The human inducible nitric oxide synthase (hiNOS) gene is regulated by nuclear factor κB (NF-κB) and has recently been shown to be a target of the Wnt/β-catenin pathway. In this study, we tested the hypothesis that Wnt/β-catenin signaling might regulate cytokine- or tumor necrosis factor α (TNFα)–induced hiNOS expression through interaction with NF-κB. A cytokine mixture of TNFα + interleukin (IL)-1β + IFNγ induced a 2- to 3-fold increase in hiNOS promoter activity in HCT116 and DLD1 colon cells, but produced a 2-fold decrease in SW480 colon cancer cells. A similar differential activity was seen in liver cancer cells (HepG2, Huh7, and Hep3B). Overexpression of β-catenin produced a dose-dependent decrease in NF-κB reporter activity and decreased cytokine mixture–induced hiNOS promoter activity. Gel shift for TNFα-induced hiNOS NF-κB activation showed decreased p50 binding and decreased NF-κB reporter activity in the β-catenin–mutant HAβ18 cells. Conversely, enhanced p50 binding and increased NF-κB reporter activity were seen in HAβ85 cells, which lack β-catenin signaling. Coimmunoprecipitation confirmed that β-catenin complexed with both p65 and p50 NF-κB proteins. NF-κB–dependent Traf1 protein expression also inversely correlated with the level of β-catenin. Furthermore, SW480 cells stably transformed with wild-type adenomatous polyposis coli showed decreased β-catenin protein and increased TNFα-induced p65 NF-κB binding as well as iNOS and Traf1 expression. Finally, β-catenin inversely correlated with iNOS and Fas expression in vivo in hepatocellular carcinoma tumor samples. Our in vitro and in vivo data show that β-catenin signaling inversely correlates with cytokine-induced hiNOS and other NF-κB–dependent gene expression. These findings underscore the complex role of Wnt/β-catenin, NF-κB, and iNOS signaling in the pathophysiology of inflammation-associated carcinogenesis. [Cancer Res 2009;69(9):3764–71]

The Wnt/β-catenin signaling pathway influences many cellular processes including cell adhesion, growth, and differentiation (1). Several lines of evidence have implicated aberrant Wnt/β-catenin signaling in the development of human cancers, especially colon cancer and hepatocellular carcinoma (2, 3). In colon cancer, mutation of the tumor suppressor gene adenomatous polyposis coli (APC) leads to stabilization of transcriptional regulator β-catenin, which accumulates in the cytoplasm, enters the nucleus, and interacts with nuclear T-cell factor 4 (Tcf-4)/lymphoid enhancer factor (LEF) to activate transcription of specific target genes leading to cell proliferation and carcinogenesis (46).

Nitric oxide (NO) is a diverse biological mediator with important roles in vascular biology, inflammation, and tumor biology (79). Studies have shown that NO promotes carcinogenesis at low levels and that NO inhibits tumor growth at high concentrations (1012). NO has been shown to cause DNA damage and is mutagenic (13). Inducible nitric oxide synthase (iNOS) has been documented in cancer specimens (14) and precancerous chronic inflammatory conditions (15, 16). Several studies have also investigated the relationship between iNOS expression, vascular endothelial growth factor, and the tumor suppressor gene product p53 (17, 18). Exposure of human cancer cells to a NO donor resulted in p53 protein accumulation, which then acted in a negative feedback loop to trans-repress human iNOS (hiNOS) gene transcription in vitro and in vivo (19, 20). Hence, the double-edged sword of NO in tumor biology clearly depends on cell type, NO concentration, oxidative stress, and tumor milieu (21, 22).

The expression of iNOS is predominantly regulated at the transcriptional level by inflammatory cytokines that signal through nuclear factor κB (NF-κB) and other nuclear factors (2327). NF-κB is also an important transcriptional factor that is involved in colorectal carcinogenesis (28, 29). Recently, we have shown that the hiNOS gene is up-regulated by Wnt/β-catenin signaling (30). Overexpression of β-catenin and Tcf-4 increased basal and cytokine-induced iNOS promoter activity in cancer cells that was dependent on intact cis-acting Tcf-4 binding element (TBE) motifs in the hiNOS promoter (30). However, another study has shown that β-catenin physically interacts with NF-κB and inhibits its activation in human colon and breast cancer (31). Therefore, we examined the effect of β-catenin signaling on NF-κB activation and tumor necrosis factor α (TNFα)–induced iNOS expression in human colon, liver, and lung cancer cells. Our findings indicate that β-catenin differentially regulates TNFα-induced hiNOS and other NF-κB target gene expression by physical interaction with NF-κB in colon and liver cancer cells.

Cell lines and reagents. The human colorectal cancer cell lines SW480, DLD1, RKO, and 293 were obtained from American Type Culture Collection; HCT116, HAβ18, and HAβ85 (gene-targeted HCT116 derivatives) were kindly given by Dr. Todd Waldman (Georgetown University, Washington, DC; ref. 32). They were cultured at 37°C in 5% CO2 in medium containing 10% fetal bovine serum (FBS; Clontech), 100 units/mL penicillin, 100 μg/mL streptomycin, and 15 mmol/L HEPES. SW480, DLD1, RKO, and 293 cells were cultured in DMEM (Invitrogen), and HCT116, HAβ18, and HAβ85 in McCoy's 5A medium (Invitrogen). Unless indicated, cells were stimulated with a cytokine mixture consisting of 1,000 units/mL human TNF-α (R&D Systems), 100 units/mL IL-1β (R&D Systems), and 250 units/mL human IFNγ (R&D Systems or Roche Pharmaceuticals).

Plasmid constructs. The hiNOS −5.8 kb NF-κB-luciferase reporter plasmid (2× NF-κB; ref. 25), hiNOS −5.8 kb NF-κB oligonucleotide cloned into the unique Sma1 site of luciferase reporter plasmid pT109, which contains 109 bp of the herpes virus thymidine kinase promoter, driving expression of firefly luciferase, was confirmed by sequencing. NF-κB −5.8 wild-type (WT), 5′-AGAGGGCTTTCCCAGAACCA-3′; NF-κB −5.8 mutant, 5′-AGAGGGCTCGCCCAGAACCA-3′ (25). The reporter plasmids pTOP-FLASH and pFOP-FLASH, WT APC expression vector (PCMV-neo-APC), and β-catenin expression vector (PCI-neo β-catenin) were kindly provided by Dr. Bert Vogelstein. Expression plasmids encoding human p50 and FLAG-taggep65 (pFL65) NF-κB proteins were kindly provided by Dr. Joseph DiDonato (Department of Cancer Biology, Cleveland Clinic Foundation, Cleveland, OH; ref. 33).

