIAPaCa-2 and MCF-7 cells were incubated with GSNO (100 and 500 μmol/L) in medium containing 10% FBS for 24, 48, and 72 h. B, MIAPaCa-2 cells were transfected with vector only (as a mock), full-length IRS-1, and IRS-1 DM2 (as a dominant negative), followed by incubation with G418. Stable transfected cells were incubated in DMEM with 10% FBS overnight and grown to 80% confluence. After incubation with SNAP (1 mmol/L) under serum starvation for 6 h, the cells were incubated with IGF-I (25 nmol/L) for 5 min and then harvested. The cell lysates were subjected to immunoblotting. All experiments were repeated three times and the same results were obtained. C, stable transfected MIAPaCa-2 cells were incubated with GSNO (200 μmol/L) in medium containing 10% FBS for 24, 48, and 72 h. D, stable transfected cells (2 × 104 per well) were incubated with GSNO (200 μmol/L) in medium with or without IGF-I (100 nmol/L; serum-free) for 48 h. *, P < 0.01, compared with control; ++, P < 0.05, compared with IGF-I only.

FIGURE 4.

NO donor inhibits proliferation through downregulation of the IGF-Akt pathway in MIAPaCa-2 cells. A, MIAPaCa-2 and MCF-7 cells were incubated with GSNO (100 and 500 μmol/L) in medium containing 10% FBS for 24, 48, and 72 h. B, MIAPaCa-2 cells were transfected with vector only (as a mock), full-length IRS-1, and IRS-1 DM2 (as a dominant negative), followed by incubation with G418. Stable transfected cells were incubated in DMEM with 10% FBS overnight and grown to 80% confluence. After incubation with SNAP (1 mmol/L) under serum starvation for 6 h, the cells were incubated with IGF-I (25 nmol/L) for 5 min and then harvested. The cell lysates were subjected to immunoblotting. All experiments were repeated three times and the same results were obtained. C, stable transfected MIAPaCa-2 cells were incubated with GSNO (200 μmol/L) in medium containing 10% FBS for 24, 48, and 72 h. D, stable transfected cells (2 × 104 per well) were incubated with GSNO (200 μmol/L) in medium with or without IGF-I (100 nmol/L; serum-free) for 48 h. *, P < 0.01, compared with control; ++, P < 0.05, compared with IGF-I only.

Close modal

Proliferation of MIAPaCa-2 cells was elevated in culture medium containing serum or IGF-I, whereas no proliferation was observed in culture medium without serum or IGF-I. Proliferation of full-length IRS-1–transfected cells was greater than that of the vector alone–transfected cells in the culture medium containing 10% FBS. On the other hand, the proliferation of IRS-1 DM2–transfected cells was attenuated compared with full-length IRS-1 and vector alone (Fig. 4C). The proliferation of full-length IRS-1–transfected cells was greater than that of vector alone–transfected cells in the culture medium containing 100 nmol/L IGF-I without 10% FBS, whereas IGF-I–stimulated proliferation of IRS-1 DM2–transfected cells was not observed (Fig. 4D). GSNO (200 μmol/L) significantly reduced the proliferation of vector alone–, full-length IRS-1–, and IRS-1 DM2–transfected cells in culture medium containing 10% FBS or IGF-I. In the culture medium containing serum or IGF-I, the reduction rate of proliferation of full-length IRS-1–transfected cells was the greatest, whereas it was the least in IRS-1 DM2–transfected cells (Fig. 4C and D). Proliferation of Panc-1 cells was not observed in the presence or absence of 1400W (100 μmol/L) when cultured without serum or IGF-I (Fig. 5A). 1400W significantly enhanced the proliferation of Panc-1 cells when cultured with 10% FBS (Fig. 5B). To further investigate the role of iNOS in IGF-I–stimulated proliferation, we evaluated the effects of the selective iNOS inhibitor 1400W in Panc-1 cells cultured with IGF-I in the absence of FBS. In the absence of 1400W, IGF-I failed to increase the cell numbers of Panc-1. The combination of IGF-I and 1400W, however, increased the number of Panc-1 cells (Fig. 5C). These results provide further evidence for the involvement of the downregulation of IGF-I signaling in NO-induced inhibition of cancer cell proliferation.

FIGURE 5.

