To determine the role of 14-3-3 in colorectal cancer apoptosis induced by nonsteroidal anti-inflammatory drugs (NSAIDs), we evaluated the effects of sulindac on 14-3-3ε protein expression in colorectal cancer cells. Sulindac sulfide inhibited 14-3-3ε proteins in HT-29 and DLD-1 cells in a time- and concentration-dependent manner. Sulindac sulfone at 600 μmol/L inhibited 14-3-3ε protein expression in HT-29. Indomethacin and SC-236, a selective cyclooxygenase-2 (COX-2) inhibitor, exerted a similar effect as sulindac. Sulindac suppressed 14-3-3ε promoter activity. As 14-3-3ε promoter activation is mediated by peroxisome proliferator–activated receptor δ (PPARδ), we determined the correlation between 14-3-3ε inhibition and PPARδ suppression by NSAIDs. Sulindac sulfide inhibited PPARδ protein expression and PPARδ transcriptional activity. Overexpression of PPARδ by adenoviral transfer rescued 14-3-3ε proteins from elimination by sulindac or indomethacin. NSAID-induced 14-3-3ε suppression was associated with reduced cytosolic Bad with elevation of mitochondrial Bad and increase in apoptosis which was rescued by Ad-PPARδ transduction. Stable expression of 14-3-3ε in HT-29 significantly protected cells from apoptosis. Our findings shed light on a novel mechanism by which NSAIDs induce colorectal cancer apoptosis via the PPARδ/14-3-3ε transcriptional pathway. These results suggest that 14-3-3ε is a target for the prevention and therapy of colorectal cancer. [Cancer Res 2007;67(7):3185–91]

Human and animal studies have shown that nonsteroidal anti-inflammatory drugs (NSAIDs) suppress colorectal tumorigenesis (1). Population-based epidemiologic studies have shown that use of aspirin and NSAID reduces risk of colorectal cancer (2, 3). Animal experiments provide evidence for control of colorectal cancer by NSAIDs such as sulindac and selective cyclooxygenase-2 (COX-2) inhibitors (4, 5). Results from clinical trials have documented the efficacy of sulindac and selective COX-2 inhibitors in reducing the size and numbers of colon adenomatous polyps in patients with familial adenomatous polyposis, a precancerous condition (6, 7). The action of NSAIDs on controlling colorectal cancer is attributed to the inhibition of COX-2 (8), which is overexpressed in a majority of patients with colorectal cancer (9, 10). NSAID could also exert its action on colorectal cancer in a COX-2–independent manner (11). It is well documented that NSAIDs, notably sulindac, indomethacin, and selective COX-2 inhibitors exert their actions by inducing colorectal cancer cell apoptosis (12, 13). The mechanism by which these agents induce apoptosis is not entirely clear. It was proposed that NSAID-induced apoptosis might be mediated by down-regulation of peroxisome proliferator–activated receptor-δ (PPARδ; ref. 14). Several studies have shown that PPARδ is overexpressed in colorectal cancer and PPARδ is activated by prostacyclin generated by colorectal cancer cells (15). Activation of PPARδ accelerates intestinal adenoma growth whereas genetic disruption of PPARδ decreases the tumorigenicity of human colorectal cancer cells (16, 17). It has been reported that PPARδ plays an important role in cellular resistance to apoptotic insults (18, 19). Activation of PPARδ was reported to be associated with increased expression of phosphoinositide-dependent kinase-1 and activation of Akt (20). Akt is recognized as a key signaling molecule for transmitting survival messages. We have recently reported that PPARδ activation by prostacyclin or L-165041, a synthetic PPARδ ligand, increases the expression of 14-3-3 proteins, especially the 14-3-3ε isoform (21). Promoter analysis identifies three PPAR response elements (PPRE) located between −1426 and −1477 of the 14-3-3ε promoter region. Deletion of these PPREs abrogates the protective effect of prostacyclin or L-165041 (21). Our previous findings suggest that PPARδ-mediated 14-3-3ε up-regulation plays a crucial role in endothelial cell resistance to apoptosis (21). It is unclear whether 14-3-3ε is involved in colorectal cancer cell survival. We postulated in this study that NSAIDs induce apoptosis by reducing 14-3-3ε via suppressing PPARδ. Reduction in 14-3-3ε protein levels results in mitochondrial Bad translocation and colorectal cancer cell apoptosis. The results show that sulindac sulfide, sulindac sulfone, indomethacin, and SC-236 suppressed HT-29 14-3-3ε protein and promoter activity, which correlated with the suppression of PPARδ protein expression. The reduction in 14-3-3ε levels was accompanied by enhanced mitochondrial Bad translocation and HT-29 apoptosis.