Transient transfection assay. DNA transfections of cells were carried out in six-well plates (Corning) by using Lipofectamine (Invitrogen) and MIRUS Trans-IT reagent (Mirus) as previously described (30). To control for transfection efficiency between groups, 0.5 μg of a plasmid containing a cytomegalovirus promoter–driven β-galactosidase was added to each well. pRSV-Luc was used as positive control, and the promoterless plasmid pXP2 served as negative control. RKO β-catenin S33Y stable transfectants were established by transfection with β-catenin S33Y plasmid (2). Stable transfectants in SW480 cells were established by transfection with full-length APC plasmid (2).

Immunoprecipitation and Western blot analysis. Immunoprecipitation, SDS-PAGE, and Western blot were done as described previously (30). Antibodies used for Western blot were rabbit anti-hiNOS polyclonal (BD Biosciences), β-catenin (H-102), p65 (A), p50 (c-19), TRAF1 (H-132, Santa Cruz), and mouse anti–β-catenin monoclonal (Sigma).

Electrophoretic mobility shift assays. Nuclear protein extraction and gel shift assay were done as previously described (30). Antibody supershifts used 0.5 to 2 μg of anti-p65 (A)X (Santa Cruz) and anti-p50 (NLS)X (Santa Cruz).

Immunofluorescent staining. Cells were cultured on coverslips and washed twice with cold PBS, fixed with 2% paraformaldehyde in PBS for 15 min, permeabilized with 0.1% Triton X-100 and 10% FBS in PBS for 30 min at room temperature, and then incubated with the specific primary antibodies for p50 (Santa Cruz), β-catenin (Sigma), APC (BD), iNOS(BD), and cleaved PARP (Asp214, Cell Signaling; ref. 30). Slides were viewed with Olympus Provis and Leica TSL-SL microscopes.

Immunohistochemistry. Tissue samples were deparaffinized and subjected to alcohol gradient and washed thrice, then treated with 0.3% H2O2 in methanol followed by 10% horse serum for 30 min. Sections were incubated overnight with primary antibodies β-catenin (Sigma; 1:200 dilution), Fas (CH11 Upstate; 1:150 dilution), or iNOS (Abcam; 1:150 dilution) and then with secondary antibodies Bio-antimouse and rabbit IgG (1:200 dilution) for 30 min at room temperature; signal was detected using ABC Elite Kit (Vector Laboratories).

NO production assessment. Culture supernatants were collected and assayed for nitrite, a stable end product of NO oxidation, using the Griess reaction as described (34).

Statistical methods. Data are presented as the mean ± SE. Cultures were done in duplicate or triplicate, and experiments were done at least thrice. Data were analyzed by the Student t test or ANOVA, and P < 0.05 was considered statistically significant.

β-Catenin inversely correlated with cytokine-induced iNOS expression. To examine the level of cytokine-induced iNOS gene expression in HCT116, DLD1, and SW480 cell lines, we transfected the cells with the WT hiNOS −7.2 kb luciferase promoter plasmid and then treated them with a cytokine mixture of TNFα + IL-1β + IFNγ for 6 hours. The cytokine mixture induced hiNOS promoter activity in HCT116 and DLD1 cells, but significantly decreased hiNOS promoter activity below basal levels in SW480 cells (Fig. 1A). This was an unexpected finding and led us to investigate in other human cell lines. A similar differential expression of cytokine-induced hiNOS transcription was seen in HepG2 cells compared with Huh7 and Hep3B liver cancer cells, where again cytokine mixture stimulation decreased hiNOS promoter activity below basal levels (Fig. 1B). We also observed that the magnitude of cytokine-induced NO production was quite different, being dramatically increased in primary human and rat hepatocytes, lower in HepG2 and DLD1 cells, and no appreciable NO synthesis detectable by nitrate assay in SW480 cells (Fig. 1C). Interestingly, endogenous β-catenin expression was high in the SW480 cells and lower in the HCT116 and DLD1 cells (Supplementary Fig. S1A). These observations led us to hypothesize that the presence of β-catenin might inversely correlate and regulate cytokine-induced iNOS expression, possibly through interaction with the NF-κB pathway.

Figure 1.

Differential induction of iNOS by cytokines in various cancer cell lines. Cytokine-induced hiNOS expression in colon (A) and liver (B) cancer cells. Cells were transiently transfected with 1 μg WT −7.2 kb hiNOS promoter luciferase reporter plasmid, and luciferase activity was determined 6 h after treatment with a cytokine mixture (CM) of TNFα, IL-1β, and IFNγ. Transfection efficiency is normalized by cotransfection with β-galactosidase. RLU, relative luciferase unit. *, P < 0.01, compared with −7.2 kb hiNOS promoter stimulated without cytokine mixture. C, human and rat hepatocytes, liver cancer cell line (HepG2), and colon cancer cell lines (DLD1 and SW480) were treated with cytokines for 24 h and the media were harvested for nitrite assay.

Figure 1.

Differential induction of iNOS by cytokines in various cancer cell lines. Cytokine-induced hiNOS expression in colon (A) and liver (B) cancer cells. Cells were transiently transfected with 1 μg WT −7.2 kb hiNOS promoter luciferase reporter plasmid, and luciferase activity was determined 6 h after treatment with a cytokine mixture (CM) of TNFα, IL-1β, and IFNγ. Transfection efficiency is normalized by cotransfection with β-galactosidase. RLU, relative luciferase unit. *, P < 0.01, compared with −7.2 kb hiNOS promoter stimulated without cytokine mixture. C, human and rat hepatocytes, liver cancer cell line (HepG2), and colon cancer cell lines (DLD1 and SW480) were treated with cytokines for 24 h and the media were harvested for nitrite assay.