1400W, a highly selective iNOS inhibitor, enhances the proliferation of Panc-1 cells. Panc-1 cells were incubated in serum-free medium or 10% FBS or IGF-I (100 nmol/L). The effects of 1400W on cell proliferation were evaluated. *, P < 0.01, compared with control. Data shown are the results of triplicate experiments. Bars, SEM.

FIGURE 5.

1400W, a highly selective iNOS inhibitor, enhances the proliferation of Panc-1 cells. Panc-1 cells were incubated in serum-free medium or 10% FBS or IGF-I (100 nmol/L). The effects of 1400W on cell proliferation were evaluated. *, P < 0.01, compared with control. Data shown are the results of triplicate experiments. Bars, SEM.

Close modal

Sensitivity of invasion to NO is dependent on IGF signaling

In the invasion assay, there was no difference in invasion among vector alone–, full-length IRS-1–, and IRS-1 DM2–transfected MIAPaCa-2 cells in the absence of the NO donor. The addition of 200 μmol/L GSNO remarkably reduced the invasion of vector alone– and full-length IRS-1–transfected MIAPaCa-2 cells, but did not alter the invasion of IRS-1 DM2–transfected MIAPaCa-2 cells (Fig. 6A and C). The invasion of Panc-1 cells incubated with 1400W (5 and 100 μmol/L) was significantly greater than that of Panc-1 cells incubated without 1400W (Fig. 6B and D).

FIGURE 6.

NO donor inhibits the invasion of MIAPaCa-2 cells and 1400W enhances the invasion of Panc-1 cells. Cell invasion was determined using BioCoat Matrigel Invasion Chambers. Stable transfected MIAPaCa-2 cells (A and C) or Panc-1 cells (B and D) were placed in the upper chambers. Conditioned medium of NIH 3T3 cells was used as a chemoattractant. Data shown are the results of triplicate experiments (A and B). Bars, SEM. C and D, representative staining of migrated cancer cells. *, P < 0.01, compared with control.

FIGURE 6.

NO donor inhibits the invasion of MIAPaCa-2 cells and 1400W enhances the invasion of Panc-1 cells. Cell invasion was determined using BioCoat Matrigel Invasion Chambers. Stable transfected MIAPaCa-2 cells (A and C) or Panc-1 cells (B and D) were placed in the upper chambers. Conditioned medium of NIH 3T3 cells was used as a chemoattractant. Data shown are the results of triplicate experiments (A and B). Bars, SEM. C and D, representative staining of migrated cancer cells. *, P < 0.01, compared with control.

Close modal

Here, we showed that NO donor reduced IRS-1 protein expression via proteasome-dependent degradation and inhibited insulin/IGF-I–stimulated phosphorylation of Akt/PKB and GSK-3β, but enhanced the phosphorylation of Erk-1/2 in pancreatic cancer cells. In this study, the COOH-terminal deletion mutants of IRS-1 (DM2) worked as a dominant negative, as previously reported (4). NO donor inhibited IGF-I–induced phosphorylation of Akt/PKB and GSK-3β in MIAPaCa-2 cells transfected with IRS-1 WT or vector, but not in cells transfected with IRS-1 DM2, thus revealing the importance of IRS-1 in the inhibition of insulin/IGF signal by NO. IRS-1 expression and IGF-I signaling have important roles in the proliferation and invasion of MIAPaCa-2 cells and Panc-1 cells, consistent with previous reports on other cancer cells (3, 31-33). NO donor inhibited the IGF-I signaling, proliferation, and invasion of MIAPaCa-2 cells transfected with full-length IRS-1 or vector. In contrast, the selective iNOS inhibitor upregulated IRS-1 protein expression and insulin/IGF signal, resulting in enhanced proliferation and invasion activity in Panc-1 cells. These results indicate that the expression of IRS-1 protein is regulated by endogenous NO production by iNOS as well as exogenous NO, resulting in downregulation of IGF-I signaling and inhibition of the proliferation and invasion of MIAPaCa-2 and Panc-1 cancer cells.

Furthermore, we identified the COOH terminus as the site responsible for IRS-1 protein degradation by NO, which is located in the SH2-containing molecule binding site next to the phosphotyrosine binding (PTB) domain (Fig. 2C). The observation of the ubiquitination and degradation of both IRS-1 DM1 and IRS-1 DM3 indicates the possibility that there may be at least two sites responsible for NO donor–induced ubiquitination in the IRS-1 protein.