Cell culture and reagents. HT-29 and DLD-1 cells were obtained from American Type Culture Collection (Manassas, VA). HT-29 were maintained in DMEM and DLD-1 were maintained in RPMI 1604 supplemented with 10% fetal bovine serum and penicillin-streptomycin at 37°C in a humidified 5% CO2 atmosphere. Cell culture media and antibiotics were purchased from BRL Life Technologies (Grand Island, NY). Sulindac sulfide and sulindac sulfone were obtained from ICN Biomedicals, Inc. (Solon, OH). Indomethacin, SC-236, MG-132, and cell-permeable caspase 3 inhibitor, ac-DVED aldehyde (DEVD-CHO), were from Calbiochem (San Diego, CA). We used 10 μmol/L of MG-132 and 50 μmol/L of DEVD-CHO, which were reported to block proteasome and caspase 3 activities, respectively (22, 23).

Recombinant adenoviral vectors. Replication-defective recombinant adenoviruses were generated by homologous recombination and amplified in 293 cells as described previously (24, 25). The recombinant viruses were purified by CsCl density gradient centrifugation and the virus titers were determined by a plaque assay as previously described (24). Ad-PPARδ was kindly provided by Drs. Kinzler and Vogelstein at Johns Hopkins University. HT-29 cells were transfected with Ad-GFP (green fluorescent protein) or Ad-PPARδ at a multiplicity of infection (moi) of 50 for 48 h before treatment with indomethacin or sulindac sulfide.

Preparation of mitochondrial fraction. HT-29 cells treated with indicated reagents (sulindac sulfide, 160 μmol/L; sulindac sulfone, 300 μmol/L; indomethacin, 800 μmol/L; or SC-236, 40 μmol/L) for 8 h were washed thrice with PBS and harvested by centrifugation. Mitochondrial fractions were prepared by a mitochondria isolation kit (Sigma, St. Louis, MO) as described previously (26). The mitochondrial and cytosolic fractions were purified by two-step gradient centrifugation and stored at −20°C. Mitochondrial Bad protein level was analyzed by Western blotting. Heat shock protein 60 (Hsp60) was used as a mitochondrial marker.

Immunoprecipitation. HT-29 cells treated with the indicated reagents were harvested and lysed in 1× radioimmunoprecipitation assay buffer (Upstate, Charlottesville, VA) with cocktail protease inhibitors (Sigma). Lysate proteins (200 μg) were immunoprecipitated with a human 14-3-3ε antibody. The immunoprecipitated complex was pulled down with protein A/G-agarose (Santa Cruz Biotechnology, Santa Cruz, CA). After washing five times, the proteins were analyzed by Western blotting using a Bad antibody.

Western blot analysis. Cells were lysed in radioimmunoprecipitation assay buffer containing protease inhibitors. Protein concentrations were determined by a Bio-Rad (Hercules, CA) protein assay kit. Cell lysate proteins (25 μg) were applied to each lane and analyzed by Western blots as previously described (25). Rabbit polyclonal antibodies against 14-3-3ε (diluted at 1:250), and goat polyclonal antibodies against Hsp60 (1:2,000) were purchased from Santa Cruz Biotechnology. Rabbit polyclonal antibody against PPARδ (1:500) was obtained from Cayman Chemical (Ann Arbor, MI). Rabbit polyclonal antibody against Bad (1:500) was purchased from Cell Signaling (Beverly, MA). Monoclonal antibody against actin (1:5,000) was obtained from Oncogene (Boston, MA). Donkey anti-rabbit or donkey anti-mouse IgG conjugated with horseradish peroxidase were purchased from Santa Cruz Biotechnology. Protein bands were visualized by an enhanced chemiluminescence system (Pierce, Rockford, IL).