Close modal

β-Catenin reduced TNFα-induced hiNOS-specific NF-κB reporter and binding activities. To determine whether β-catenin interacts with NF-κB and regulates hiNOS expression, we synthesized hiNOS-specific −5.8 kb NF-κB oligonucleotides and constructed two tandem copies of the NF-κB element at −5.8 kb in the hiNOS promoter for use in gel shift assay, as well as hiNOS-specific NF-κB reporter plasmid driving the minimal herpes thymidine kinase-luciferase promoter. We initially determined the effects of β-catenin overexpression on NF-κB p65–mediated activation of the hiNOS NF-κB reporter. A p65 expression plasmid (0.2 μg) was cotransfected with increasing doses of β-catenin expression plasmid (mutant β-cateninS33Y) along with the hiNOS NF-κBx2 reporter. β-Catenin overexpression inhibited the p65 activation of hiNOS NF-κB reporter in a dose-dependent manner (Fig. 2A). β-Catenin overexpression also abrogated the cytokine mixture–induced hiNOS promoter activity in the HCT116, DLD1, and HepG2 cells previously noted and actually produced a 30% to 50% drop in luciferase activity compared with basal cells (Fig. 2B). These findings suggest that β-catenin can antagonize cytokine-mediated induction of the NF-κB–dependent hiNOS gene.

Figure 2.

Effects of β-catenin on hiNOS promoter activity. A, 293 cells were cotransfected with 2xNF-κB luc (−5.8 kb in hiNOS promoter; 0.1 μg), p65 (0.2 μg), and active mutant β-catenin S33Y (0.6, 1.2, and 1.8 μg). Luciferase activities were determined 36 h after cotransfection. *, P < 0.01, compared with 2xNF-κB luciferase reporter empty vector; **, P < 0.05, compared with 2xNF-κB luciferase reporter cotransfected with β-catenin expression empty vector. B, cancer cells were cotransfected with WT −7.2 kb hiNOS promoter-luciferase (0.5 μg) and active mutant β-catenin S33Y (1.5 μg) for 24 h, and then luciferase activities determined 6 h after treatment with or without cytokine mixture of TNFα, IL-1β, and IFNγ. Total amounts of plasmid DNA were kept constant by adding the empty pcDNA vector. *, P < 0.01, compared with −7.2 kb hiNOS promoter luciferase reporter cotransfected with β-catenin expression empty vector.

Figure 2.

Effects of β-catenin on hiNOS promoter activity. A, 293 cells were cotransfected with 2xNF-κB luc (−5.8 kb in hiNOS promoter; 0.1 μg), p65 (0.2 μg), and active mutant β-catenin S33Y (0.6, 1.2, and 1.8 μg). Luciferase activities were determined 36 h after cotransfection. *, P < 0.01, compared with 2xNF-κB luciferase reporter empty vector; **, P < 0.05, compared with 2xNF-κB luciferase reporter cotransfected with β-catenin expression empty vector. B, cancer cells were cotransfected with WT −7.2 kb hiNOS promoter-luciferase (0.5 μg) and active mutant β-catenin S33Y (1.5 μg) for 24 h, and then luciferase activities determined 6 h after treatment with or without cytokine mixture of TNFα, IL-1β, and IFNγ. Total amounts of plasmid DNA were kept constant by adding the empty pcDNA vector. *, P < 0.01, compared with −7.2 kb hiNOS promoter luciferase reporter cotransfected with β-catenin expression empty vector.

Close modal

Next, we examined for β-catenin/Tcf-4 transcriptional activity in HCT116 cells (β-catenin WT/mut) compared with HAβ18 (del/mut) and HAβ85 (WT/del) cell lines (32) by pTOP/pFOP transfection. The HAβ85 (WT/del) cells lack β-catenin signaling due to deletion of the mutant β-catenin allele. In these studies, the WT β-catenin is subject to degradation, whereas the mutant β-catenin is stabilized and active for nuclear translocation. The pTOP expression plasmid is a positive reporter plasmid for β-catenin transcriptional activity (containing three copies of an optimal Tcf binding motif), whereas the pFOP is a negative control mutant expression reporter. As predicted, the presence of a mutant β-catenin allele increased β-catenin/Tcf-4 transcriptional activity (pTOP reporter activity) in HCT116 and HAβ18 cell lines, but not in HAβ85 cells (Fig. 3A). As a negative control, the pFOP reporter was used and did not show any β-catenin activation. TNFα stimulation of the HAβ18 cells reduced resting hiNOS-specific NF-κB reporter activity below basal level, whereas it induced NF-κB reporter activity in the HAβ85 cells that lacked β-catenin signaling (Fig. 3B). Dominance of the mutant β-catenin in the HAβ18 cells also suppressed TNFα-induced p50 DNA binding (Fig. 3C,, lane 4, lower band). Immunofluorescent staining also indicated that activated β-catenin decreased TNFα-induced NF-κB p50 translocation to the nucleus of HAβ18 cells, compared with abundant p50 staining in the HAβ85 cells (Fig. 3D), which is consistent with the β-catenin–mediated repression of NF-κB reporter activity and gel shift binding (Fig. 3B and C).

Figure 3.

β-Catenin decreased TNF-induced hiNOS-specific NF-κB reporter and binding activities. A, β-catenin transcriptional activity in CTNNB1 knockout cells was evaluated by transfecting p-TOP/p-FOP (1 μg) for 24 h in HCT116 (β-catenin WT/mut Δ45), HAβ18 (β-catenin −/mut Δ45), and HAβ85 (β-catenin WT/−) colon cancer cells. B, colon cancer cell lines were transfected with NF-κB-luc reporter (1.0 μg), and then luciferase activities determined 9 h after treatment with or without TNFα (1 ng/mL). *, P < 0.01, compared with 2xNF-κB luciferase reporter stimulated without TNFα. C, gel shift assay for hiNOS-specific NF-κB DNA binding activity. Cells were treated with TNFα (10 ng/mL) for 15 min. D, colon cancer cell lines HAβ18 (β-catenin −/mut Δ45) and HAβ85 (β-catenin WT/−) were treated with TNFα (10 ng/mL) for 15 min. p50 was stained with FITC (green), and F-actin was stained by rhodamine-conjugated phalloidin (red; ×400).