The mechanism explaining the degradation of the IRS-1 protein was previously reported (19, 34). Yu et al. showed that activated R-Ras induces degradation of IRS-1 protein associated with suppression of estrogen action in MCF-7 cells (34). However, in contrast to H-Ras and N-Ras, both of which contain Cys118 residue in the conserved region, R-Ras not containing a Cys residue in the region is not modified by NO (35, 36). In addition, they described that R-Ras–dependent IRS-1 protein degradation was ubiquitin independent. Thus, R-Ras does not seem to contribute to NO-mediated IRS-1 degradation. We previously reported that PI3K, mammalian target of rapamycin, and c-jun NH2-terminal kinase/stress-activated protein kinase inhibitors failed to block NO-induced IRS-1 protein reduction, despite having these inhibitors attenuated insulin-induced IRS-1 protein reduction in mouse C2C12 cells (19). Therefore, further investigation will be necessary to clarify the mechanism of NO-mediated ubiquitination and degradation of IRS-1.

In contrast to NO-induced downregulation of the IRS-1-PI3K-Akt pathway, NO donor upregulates the phosphorylation of Erk-1/2 in MIAPaCa-2 cells. These results are in accordance with previous studies reporting that NO modifies H-Ras and N-Ras directly by S-nitrosylation, resulting in the activation of the signaling (35, 37). Lim et al. showed that the PI3K-Akt pathway phosphorylates and activates eNOS, resulting in the activity of H-Ras and N-Ras and tumor growth (36). In addition, other previous studies reported NO-induced cell proliferation and cell invasion (38, 39). The authors of these reports argue that NO-induced activation of Erk-1/2 leads to cell survival, in opposition to our data indicating the tumoricidal effects of NO. However, in light of this discrepancy, we hypothesize that the outcome of cancer cells treated with NO may depend on the balance of the PI3K-Akt and Ras-Erk pathways. For instance, the rate of NO-induced inhibition of proliferation was highest in MIAPaCa-2 cells overexpressing full-length IRS-1, but minimal in cells expressing IRS-1 DM2. This means that MIAPaCa-2 cells overexpressing IRS-1 may be dependent on the IRS-1-Akt pathway, whereas MIAPaCa-2 cells overexpressing IRS-1 DM2 may be independent of the pathway.

NO donor exhibited minimal inhibition in the proliferation of stable IRS-1 DM2–transfected MIAPaCa-2 cells, although NO donor failed to reduce the invasion of the cells. These results indicate that NO may downregulate the proliferation of MIAPaCa-2 cells partially through mechanisms other than the reduction of IRS-1 protein expression and phosphorylation. Namely, as previously reported, NO donor inhibits the activity of Akt/PKB through S-nitrosylation (12), indicating the possibility that it may be one of several mechanisms. Furthermore, NO induced DNA damage, and the activation of p53 and mitogen-activated protein kinase may be related to the phenomenon, as reported previously by Hofseth et al. (40).

This study shows that GSNO and SNAP provide high concentration of nitrite in the culture medium at 6 hours after administration. Furthermore, Panc-1 cells produced nitrite, a metabolite of NO, in culture medium. The level of NO in the cells is an important factor that influences the pathophysiologic activity of NO. iNOS can provide high concentrations of NO in Panc-1 cells, and NO derived from 100 to 500 μmol/L GSNO and 1 mmol/L SNAP may result in a high NO concentration in the cytosol of cancer cells. High levels of NO have been shown to exhibit cytotoxicity in various cells (41). Because the half time in which GSNO provides NO is longer than that of SNAP, we intentionally used SNAP for short-term experiments and GSNO for long-term experiments (42, 43). GSNO inhibited IRS-1 protein expression at dosages of 100 to 200 μmol/L. These dosages of GSNO also inhibited cell proliferation and invasion. These findings suggest that NO-mediated IRS-1 degradation contributes to the inhibition of cell proliferation and invasion by NO donor. GSNO reduces IRS-1 protein expression, which is upregulated by 1400W in Panc-1 cells (Fig. 3C). These results suggest that endogenous NO is sufficient to reduce IRS-1 expression; therefore, GSNO does not reduce IRS-1 expression in Panc-1 cells without 1400W.

The usefulness of cancer therapy using NO, including iNOS gene therapy and administration of NO donor, was recently confirmed in animal models (44-46). Consequently, NO therapy has been focused on and is currently undergoing clinical evaluation for cancer prevention (47). However, the molecular mechanism still remains unclear. Our data suggest that NO-induced downregulation of the insulin/IGF-IRS-1-Akt pathway may play an important role in the proliferation and invasion of pancreatic cancer cells.