Assay of apoptosis. Apoptotic cells were analyzed by Hoechst staining according to the instructions of the manufacturer (Invitrogen, Carlsbad, CA). Hoechst 33258 is a cell-permeable dye which is sensitive to nuclear DNA damage and is useful in detecting apoptosis of unfixed living cells (27). In brief, HT-29 cells transiently transfected with Ad-PPARδ, Ad-GFP, or stably transfected HT-29 cells were treated with 160 μmol/L of sulindac sulfide for 24 h. After washing, they were incubated with 1 μg/mL of Hoechst 33258 in the dark for 15 min, centrifuged and washed with PBS. The washed cells were examined under fluorescent microscopy (excitation at 352 nm and emission at 461 nm). Apoptotic cells exhibited strong nuclear staining with overt nuclear morphologic changes as previously described (27). The percentage of apoptotic cells was determined by counting at least 200 cells.

Plasmid constructs and luciferase reporter assay. A human 14-3-3ε expression construct was amplified by PCR and subcloned into pCDNA 3.1+ vector (Invitrogen) and a 14-3-3ε promoter subcloned into pGL3 vector were prepared as previously described (21). PPRE reporter (14) was kindly provided by Drs. Kinzler and Vogelstein at Johns Hopkins University. For the luciferase assay, cells were transfected with the reporter constructs by Effectene transfection kit (Qiagen, Valencia, CA) for 48 h. After treatment with the indicated reagents, cells were lysed and luciferase activity was measured using a kit from Promega (Madison, WI), and the emitted light was determined in a luminometer.

Establishment of 14-3-3ε stable cell line. Control vector (pCDNA3.1+) and human 14-3-3ε expression vectors which contain a neomycin-resistant cassette were transfected into HT-29 cells by using an Effectene transfection kit for 48 h. The transfected cells were screened by incubation with 500 μg/mL of G418 (Sigma) for 4 weeks. The cell lines which stably expressed 14-3-3ε were isolated from individual colonies and maintained in DMEM with 10% fetal bovine serum and 200 μg/mL of G418.

Statistical analysis. ANOVA was used to analyze statistical differences among multiple groups. P < 0.05 was considered to be statistically significant.

Suppression of 14-3-3ε by sulindac sulfide and COX-2 inhibitors. To evaluate the effect of sulindac on 14-3-3ε protein levels, we treated HT-29 with sulindac sulfide at 0 to 160 μmol/L for 4 to 24 h. 14-3-3ε proteins were analyzed by Western blotting. Sulindac sulfide did not have an effect at 4 h of treatment, suppressed 14-3-3ε at 8 h only at 160 μmol/L, and exerted a concentration-dependent inhibition of 14-3-3ε at 24 h of treatment (Fig. 1A). Sulindac sulfone had no effect at 300 μmol/L but suppressed 14-3-3ε expression at 600 μmol/L (Fig. 1B). Indomethacin, a nonselective COX inhibitor, or SC-236, a selective COX-2 inhibitor, also concentration-dependently inhibited 14-3-3ε protein levels (Fig. 1C). Sulindac sulfide suppressed 14-3-3ε protein levels in DLD-1 with a pattern similar to that of HT-29 (Fig. 1D).

Figure 1.

NSAIDs suppressed 14-3-3ε expression. HT-29 cells were treated with (A) sulindac (Sul) sulfide for 4 to 24 h, (B) sulindac sulfone for 24 h, or (C) indomethacin (Indo) or SC-236 for 24 h. D, DLD-1 cells were treated with sulindac sulfide for 24 h. 14-3-3ε proteins were determined by Western blotting. Actin was determined as control. Representative of three independent experiments with similar results.