Figure 3.

β-Catenin decreased TNF-induced hiNOS-specific NF-κB reporter and binding activities. A, β-catenin transcriptional activity in CTNNB1 knockout cells was evaluated by transfecting p-TOP/p-FOP (1 μg) for 24 h in HCT116 (β-catenin WT/mut Δ45), HAβ18 (β-catenin −/mut Δ45), and HAβ85 (β-catenin WT/−) colon cancer cells. B, colon cancer cell lines were transfected with NF-κB-luc reporter (1.0 μg), and then luciferase activities determined 9 h after treatment with or without TNFα (1 ng/mL). *, P < 0.01, compared with 2xNF-κB luciferase reporter stimulated without TNFα. C, gel shift assay for hiNOS-specific NF-κB DNA binding activity. Cells were treated with TNFα (10 ng/mL) for 15 min. D, colon cancer cell lines HAβ18 (β-catenin −/mut Δ45) and HAβ85 (β-catenin WT/−) were treated with TNFα (10 ng/mL) for 15 min. p50 was stained with FITC (green), and F-actin was stained by rhodamine-conjugated phalloidin (red; ×400).

Close modal

Inactivation of β-catenin by integrating full-length APC in SW480 restored NF-κB activity. To support our hypothesis that high level of β-catenin was inhibiting NF-κB–mediated gene activation, we generated stably transformed SW480 cells overexpressing the APC gene. Because the SW480 cells contain a truncated APC gene and exhibited very high constitutive β-catenin levels (Supplementary Fig. S1A), we reasoned that adding back full-length APC could bind and degrade β-catenin in the multiprotein complex. If so, then NF-κB–mediated gene transcription should be restored in SW480-APC cells compared with control SW480-vector cells. Immunoblot of stably transformed SW480-APC cells showed strong APC, decreased β-catenin, and increased hiNOS protein compared with the SW480 cells expressing empty vector (Fig. 4A,, top). In addition, overexpression of APC led to decreased β-catenin transcriptional activity with pTOP transfection (Fig. 4A,, bottom). This was also consistent with the immunofluorescent staining, with SW480-APC exhibiting decreased β-catenin and increased iNOS staining (Fig. 4B). Likewise, SW480-APC showed a higher level of TNFα-induced p65 binding on gel shift (Fig. 4C), as well as increased TNFα-induced hiNOS mRNA and protein expressions (Fig. 4D). In general, iNOS protein exists as a homodimer. However, a doublet type of iNOS protein band is often exhibited in published Western blots, and some authors have suggested that the lower band may reflect monomers or iNOS degradation products (35).

Figure 4.

Inactivation of β-catenin by integrating full-length APC (WT) into SW480 cells restored NF-κB activity. A, basal levels of protein expression of APC, β-catenin, and hiNOS in SW480-APC and SW480-vector cells were analyzed by immunoblot. SW480-vector and SW480-APC cells were transfected with pTOP/pFOP plasmids (1 μg) for 36 h. β-Catenin/Tcf-4–regulated transcription was evaluated in SW480-APC. *, P < 0.01, compared with SW480 vector cells. B, APC overexpression (red) up-regulated expression of NF-κB target gene iNOS (green) and down-regulated expression of β-catenin (red). C, gel shift assay for hiNOS-specific NF-κB DNA binding activity. TNFα increased NF-κB (−5.8 kb hiNOS promoter) p65 DNA binding (lane 2) in SW480-APC. D, TNFα induced iNOS expression in SW480-APC cells. The cells were treated with TNFα (10 ng/mL) for 0, 0.5, 2, 3, and 4 h. Human hepatocytes (HC) were also treated with cytokines for 8 h and used as positive control. iNOS protein and mRNA expressions were analyzed by immunoblot and reverse transcription-PCR (RT-PCR).

Figure 4.

Inactivation of β-catenin by integrating full-length APC (WT) into SW480 cells restored NF-κB activity. A, basal levels of protein expression of APC, β-catenin, and hiNOS in SW480-APC and SW480-vector cells were analyzed by immunoblot. SW480-vector and SW480-APC cells were transfected with pTOP/pFOP plasmids (1 μg) for 36 h. β-Catenin/Tcf-4–regulated transcription was evaluated in SW480-APC. *, P < 0.01, compared with SW480 vector cells. B, APC overexpression (red) up-regulated expression of NF-κB target gene iNOS (green) and down-regulated expression of β-catenin (red). C, gel shift assay for hiNOS-specific NF-κB DNA binding activity. TNFα increased NF-κB (−5.8 kb hiNOS promoter) p65 DNA binding (lane 2) in SW480-APC. D, TNFα induced iNOS expression in SW480-APC cells. The cells were treated with TNFα (10 ng/mL) for 0, 0.5, 2, 3, and 4 h. Human hepatocytes (HC) were also treated with cytokines for 8 h and used as positive control. iNOS protein and mRNA expressions were analyzed by immunoblot and reverse transcription-PCR (RT-PCR).

Close modal

β-Catenin physically interacts with NF-κB subunits in colon cancer cells. To document that β-catenin was physically binding to NF-κB proteins, we performed coimmunoprecipitation experiments. The SW480 cells have strong β-catenin expression, DLD1 and HCT116 have moderate β-catenin expression, whereas the RKO cells show nearly undetectable β-catenin protein levels (Supplementary Fig. S1A). On coimmunoprecipitation, β-catenin complexed with both p50 and p65 (Supplementary Fig. S1B). This confirms a direct interaction of β-catenin with NF-κB subunits in these cells, as has been shown in other cells (31), and provides a potential regulatory mechanism for modulating NF-κB signaling. We also performed immunoprecipitation of cell lysates from primary human hepatocytes and HepG2 cells and found that β-catenin complexed with either p65 or p50 (data not shown).