No potential conflicts of interest were disclosed.

Grant Support: Grant-in-Aid for Scientific Research (grant nos. 18591517 and 20591633) from the Japanese Society for the Promotion of Science.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Bergmann
U
,
Funatomi
H
,
Yokoyama
M
,
Beger
HG
,
Korc
M
. 
Insulin-like growth factor I overexpression in human pancreatic cancer: evidence for autocrine and paracrine roles
.
Cancer Res
1995
;
55
:
2007
11
.
2
Furukawa
M
,
Raffeld
M
,
Mateo
C
, et al
. 
Increased expression of insulin-like growth factor I and/or its receptor in gastrinomas is associated with low curability, increased growth, and development of metastases
.
Clin Cancer Res
2005
;
11
:
3233
42
.
3
Kim
HJ
,
Litzenburger
BC
,
Cui
X
, et al
. 
Constitutively active type I insulin-like growth factor receptor causes transformation and xenograft growth of immortalized mammary epithelial cells and is accompanied by an epithelial-to-mesenchymal transition mediated by NF-κB and snail
.
Mol Cell Biol
2007
;
27
:
3165
75
.
4
Tanaka
S
,
Wands
JR
. 
A carboxy-terminal truncated insulin receptor substrate-1 dominant negative protein reverses the human hepatocellular carcinoma malignant phenotype
.
J Clin Invest
1996
;
98
:
2100
8
.
5
Ito
T
,
Sasaki
Y
,
Wands
JR
. 
Overexpression of human insulin receptor substrate 1 induces cellular transformation with activation of mitogen-activated protein kinases
.
Mol Cell Biol
1996
;
16
:
943
51
.
6
Chang
Q
,
Li
Y
,
White
MF
,
Fletcher
JA
,
Xiao
S
. 
Constitutive activation of insulin receptor substrate 1 is a frequent event in human tumors: therapeutic implications
.
Cancer Res
2002
;
62
:
6035
8
.
7
Asano
T
,
Yao
Y
,
Shin
S
,
McCubrey
J
,
Abbruzzese
JL
,
Reddy
SA
. 
Insulin receptor substrate is a mediator of phosphoinositide 3-kinase activation in quiescent pancreatic cancer cells
.
Cancer Res
2005
;
65
:
9164
8
.
8
Dearth
RK
,
Cui
X
,
Kim
HJ
, et al
. 
Mammary tumorigenesis and metastasis caused by overexpression of insulin receptor substrate 1 (IRS-1) or IRS-2
.
Mol Cell Biol
2006
;
26
:
9302
14
.
9
Park
HS
,
Huh
SH
,
Kim
MS
,
Lee
SH
,
Choi
EJ
. 
Nitric oxide negatively regulates c-Jun N-terminal kinase/stress-activated protein kinase by means of S-nitrosylation
.
Proc Natl Acad Sci U S A
2000
;
97
:
14382
7
.
10
Reynaert
NL
,
Ckless
K
,
Korn
SH
, et al
. 
Nitric oxide represses inhibitory κB kinase through S-nitrosylation
.
Proc Natl Acad Sci U S A
2004
;
101
:
8945
50
.
11
Mannick
JB
,
Hausladen
A
,
Liu
L
, et al
. 
Fas-induced caspase denitrosylation
.
Science
1999
;
284
:
651
4
.
12
Yasukawa
T
,
Tokunaga
E
,
Ota
H
,
Sugita
H
,
Martyn
JA
,
Kaneki
M
. 
S-nitrosylation-dependent inactivation of Akt/protein kinase B in insulin resistance
.
J Biol Chem
2005
;
280
:
7511
8
.
13
Ignarro
LJ
,
Buga
GM
,
Wood
KS
,
Byrns
RE
,
Chaudhuri
G
. 
Endothelium-derived relaxing factor produced and released from artery and vein is nitric oxide
.
Proc Natl Acad Sci U S A
1987
;
84
:
9265
9
.
14
Palmer
RM
,
Ferrige
AG
,
Moncada
S
. 
Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor
.
Nature
1987
;
327
:
524
6
.
15
Kuo
PC
,
Schroeder
RA
. 
The emerging multifaceted roles of nitric oxide
.
Ann Surg
1995
;
221
:
220
35
.