Figure 1.

NSAIDs suppressed 14-3-3ε expression. HT-29 cells were treated with (A) sulindac (Sul) sulfide for 4 to 24 h, (B) sulindac sulfone for 24 h, or (C) indomethacin (Indo) or SC-236 for 24 h. D, DLD-1 cells were treated with sulindac sulfide for 24 h. 14-3-3ε proteins were determined by Western blotting. Actin was determined as control. Representative of three independent experiments with similar results.

Close modal

Inhibition of 14-3-3ε promoter activity by sulindac sulfide. To determine whether sulindac sulfide inhibits 14-3-3ε at the transcription level, we transfected HT-29 with a human 14-3-3ε promoter construct (−1628/+24) and treated the transfected cells with various concentrations of sulindac sulfide for 24 h. Sulindac sulfide concentration-dependently inhibited 14-3-3ε promoter activity (Fig. 2A). Sulindac-induced 14-3-3ε protein suppression was not influenced by MG-132, a proteasome inhibitor, or DEVD-CHO, a caspase 3 inhibitor (Fig. 2B). Taken together, these results suggest that sulindac suppresses 14-3-3ε primarily at the transcriptional level.

Figure 2.

Sulindac sulfide suppressed 14-3-3ε expression through transcriptional inactivation. A, HT-29 cells transfected with a 14-3-3ε promoter construct were treated with increasing concentrations of sulindac sulfide for 24 h. Luciferase expression was measured and expressed as relative light units (RLU)/μg protein. Columns, mean of three independent experiments; bars, SD. Statistical difference was analyzed by ANOVA. B, HT-29 cells were pretreated with MG-132 (10 μmol/L) or DEVD-CHO (50 μmol/L) for 30 min followed by sulindac sulfide (160 μmol/L) for 24 h. 14-3-3ε proteins were determined by Western blotting. This blot is representative of three experiments with similar results.

Figure 2.

Sulindac sulfide suppressed 14-3-3ε expression through transcriptional inactivation. A, HT-29 cells transfected with a 14-3-3ε promoter construct were treated with increasing concentrations of sulindac sulfide for 24 h. Luciferase expression was measured and expressed as relative light units (RLU)/μg protein. Columns, mean of three independent experiments; bars, SD. Statistical difference was analyzed by ANOVA. B, HT-29 cells were pretreated with MG-132 (10 μmol/L) or DEVD-CHO (50 μmol/L) for 30 min followed by sulindac sulfide (160 μmol/L) for 24 h. 14-3-3ε proteins were determined by Western blotting. This blot is representative of three experiments with similar results.

Close modal

Restoration of 14-3-3ε by adenoviral PPARδ transfer. We have recently discovered that 14-3-3ε transcription is mediated by PPARδ (21). We were, therefore, interested in learning whether sulindac-induced 14-3-3ε suppression involves PPARδ. HT-29 cells were treated with sulindac sulfide (160 μmol/L) or indomethacin (800 μmol/L) for 24 h and PPARδ proteins were analyzed by Western blots. PPARδ levels became undetectable following treatment with either agent (Fig. 3A). To ensure that PPARδ is active in HT-29, we transfected the cells with a PPRE construct and treated the cells with sulindac sulfide for 24 h. Sulindac reduced the PPRE activity in a concentration-dependent manner (Fig. 3B). To determine whether PPARδ overexpression rescues 14-3-3ε from sulindac or indomethacin suppression, we transduced HT-29 with Ad-PPARδ (50 moi) or Ad-GFP control (50 moi). PPARδ protein levels were increased by Ad-PPARδ transduction (Fig. 4A), which is accompanied by enhanced PPRE activity (Fig. 4B) and 14-3-3ε promoter activity (Fig. 4C). PPARδ proteins in Ad-PPARδ–transduced cells were not altered by sulindac or indomethacin (Fig. 4A), but the PPARδ transcriptional activity (Fig. 4B) and the 14-3-3ε promoter activity (Fig. 4C) were partially suppressed. 14-3-3ε proteins were increased by Ad-PPARδ overexpression which were suppressed by sulindac sulfide and indomethacin (Fig. 4D).