Effects of β-catenin on NF-κB–mediated transcription in other target genes. To show that the inhibitory effects of β-catenin on NF-κB activity were not limited to hiNOS, we also examined Traf1 gene expression, which is known to be regulated by NF-κB activity (31). Strong TNFα-induced Traf1 protein was seen in the HAβ85 cells that lack significant β-catenin transcriptional activity, whereas the HAβ18 cells did not show inducible Traf1 expression (Fig. 5A). Similarly, the RKO cancer cell line, which lacks β-catenin protein (Supplementary Fig. S1A), also showed strong TNFα-mediated Traf1 expression (Fig. 5B). In agreement with the notion of β-catenin–mediated inhibition of NF-κB activation, we also generated RKO cells stably transformed to overexpress β-catenin and observed a significant down-regulation of TNFα-induced Traf1 protein expression (Fig. 5C). Finally, consistent with β-catenin degradation by APC binding, inducible Traf1 expression was shown in the SW480-APC cells, but not in the control SW480-vector cells that exhibit high β-catenin (Fig. 5D).

Figure 5.

High-level β-catenin decreased TNF-induced NF-κB target gene expression. HAβ18 and HAβ85 cells (A) as well as RKO and HCT116 cells (B) were treated with TNFα (10 ng/mL) for 3 h. Traf1 or iNOS protein expression was analyzed by immunoblot. C, RKO cells with or without β-catenin stable expression were treated with TNFα (10 ng/mL) for 3 h, and Traf1 protein expression was analyzed. D, SW480-APC and SW480-vector cells were treated with TNFα (10 ng/mL) for 3 h, and Traf1 expression was analyzed.

Figure 5.

High-level β-catenin decreased TNF-induced NF-κB target gene expression. HAβ18 and HAβ85 cells (A) as well as RKO and HCT116 cells (B) were treated with TNFα (10 ng/mL) for 3 h. Traf1 or iNOS protein expression was analyzed by immunoblot. C, RKO cells with or without β-catenin stable expression were treated with TNFα (10 ng/mL) for 3 h, and Traf1 protein expression was analyzed. D, SW480-APC and SW480-vector cells were treated with TNFα (10 ng/mL) for 3 h, and Traf1 expression was analyzed.

Close modal

β-Catenin inhibited NO-induced apoptosis in HAβ18 cells. To test the relationship between β-catenin and NO-induced apoptosis, HAβ18 and HAβ85 cells were treated with the NO donor S-nitroso-N-acetylpenicillamine. The results showed that NO induced activation of caspase-3 (Fig. 6A) and PARP (Fig. 6B) in the HAβ85 cells, but not in the HAβ18 cells, and suggests that the presence of mutated β-catenin in the HAβ18 cells inhibits NO-induced apoptosis. A recent study also reported similar findings in stably transformed RKO-β-catenin cells (36).

Figure 6.

Effect of β-catenin on NO-induced apoptosis and iNOS/Fas expression in human hepatocellular carcinoma tissues. A, HAβ18 and HAβ85 cells were treated with NO donor S-nitroso-N-acetylpenicillamine (SNAP; 0 or 500 μmol/L) for 14 h, and lysates were analyzed for cleaved caspase-3 protein expression. B, HAβ18 and HAβ85 cells were treated with NO donor S-nitroso-N-acetylpenicillamine (500 μmol/L) for 18 h as indicated and labeled with cleaved PARP (green). Actin filaments have been labeled with phalloidin (red). DRAQ5 (blue) fluorescent DNA dye is used as nuclear stain (Leica TSL-SL confocal microscope 63×; zoom, 200). C, representative human primary hepatocellular carcinoma tissue samples were analyzed by immunohistochemistry. In case 1, when β-catenin was inactive (predominantly in the membrane; a), iNOS (b) and Fas (c) were strongly expressed; in case 2, when β-catenin was active (in the cytoplasm and nucleus; d), iNOS (e) and Fas (f) expression levels were much lower.

Figure 6.

Effect of β-catenin on NO-induced apoptosis and iNOS/Fas expression in human hepatocellular carcinoma tissues. A, HAβ18 and HAβ85 cells were treated with NO donor S-nitroso-N-acetylpenicillamine (SNAP; 0 or 500 μmol/L) for 14 h, and lysates were analyzed for cleaved caspase-3 protein expression. B, HAβ18 and HAβ85 cells were treated with NO donor S-nitroso-N-acetylpenicillamine (500 μmol/L) for 18 h as indicated and labeled with cleaved PARP (green). Actin filaments have been labeled with phalloidin (red). DRAQ5 (blue) fluorescent DNA dye is used as nuclear stain (Leica TSL-SL confocal microscope 63×; zoom, 200). C, representative human primary hepatocellular carcinoma tissue samples were analyzed by immunohistochemistry. In case 1, when β-catenin was inactive (predominantly in the membrane; a), iNOS (b) and Fas (c) were strongly expressed; in case 2, when β-catenin was active (in the cytoplasm and nucleus; d), iNOS (e) and Fas (f) expression levels were much lower.

Close modal

Lithium chloride stabilizes β-catenin and inhibits TNFα-mediated NF-κB activation. As another approach to support our hypothesis, we used lithium chloride (LiCl), a well-known inhibitor of glycogen synthase kinase 3β (GSK-3β), to block β-catenin phosphorylation and degradation. LiCl was added to the carcinoid cell line NCI H727 and dramatically induced β-catenin expression (Supplementary Fig. S2A). LiCl also decreased TNFα-induced gel shift for NF-κB (Supplementary Fig. S2B, lane 4). Finally, LiCl also inhibited TNFα-stimulated p65 staining in cultures of a freshly isolated carcinoid metastasis to the liver from a pancreatic carcinoid primary tumor (Supplementary Fig. S2C).

β-Catenin inversely correlated with iNOS or Fas expression in human liver tumors. Both iNOS and Fas are target genes of the NF-κB pathway, which are also regulated by β-catenin signaling in cancer cells. To determine if there was also an inverse correlation between β-catenin and iNOS or Fas expression in vivo, we stained 25 samples of hepatocellular carcinoma or metastatic colorectal cancer to liver by immunohistochemistry. We observed that β-catenin inversely correlated with iNOS or Fas expression in these tumor tissue samples (Fig. 6C; Supplementary Table S1). Cases #1 and #2 are primary hepatocellular carcinoma tumor samples with opposite findings: In case #1, β-catenin levels are low and the tumor shows strong staining for both iNOS and Fas, whereas in case #2, high β-catenin levels inversely result in low iNOS and Fas expression (Fig. 6C). In 10 hepatocellular carcinoma tumors that were resected, all of the β-catenin–positive tumors were negative for iNOS or Fas staining, whereas the β-catenin–negative tumors stained for both Fas and iNOS (Supplementary Table S1). For the resected hepatic colorectal cancer metastases, 9 of 11 Fas-negative tumors were β-catenin positive whereas all of the Fas-positive tumors were negative for β-catenin staining. Similar findings were observed for iNOS staining.