16
Wink
DA
,
Miranda
KM
,
Espey
MG
. 
Effects of oxidative and nitrosative stress in cytotoxicity
.
Semin Perinatol
2000
;
24
:
20
3
.
17
Perreault
M
,
Marette
A
. 
Targeted disruption of inducible nitric oxide synthase protects against obesity-linked insulin resistance in muscle
.
Nat Med
2001
;
7
:
1138
43
.
18
Xie
K
,
Fidler
IJ
. 
Therapy of cancer metastasis by activation of the inducible nitric oxide synthase
.
Cancer Metastasis Rev
1998
;
17
:
55
75
.
19
Sugita
H
,
Fujimoto
M
,
Yasukawa
T
, et al
. 
Inducible nitric-oxide synthase and NO donor induce insulin receptor substrate-1 degradation in skeletal muscle cells
.
J Biol Chem
2005
;
280
:
14203
11
.
20
Ambs
S
,
Merriam
WG
,
Ogunfusika
MO
, et al
. 
p53 and vascular endothelial growth factor regulate tumor growth of NOS2-expressing human carcinoma cells
.
Nat Med
1998
;
4
:
1371
6
.
21
Camp
ER
,
Yang
A
,
Liu
W
, et al
. 
Roles of nitric oxide synthase inhibition and vascular endothelial growth factor receptor-2 inhibition on vascular morphology and function in an in vivo model of pancreatic cancer
.
Clin Cancer Res
2006
;
12
:
2628
33
.
22
Kalivendi
SV
,
Kotamraju
S
,
Zhao
H
,
Joseph
J
,
Kalyanaraman
B
. 
Doxorubicin-induced apoptosis is associated with increased transcription of endothelial nitric-oxide synthase. Effect of antiapoptotic antioxidants and calcium
.
J Biol Chem
2001
;
276
:
47266
76
.
23
Wang
B
,
Wei
D
,
Crum
VE
, et al
. 
A novel model system for studying the double-edged roles of nitric oxide production in pancreatic cancer growth and metastasis
.
Oncogene
2003
;
22
:
1771
82
.
24
Peshes-Yaloz
N
,
Rosen
D
,
Sondel
PM
,
Krammer
PH
,
Berke
G
. 
Up-regulation of Fas (CD95) expression in tumour cells in vivo
.
Immunology
2007
;
120
:
502
11
.
25
Kotamraju
S
,
Williams
CL
,
Kalyanaraman
B
. 
Statin-induced breast cancer cell death: role of inducible nitric oxide and arginase-dependent pathways
.
Cancer Res
2007
;
67
:
7386
94
.
26
Notas
G
,
Nifli
AP
,
Kampa
M
,
Vercauteren
J
,
Kouroumalis
E
,
Castanas
E
. 
Resveratrol exerts its antiproliferative effect on HepG2 hepatocellular carcinoma cells, by inducing cell cycle arrest, and NOS activation
.
Biochim Biophys Acta
2006
;
1760
:
1657
66
.
27
Jarry
A
,
Charrier
L
,
Bou-Hanna
C
, et al
. 
Position in cell cycle controls the sensitivity of colon cancer cells to nitric oxide-dependent programmed cell death
.
Cancer Res
2004
;
64
:
4227
34
.
28
Chawla-Sarkar
M
,
Bauer
JA
,
Lupica
JA
, et al
. 
Suppression of NF-κB survival signaling by nitrosylcobalamin sensitizes neoplasms to the anti-tumor effects of Apo2L/TRAIL
.
J Biol Chem
2003
;
278
:
39461
9
.
29
Misko
TP
,
Schilling
RJ
,
Salvemini
D
,
Moore
WM
,
Currie
MG
. 
A fluorometric assay for the measurement of nitrite in biological samples
.
Anal Biochem
1993
;
214
:
11
6
.
30
Ceyhan
GO
,
Giese
NA
,
Erkan
M
, et al
. 
The neurotrophic factor artemin promotes pancreatic cancer invasion
.
Ann Surg
2006
;
244
:
274
81
.
31
Shi
B
,
Sepp-Lorenzino
L
,
Prisco
M
,
Linsley
P
,
deAngelis
T
,
Baserga
R
. 
Micro RNA 145 targets the insulin receptor substrate-1 and inhibits the growth of colon cancer cells
.
J Biol Chem
2007
;
282
:
32582
90
.
32
Scamuffa
N
,
Siegfried
G
,
Bontemps
Y
, et al