Figure 3.

NSAIDs inhibited PPARδ. A, HT-29 cells treated with sulindac sulfide (160 μmol/L) or indomethacin (800 μmol/L) for 24 h. PPARδ protein was determined by Western blotting. This blot is representative of three experiments with similar results. B, HT-29 cells transfected with a PPRE construct were treated with sulindac sulfide for 24 h. Luciferase expression was measured. Columns, mean of three independent experiments; bars, SD.

Figure 3.

NSAIDs inhibited PPARδ. A, HT-29 cells treated with sulindac sulfide (160 μmol/L) or indomethacin (800 μmol/L) for 24 h. PPARδ protein was determined by Western blotting. This blot is representative of three experiments with similar results. B, HT-29 cells transfected with a PPRE construct were treated with sulindac sulfide for 24 h. Luciferase expression was measured. Columns, mean of three independent experiments; bars, SD.

Close modal
Figure 4.

PPARδ overexpression restored 14-3-3ε expression. A, HT-29 cells were transfected with Ad-GFP or Ad-PPARδ for 24 h followed by treatment with sulindac sulfide (160 μmol/L) or indomethacin (800 μmol/L) for an additional 24 h. PPARδ proteins were determined by Western blotting. B to D, HT-29 transfected with Ad-GFP or Ad-PPARδ for 24 h followed by treatment of sulindac sulfide or indomethacin at indicated concentrations for an additional 24 h; PPRE (B) and 14-3-3ε (C) promoter activity were determined by luciferase expression. D, 14-3-3ε protein was determined by Western blotting. Each blot is representative of three experiments with similar results. NS, statistically nonsignificant; *, P < 0.05; **, P < 0.01.

Figure 4.

PPARδ overexpression restored 14-3-3ε expression. A, HT-29 cells were transfected with Ad-GFP or Ad-PPARδ for 24 h followed by treatment with sulindac sulfide (160 μmol/L) or indomethacin (800 μmol/L) for an additional 24 h. PPARδ proteins were determined by Western blotting. B to D, HT-29 transfected with Ad-GFP or Ad-PPARδ for 24 h followed by treatment of sulindac sulfide or indomethacin at indicated concentrations for an additional 24 h; PPRE (B) and 14-3-3ε (C) promoter activity were determined by luciferase expression. D, 14-3-3ε protein was determined by Western blotting. Each blot is representative of three experiments with similar results. NS, statistically nonsignificant; *, P < 0.05; **, P < 0.01.

Close modal

Mitochondrial Bad translocation and cell death by sulindac and COX-2 inhibitors. 14-3-3 proteins bind phosphorylated Bad and sequester it in cytosol. Apoptotic signals induce Bad to release from 14-3-3 and translocate to the mitochondria where it disrupts the protective action of Bcl-XL, thereby promoting apoptosis via the mitochondrial pathway (28, 29). It was reported that up-regulation of 14-3-3ε protein levels by PPARδ ligands enhances Bad binding and reduces Bad translocation to mitochondria (21). Because sulindac suppresses 14-3-3ε in HT-29, it is predicted that sulindac reduces cytosolic Bad sequestration and reciprocally enhances Bad localization to mitochondria. To confirm this, we did two types of experiments. First, we prepared mitochondrial and cytosolic fractions from HT-29 treated with or without sulindac sulfide or sulfone, indomethacin, or SC-236, and Bad in the mitochondrial fraction was analyzed by Western blotting. As a control, the mitochondria-resident protein Hsp60 was concurrently analyzed. Second, we immunoprecipitated cell lysates of HT-29 treated with or without sulindac sulfide or indomethacin by a 14-3-3ε antibody and analyzed Bad in the immunoprecipitate with a Bad antibody. The results show that in the absence of NSAIDs, most Bad were localized to the cytosol, whereas Hsp60 was predominantly detected in the mitochondrial fraction with a trace quantity detected in the cytosolic fraction (Fig. 5A). Bad was detected at a very low level in mitochondrial fraction (Fig. 5A). Sulindac and indomethacin enhanced mitochondrial Bad (Fig. 5A) and suppressed binding of Bad to 14-3-3ε (Fig. 5B). SC-236 enhanced mitochondrial Bad to an extent comparable to sulindac sulfide and indomethacin (Fig. 5A). These results suggest that sulindac, indomethacin and SC-236 induce Bad translocation to mitochondria as a consequence of severe 14-3-3ε suppression.