Previously we identified that the hiNOS (NOS2) gene was regulated by Wnt/β-catenin signaling and identified two TBEs in the hiNOS promoter that accounted for a direct induction of basal and cytokine-induced hiNOS promoter activity dependent on intact TBE (30). However, in the course of those studies, we observed several different human liver and colon cancer cells that actually exhibited decreased iNOS gene expression after cytokine stimulation, which markedly contrasted with primary human and rodent hepatocytes, as well as the findings in several other human cancer cell lines. Because the level of activated β-catenin varies among different cancers (1, 2) and because NF-κB has been shown to have an important role in cytokine-mediated hiNOS transcription (2426) as well as inflammation-associated cancer (37, 38), we hypothesized that an inverse effect might exist between β-catenin signaling and NF-κB–dependent hiNOS gene transcription. This study presents major and novel findings: (a) β-catenin signaling in tumor cell lines inversely correlated with NF-κB–dependent hiNOS gene expression; (b) overexpression of β-catenin inhibited cytokine-induced hiNOS transcription to below basal levels mediated by β-catenin interaction with NF-κB p50 and p65 proteins; (c) high levels of endogenous β-catenin in tumor cells repressed TNFα-induced NF-κB activation that was reversed by APC overexpression or enhanced by GSK-3β inhibition with LiCl; (d) the inverse relationship of β-catenin and NF-κB–dependent gene expression was not confined to hiNOS and was also seen with other NF-κB–dependent genes such as Traf1 and Fas and; (e) the inverse correlation of β-catenin levels and NF-κB activation was also seen in vivo in primary hepatocellular carcinoma, colorectal cancer metastases, and neuroendocrine cancer.

NF-κB plays a key role in inflammation and carcinogenesis (28, 29). NF-κB is also a central transcription factor mediating TNFα- and IL-1β–induced iNOS gene expression in rat and human hepatocytes (25, 26, 33, 39, 40). Studies indicate that higher levels of iNOS and NO contribute to apoptosis through cytotoxicity (41), whereas lower NO levels may be involved in carcinogenesis (42). NF-κB activation is up-regulated by inflammatory cytokines and growth factors although it is constitutively activated in many cancer cells (43).

Although our previous work showed that the hiNOS gene was directly induced by Wnt/β-catenin signaling (30), we did not examine the relationship between differential β-catenin levels and NF-κB–induced hiNOS expression after cytokine stimulation. Our findings in the current study showed that β-catenin expression indeed reduced cytokine-induced and p65-driven hiNOS-specific NF-κB reporter activity (Fig. 2). Moreover, β-catenin overexpression abolished cytokine-induced hiNOS promoter activity in the HCT116, DLD1, and HepG2 cells. Hence, high levels of β-catenin antagonized cytokine-mediated induction of the NF-κB–dependent hiNOS gene.

By using a pair of cell lines derived from the HCT116 human colon cancer cells, we found that stabilization of β-catenin by mutation in the HAβ18 cells (CTNNB1 −/Δ45) decreased TNFα-induced p50 nuclear translocation, NF-κB p50 DNA binding, and hiNOS-specific NF-κB reporter activity (Fig. 3B–D). In contrast, the absence of β-catenin signaling in the HAβ85 cells produced the opposite results (Fig. 3B–D) and augmented NF-κB–dependent hiNOS and Traf1 protein expression (Fig. 5A). Through these experiments, we can conclude that stabilized expression of β-catenin attenuates hiNOS expression by inhibiting NF-κB pathway signaling.

In addition to testing mutant β-catenin cell lines, we also used SW480 colon cancer cells stably expressing WT APC (SW480-APC) to restore the truncated APC in the endogenous SW480 cells. The SW480-APC cells exhibited decreased β-catenin protein by Western blot and immunoflourescent staining (Fig. 4A and B) and enhanced basal and TNFα-induced hiNOS expression and NF-κB DNA binding activity (Fig. 4A–D). These results also support our finding of an inverse correlation between β-catenin and TNFα-induced NF-κB–dependent gene expression. These results also highlight that mutations in either β-catenin or APC (leading to altered cytosolic β-catenin levels) can modulate NF-κB–dependent target gene expression.

Two other recent studies have shown a relationship between β-catenin and NF-κB signaling (31, 37). Deng and colleagues (31) first showed that β-catenin inhibited NF-κB activity in human colon and breast cancer cell lines; however, they did not examine for β-catenin or NF-κB–dependent gene expression in primary liver cancer specimens. Subsequently, Sun and colleagues (37) reported a cross talk between NF-κB and β-catenin in bacterial-colonized HCT116 intestinal epithelial cells. In this study, constitutively expressed β-catenin indirectly stabilized IκBα and thereby inhibited NF-κB. In our study, we did not find a major change in IκBα levels among cancer cell lines with different degrees of β-catenin expression (Supplementary Fig. S1). Instead, we provided evidence by coimmunoprecipitation for a direct interaction between β-catenin and both p50 and p65 proteins. Neither of these studies determined the effect of β-catenin on hiNOS gene expression. Further, we developed stably transformed SW480 cells overexpressing APC and showed that this resulted in lower β-catenin protein levels, lower β-catenin transcriptional activity, and higher NF-κB DNA binding activity, and subsequently increased hiNOS mRNA and protein expressions in colon cancer cells. This is very important because it shows that manipulation of Wnt signaling by either APC or β-catenin can influence NF-κB–mediated target gene expression. Taken together, the inverse correlation between β-catenin and hiNOS expression indicates that activated β-catenin predominantly controls TNFα-induced hiNOS expression and NO production via interaction between β-catenin and NF-κB in cancer cells.