. 
Selective inhibition of proprotein convertases represses the metastatic potential of human colorectal tumor cells
.
J Clin Invest
2008
;
118
:
352
63
.
33
Jiang
P
,
Enomoto
A
,
Jijiwa
M
, et al
. 
An actin-binding protein Girdin regulates the motility of breast cancer cells
.
Cancer Res
2008
;
68
:
1310
8
.
34
Yu
Y
,
Hao
Y
,
Feig
LA
. 
The R-Ras GTPase mediates cross talk between estrogen and insulin signaling in breast cancer cells
.
Mol Cell Biol
2006
;
26
:
6372
80
.
35
Lander
HM
,
Hajjar
DP
,
Hempstead
BL
, et al
. 
A molecular redox switch on p21(ras). Structural basis for the nitric oxide-p21(ras) interaction
.
J Biol Chem
1997
;
272
:
4323
6
.
36
Lim
KH
,
Ancrile
BB
,
Kashatus
DF
,
Counter
CM
. 
Tumour maintenance is mediated by eNOS
.
Nature
2008
;
452
:
646
9
.
37
Ibiza
S
,
Perez-Rodriguez
A
,
Ortega
A
, et al
. 
Endothelial nitric oxide synthase regulates N-Ras activation on the Golgi complex of antigen-stimulated T cells
.
Proc Natl Acad Sci U S A
2008
;
105
:
10507
12
.
38
Ellerhorst
JA
,
Ekmekcioglu
S
,
Johnson
MK
,
Cooke
CP
,
Johnson
MM
,
Grimm
EA
. 
Regulation of iNOS by the p44/42 mitogen-activated protein kinase pathway in human melanoma
.
Oncogene
2006
;
25
:
3956
62
.
39
Donnini
S
,
Finetti
F
,
Solito
R
, et al
. 
EP2 prostanoid receptor promotes squamous cell carcinoma growth through epidermal growth factor receptor transactivation and iNOS and ERK1/2 pathways
.
FASEB J
2007
;
21
:
2418
30
.
40
Hofseth
LJ
,
Saito
S
,
Hussain
SP
, et al
. 
Nitric oxide-induced cellular stress and p53 activation in chronic inflammation
.
Proc Natl Acad Sci U S A
2003
;
100
:
143
8
.
41
Heller
R
,
Polack
T
,
Grabner
R
,
Till
U
. 
Nitric oxide inhibits proliferation of human endothelial cells via a mechanism independent of cGMP
.
Atherosclerosis
1999
;
144
:
49
57
.
42
Mancuso
C
,
Bonsignore
A
,
Di Stasio
E
,
Mordente
A
,
Motterlini
R
. 
Bilirubin and S-nitrosothiols interaction: evidence for a possible role of bilirubin as a scavenger of nitric oxide
.
Biochem Pharmacol
2003
;
66
:
2355
63
.
43
Janssens
MY
,
Verovski
VN
,
Van den Berge
DL
,
Monsaert
C
,
Storme
GA
. 
Radiosensitization of hypoxic tumour cells by S-nitroso-N-acetylpenicillamine implicates a bioreductive mechanism of nitric oxide generation
.
Br J Cancer
1999
;
79
:
1085
9
.
44
Adams
C
,
McCarthy
HO
,
Coulter
JA
, et al
. 
Nitric oxide synthase gene therapy enhances the toxicity of cisplatin in cancer cells
.
J Gene Med
2009
;
11
:
160
8
.
45
Kiziltepe
T
,
Hideshima
T
,
Ishitsuka
K
, et al
. 
JS-K, a GST-activated nitric oxide generator, induces DNA double-strand breaks, activates DNA damage response pathways, and induces apoptosis in vitro and in vivo in human multiple myeloma cells
.
Blood
2007
;
110
:
709
18
.
46
Wang
Z
,
Cook
T
,
Alber
S
, et al
. 
Adenoviral gene transfer of the human inducible nitric oxide synthase gene enhances the radiation response of human colorectal cancer associated with alterations in tumor vascularity
.
Cancer Res
2004
;
64
:
1386
95
.
47
Ma
Q
,
Wang
Y
,
Gao
X
,
Ma
Z
,
Song
Z
. 
l-Arginine reduces cell proliferation and ornithine decarboxylase activity in patients with colorectal adenoma and adenocarcinoma
.
Clin Cancer Res
2007
;
13
:
7407
12
.