Figure 5.

NSAIDs induced Bad translocation to mitochondria and apoptosis. A, HT-29 cells were treated with sulindac sulfone (300 μmol/L), sulindac sulfide (160 μmol/L), indomethacin (800 μmol/L), or SC-236 (40 μmol/L) for 8 h. Mitochondrial and cytosolic fractions were isolated and Bad proteins were determined by Western blotting. Hsp60 was analyzed to validate the homogeneity of mitochondrial fraction. B, HT-29 cells were treated with sulindac sulfide (160 μmol/L) or indomethacin (800 μmol/L) for 8 h. Cell lysates were immunoprecipitated with a 14-3-3ε antibody. Bad proteins in the precipitate were analyzed by Western blotting. C, Ad-GFP and Ad-PPARδ transfected HT-29 cells were treated with 160 μmol/L of sulindac sulfide or 800 μmol/L of indomethacin for 24 h. Percentages of apoptotic cells were determined by Hoechst staining. Columns, mean of three independent experiments; bars, SD.

Figure 5.

NSAIDs induced Bad translocation to mitochondria and apoptosis. A, HT-29 cells were treated with sulindac sulfone (300 μmol/L), sulindac sulfide (160 μmol/L), indomethacin (800 μmol/L), or SC-236 (40 μmol/L) for 8 h. Mitochondrial and cytosolic fractions were isolated and Bad proteins were determined by Western blotting. Hsp60 was analyzed to validate the homogeneity of mitochondrial fraction. B, HT-29 cells were treated with sulindac sulfide (160 μmol/L) or indomethacin (800 μmol/L) for 8 h. Cell lysates were immunoprecipitated with a 14-3-3ε antibody. Bad proteins in the precipitate were analyzed by Western blotting. C, Ad-GFP and Ad-PPARδ transfected HT-29 cells were treated with 160 μmol/L of sulindac sulfide or 800 μmol/L of indomethacin for 24 h. Percentages of apoptotic cells were determined by Hoechst staining. Columns, mean of three independent experiments; bars, SD.

Close modal

Rescue of NSAID-induced apoptosis by Ad-PPARδ and 14-3-3ε. Sulindac sulfide and indomethacin induced HT-29 apoptosis as evidenced by an increase in Hoechst-positive cells. Apoptosis was reduced by transduction of cells with Ad-PPARδ (Fig. 5C). To ascertain the role of 14-3-3ε in NSAID-induced HT-29 apoptosis, we prepared a HT-29 cell line overexpressing 14-3-3ε (HT-2914-3-3ε) and a control cell line (HT-29CTR; Fig. 6A). In HT-29 control cells, sulindac suppressed 14-3-3ε proteins (Fig. 6A) accompanied by a large increase in apoptosis (Fig. 6B). Overexpression of 14-3-3ε proteins in HT-2914-3-3ε cells (Fig. 6A) prevented apoptosis by >50% (Fig. 6B).

Figure 6.