In contrast, Bandino and colleagues (44) recently reported that EGTA/LiCl inactivation of GSK-3β increased β-catenin and resulted in NF-κB–dependent activation of iNOS in rat hepatocytes. Their experimental design examined cell adherens junction disruption by collagenase liver perfusion and did not address the role of β-catenin signaling in cytokine-stimulated iNOS expression or the direct effects of β-catenin on NF-κB–dependent gene activation in tumor cells.

To strengthen the significance of the findings in the cancer cells in vitro, we also investigated the relationship between β-catenin levels and NF-κB–dependent hiNOS and Fas expression in vivo in resected hepatocellular carcinoma and metastatic colorectal cancer liver tumors. We observed that β-catenin inversely correlated with iNOS or Fas expression in these tumors (Fig. 6C; Supplementary Table S1). The in vivo tumor findings suggest that the cross talk between Wnt/β-catenin and NF-κB signaling occurs in the setting of inflammation-associated liver cancer and has also been reported in breast cancer specimens (45). We acknowledge that this study does not provide correlation between the level of β-catenin/iNOS expression and the aggressiveness of the cancer and overall patient prognosis. Such a study would require long-term follow-up in a larger group of patients.

Our results add insight to the complexity of the Wnt/β-catenin and NF-κB/iNOS/NO signaling pathways involved in carcinogenesis (38, 4648). Kinzler and Vogelstein (48) defined the important role of Wnt/β-catenin in colon cancer, and Pikarsky and colleagues (38) pointed out that NF-κB functions as a tumor promoter in inflammation-associated cancer. Hofseth and colleagues (11) highlighted the interaction between NO and p53 as a crucial pathway in inflammatory-mediated carcinogenesis, and also reported that cyclooxygenase-2 (COX-2) is a target of Wnt/β-catenin signaling (49). Another study showed that matrix metalloproteinases mediate NO-induced dissociation of β-catenin from membrane-bound E-cadherin and the formation of nuclear β-catenin/LEF-1 complex in mouse colonic epithelial cells (50). Our data show that Wnt/β-catenin signaling in cancer cells can diminish the overproduction of cytokine-induced iNOS, which contributes to apoptosis by NO-mediated cytotoxic mechanisms (41), and is consistent with a study wherein β-catenin overexpression blocked NO-induced apoptosis in colon cancer cells (49). Further efforts will study the working model of Wnt/β-catenin, NF-κB, iNOS/NO, and COX-2 contribution to carcinogenesis.

No potential conflicts of interest were disclosed.

Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

Grant support: NIH grants R01-GM52021/RO1-DK62313 (D.A. Geller).