14-3-3ε overexpression rescued HT-29 from apoptosis. HT-29 stably transfected with 14-3-3ε (HT-2914-3-3ε) and control (HT-29CTR) were treated with or without sulindac sulfide (160 μmol/L) for 24 h. A, 14-3-3ε proteins were analyzed by Western blotting. B, apoptosis was analyzed by Hoechst staining. Columns, mean of three independent experiments; bars, SD. C, a schematic illustration of multiple biochemical processes via which NSAIDs induce apoptosis. Process 1 refers to a COX-2–dependent pathway in which NSAIDs inhibit COX-2–derived PGI2 and PGE2 productions, and thereby suppress PPARδ-mediated 14-3-3ε expression. Processes 2 and 3 are considered COX-2–independent. In process 2, NSAIDs block extracellular signal–regulated kinase (ERK) activation, thereby reducing Bad phosphorylation and enhancing Bad translocation to the mitochondria. In process 3, NSAIDs inhibit Bcl-XL and Bcl-2 protein levels. A common result of these processes is a reduced ratio of Bcl-XL and Bcl-2 to Bad and Bax at mitochondria, and an increased release of proapoptotic factors, notably Smac/DIABLO from mitochondria into the cytosol which contribute to caspase 3 activation and apoptosis.

Figure 6.

14-3-3ε overexpression rescued HT-29 from apoptosis. HT-29 stably transfected with 14-3-3ε (HT-2914-3-3ε) and control (HT-29CTR) were treated with or without sulindac sulfide (160 μmol/L) for 24 h. A, 14-3-3ε proteins were analyzed by Western blotting. B, apoptosis was analyzed by Hoechst staining. Columns, mean of three independent experiments; bars, SD. C, a schematic illustration of multiple biochemical processes via which NSAIDs induce apoptosis. Process 1 refers to a COX-2–dependent pathway in which NSAIDs inhibit COX-2–derived PGI2 and PGE2 productions, and thereby suppress PPARδ-mediated 14-3-3ε expression. Processes 2 and 3 are considered COX-2–independent. In process 2, NSAIDs block extracellular signal–regulated kinase (ERK) activation, thereby reducing Bad phosphorylation and enhancing Bad translocation to the mitochondria. In process 3, NSAIDs inhibit Bcl-XL and Bcl-2 protein levels. A common result of these processes is a reduced ratio of Bcl-XL and Bcl-2 to Bad and Bax at mitochondria, and an increased release of proapoptotic factors, notably Smac/DIABLO from mitochondria into the cytosol which contribute to caspase 3 activation and apoptosis.

Close modal

Our findings show for the first time that sulindac and other NSAIDs drastically suppress 14-3-3ε expression in colorectal cancer cells, resulting in reduced Bad sequestration in cytosol, increased mitochondrial Bad, and enhanced apoptosis. The 14-3-3 family comprises at least seven members in mammalian cells (30). They share sequence homology and biochemical properties, interact with more than 100 cellular proteins, and have diverse biological activities (31). They bind phosphorylated Bad, thereby sequestering Bad in cytosol and suppressing Bad-induced mitochondrial injury and the consequential apoptosis via the intrinsic (mitochondrial) apoptotic pathway (32). It was reported in endothelial cells that among the 14-3-3 members, 14-3-3ε is expressed in abundance, and its cellular level is regulated and is crucial in protecting cells from oxidative damage (21). The results in this study show that 14-3-3ε is constitutively expressed in HT-29 and DLD-1 cells and is a target of NSAID-induced apoptosis. Sulindac, indomethacin, and SC-236 suppress 14-3-3ε expression in HT-29 accompanied by increased translocation of Bad to mitochondria and apoptosis. Stable overexpression of 14-3-3ε alleviates sulindac-induced apoptosis. Taken together, these results strongly suggest that the suppression of 14-3-3ε is a major mechanism by which NSAIDs induce colorectal cancer apoptosis.