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
Nusse R. Wnt signaling in disease and in development.
Cell Res
2005
;
15
:
28
–32.
2
Morin PJ, Sparks AB, Korinek V, et al. Activation of β-catenin-Tcf signaling in colon cancer by mutations in β-catenin or APC.
Science
1997
;
275
:
1787
–90.
3
Korinek V, Barker N, Morin PJ, et al. Constitutive transcriptional activation by a β-catenin-Tcf complex in APC−/− colon carcinoma.
Science
1997
;
275
:
1784
–7.
4
Clevers H. Colon cancer—understanding how NSAIDs work.
N Engl J Med
2006
;
354
:
761
–3.
5
Clevers H. At the crossroads of inflammation and cancer.
Cell
2004
;
118
:
671
–4.
6
van de WM, Sancho E, Verweij C, et al. The β-catenin/TCF-4 complex imposes a crypt progenitor phenotype on colorectal cancer cells.
Cell
2002
;
111
:
241
–50.
7
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.
8
Stuehr DJ, Marletta MA. Mammalian nitrate biosynthesis: mouse macrophages produce nitrite and nitrate in response to Escherichia coli lipopolysaccharide.
Proc Natl Acad Sci U S A
1985
;
82
:
7738
–42.
9
Bogdan C. Nitric oxide and the immune response.
Nat Immunol
2001
;
2
:
907
–16.
10
Wink DA, Ridnour LA, Hussain SP, Harris CC. The reemergence of nitric oxide and cancer.
Nitric Oxide
2008
;
19
:
65
–7.
11
Hofseth LJ, Hussain SP, Wogan GN, Harris CC. Nitric oxide in cancer and chemoprevention.
Free Radic Biol Med
2003
;
34
:
955
–68.
12
Hussain SP, Hofseth LJ, Harris CC. Radical causes of cancer.
Nat Rev Cancer
2003
;
3
:
276
–85.
13
Wink DA, Kasprzak KS, Maragos CM, et al. DNA deaminating ability and genotoxicity of nitric oxide and its progenitors.
Science
1991
;
254
:
1001
–3.
14
Jenkins DC, Charles IG, Thomsen LL, et al. Roles of nitric oxide in tumor growth.
Proc Natl Acad Sci U S A
1995
;
92
:
4392
–6.
15
Middleton SJ, Shorthouse M, Hunter JO. Increased nitric oxide synthesis in ulcerative colitis.
Lancet
1993
;
341
:
465
–6.
16
Majano PL, Garcia-Monzon C, Lopez-Cabrera M, et al. Inducible nitric oxide synthase expression in chronic viral hepatitis. Evidence for a virus-induced gene up-regulation.
J Clin Invest
1998
;
101
:
1343
–52.
17
Ambs S, Hussain SP, Harris CC. Interactive effects of nitric oxide and the p53 tumor suppressor gene in carcinogenesis and tumor progression.
FASEB J
1997
;
11
:
443
–8.
18
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.
19
Forrester K, Ambs S, Lupold SE, et al. Nitric oxide-induced p53 accumulation and regulation of inducible nitric oxide synthase expression by wild-type p53.
Proc Natl Acad Sci U S A
1996
;
93
:
2442
–7.
20
Ambs S, Ogunfusika MO, Merriam WG, et al. Up-regulation of inducible nitric oxide synthase expression in cancer-prone p53 knockout mice.
Proc Natl Acad Sci U S A
1998
;
95
:
8823
–8.
21
Stamler JS, Singel DJ, Loscalzo J. Biochemistry of nitric oxide and its redox-activated forms.
Science
1992
;
258
:
1898
–902.
22
Lancaster JR, Jr., Xie K. Tumors face NO problems?
Cancer Res
2006
;
66
:
6459
–62.
23
Lowenstein CJ, Alley EW, Raval P, et al. Macrophage nitric oxide synthase gene: two upstream regions mediate induction by interferon γ and lipopolysaccharide.
Proc Natl Acad Sci U S A
1993
;
90
:
9730
–4.
24
de Vera ME, Shapiro RA, Nussler AK, et al. Transcriptional regulation of human inducible nitric oxide synthase (NOS2) gene by cytokines: initial analysis of the human NOS2 promoter.
Proc Natl Acad Sci U S A
1996
;
93
:
1054
–9.
25
Taylor BS, de Vera ME, Ganster RW, et al. Multiple NF-κB enhancer elements regulate cytokine induction of the human inducible nitric oxide synthase gene.
J Biol Chem
1998
;
273
:
15148
–56.
26
Ganster RW, Taylor BS, Shao L, Geller DA. Complex regulation of human inducible nitric oxide synthase gene transcription by Stat 1 and NF-κB.
Proc Natl Acad Sci U S A
2001
;
98
:
8638
–43.
27
Feng X, Guo Z, Nourbakhsh M, et al. Identification of a negative response element in the human inducible nitric-oxide synthase (hiNOS) promoter: The role of NF-κB-repressing factor (NRF) in basal repression of the hiNOS gene.
Proc Natl Acad Sci U S A
2002
;
99
:
14212
–7.
28
Karin M. NF-κB and cancer: mechanisms and targets.
Mol Carcinog
2006
;
45
:
355
–61.
29
Karin M. Nuclear factor-κB in cancer development and progression.
Nature
2006
;
441
:
431
–6.
30
Du Q, Park KS, Guo Z, et al. Regulation of human nitric oxide synthase 2 expression by Wnt β-catenin signaling.
Cancer Res
2006
;
66
:
7024
–31.
31
Deng J, Miller SA, Wang HY, et al. β-Catenin interacts with and inhibits NF-κB in human colon and breast cancer.
Cancer Cell
2002
;
2
:
323
–34.
32
Kim JS, Crooks H, Dracheva T, et al. Oncogenic β-catenin is required for bone morphogenetic protein 4 expression in human cancer cells.
Cancer Res
2002
;
62
:
2744
–8.
33
Ganster RW, Guo Z, Shao L, Geller DA. Differential effects of TNF-α and IFN-γ on gene transcription mediated by NF-κB-Stat1 interactions.
J Interferon Cytokine Res
2005
;
25
:
707
–19.
34
Geller DA, de Vera ME, Russell DA, et al. A central role for IL-1β in the in vitro and in vivo regulation of hepatic inducible nitric oxide synthase. IL-1β induces hepatic nitric oxide synthesis.
J Immunol
1995
;
155
:
4890
–8.
35
Kolodziejski PJ, Rashid MB, Eissa NT. Intracellular formation of “undisruptable” dimers of inducible nitric oxide synthase.
Proc Natl Acad Sci U S A
2003
;
100
:
14263
–8.
36
Wang H, MacNaughton WK. Overexpressed β-catenin blocks nitric oxide-induced apoptosis in colonic cancer cells.
Cancer Res
2005
;
65
:
8604
–7.
37
Sun J, Hobert ME, Duan Y, et al. Crosstalk between NF-κB and β-catenin pathways in bacterial-colonized intestinal epithelial cells.
Am J Physiol Gastrointest Liver Physiol
2005
;
289
:
G129
–137.
38
Pikarsky E, Porat RM, Stein I, et al. NF-κB functions as a tumour promoter in inflammation-associated cancer.
Nature
2004
;
431
:
461
–6.
39
Geller DA, Nussler AK, Di SM, et al. Cytokines, endotoxin, and glucocorticoids regulate the expression of inducible nitric oxide synthase in hepatocytes.
Proc Natl Acad Sci U S A
1993
;
90
:
522
–6.
40
Kitade H, Sakitani K, Inoue K, et al. Interleukin 1β markedly stimulates nitric oxide formation in the absence of other cytokines or lipopolysaccharide in primary cultured rat hepatocytes but not in Kupffer cells.
Hepatology
1996
;
23
:
797
–802.
41
Kim PK, Zamora R, Petrosko P, Billiar TR. The regulatory role of nitric oxide in apoptosis.
Int Immunopharmacol
2001
;
1
:
1421
–41.
42
Mannick JB, Asano K, Izumi K, Kieff E, Stamler JS. Nitric oxide produced by human B lymphocytes inhibits apoptosis and Epstein-Barr virus reactivation.
Cell
1994
;
79
:
1137
–46.
43
Lu T, Sathe SS, Swiatkowski SM, Hampole CV, Stark GR. Secretion of cytokines and growth factors as a general cause of constitutive NFκB activation in cancer.
Oncogene
2004
;
23
:
2138
–45.
44
Bandino A, Compagnone A, Bravoco V, et al. β-Catenin triggers nuclear factor κB-dependent up-regulation of hepatocyte inducible nitric oxide synthase.
Int J Biochem Cell Biol
2008
;
40
:
1861
–71.
45
Deng J, Xia W, Miller SA, et al. Crossregulation of NF-κB by the APC/GSK-3β/β-catenin pathway.
Mol Carcinog
2004
;
39
:
139
–46.
46
Wang CY, Mayo MW, Korneluk RG, Goeddel DV, Baldwin AS, Jr. NF-κB antiapoptosis: induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation.
Science
1998
;
281
:
1680
–3.
47
Rubinfeld B, Robbins P, El-Gamil M, et al. Stabilization of β-catenin by genetic defects in melanoma cell lines.
Science
1997
;
275
:
1790
–2.
48
Kinzler KW, Vogelstein B. Lessons from hereditary colorectal cancer.
Cell
1996
;
87
:
159
–70.
49
Araki Y, Okamura S, Hussain SP, et al. Regulation of cyclooxygenase-2 expression by the Wnt and ras pathways.
Cancer Res
2003
;
63
:
728
–34.
50
Mei JM, Borchert GL, Donald SP, Phang JM. Matrix metalloproteinase(s) mediate(s) NO-induced dissociation of β-catenin from membrane bound E-cadherin and formation of nuclear β-catenin/LEF-1 complex.
Carcinogenesis
2002
;
23
:
2119
–22.