14-3-3ε transcription requires PPARδ and is regulated by the level of PPARδ, as evidenced by increased 14-3-3ε proteins in Ad-PPARδ–transduced cells. Our results are consistent with a previous report that sulindac suppresses PPARδ transcription activity (14), and provide new data which attribute reduced PPARδ transcription activity to the suppression of PPARδ protein expression. The results show a good correlation between sulindac- and indomethacin-induced 14-3-3ε and PPARδ suppressions. Furthermore, PPARδ overexpression by Ad-PPARδ transduction rescues 14-3-3ε from suppressions by sulindac sulfide at 80 μmol/L or indomethacin at 400 μmol/L. These data indicate that sulindac and other NSAIDs suppress PPARδ expression, thereby blocking PPARδ-mediated 14-3-3ε transcription. Although Ad-PPARδ abrogates 14-3-3ε suppression by sulindac and indomethacin at 80 and 300 μmol/L, respectively, it only partially corrected 14-3-3ε levels suppressed by higher concentrations of sulindac (160 μmol/L) or indomethacin (800 μmol/L). Lack of complete correction by Ad-PPARδ may be due to the following reasons. First, activated PPARδ promotes transcription by forming a heterodimer with RXR nuclear receptor (14). Overexpression of PPARδ alone without concurrent RXR overexpression may, therefore, be inadequate to exert maximal transcriptional activation. Second, sulindac and other NSAIDs have diverse COX-2–dependent and COX-2–independent effects on signaling transduction and gene expression. Lack of a complete rescue of 14-3-3ε suppression and cell apoptosis may suggest that PPARδ is not the only pathway via which NSAIDs suppress 14-3-3ε. Furthermore, differential effects of Ad-PPARδ (50 moi) on rescuing 80 versus 160 μmol/L sulindac-induced 14-3-3ε suppression may reflect dose-dependent effects on diverse cellular functions. Lastly, lack of a correlation between the magnitude of PPARδ overexpression and 14-3-3ε rescue could also be attributed to the requirement of PPARδ posttranslational modification for its full transactivation activity. Because neither MG-132 nor DEVD-CHO blocks the 14-3-3ε–suppressing effect of sulindac, it is unlikely that the reduction in 14-3-3ε protein levels are due to degradation by ubiquitin-proteasome or caspase 3.

PPARδ expression is regulated by the β-catenin-Tcf (T cell factor) transcriptional program (14). The wild-type adenomatous polyposis coli tumor suppressor gene product binds and targets β-catenin for degradation, thereby suppressing PPARδ expression (14). Adenomatous polyposis coli mutation results in increased β-catenin, which translocates to the nucleus and complexes with Tcf to form an active transcription factor that binds to specific responsive elements on PPARδ and up-regulates PPARδ expression in familial adenomatous polyposis (14, 33). It is reported that sulindac and celecoxib inhibit β-catenin expression and/or the transcriptional activity of the β-catenin/Tcf complex (34, 35). It is, therefore, possible that sulindac, SC-236, and other NSAIDs suppress PPARδ expression through the β-catenin/Tcf transcription pathway.

Sulindac and other NSAIDs may induce colorectal cancer cell apoptosis by COX-2–dependent and COX-2–independent mechanisms (3639). COX-2 is overexpressed in colorectal cancer and COX-2-derived prostaglandin I2 (PGI2) has been shown to activate PPARδ in colorectal cancer cells (15). PPARδ activation confers resistance to apoptosis in colorectal cancer as well as a number of non–cancer cells such as endothelial, renal interstitial, cardiomyocytes and keratinocytes (18, 19, 21). Sulindac sulfide and selective COX-2 inhibitors may induce apoptosis by blocking COX-2 derived PGI2 production, thereby suppressing PPARδ activation and the antiapoptosis action of PPARδ. COX-2–derived prostaglandin E2 (PGE2) up-regulates β-catenin expression (40) and NSAIDs may further suppress β-catenin–mediated PPARδ expression by blocking PGE2 production. However, a recent report has shown that sulindac, selective COX-2 inhibitors, and a host of classic NSAIDs inhibit β-catenin by a COX-2–independent mechanism (41). Our results reveal that sulindac sulfone, which does not inhibit COX-2, is capable of inhibiting 14-3-3ε expression, suggesting that NSAIDs suppress 14-3-3ε by a COX-2–independent mechanism. Regardless of the COX-2 involvement, an important finding is that reduction in 14-3-3ε levels by sulindac and other NSAIDs results in a decline in Bad sequestration and an increase in Bad translocation to the mitochondria where Bad binds and inactivates the antiapoptotic Bcl-2 family proteins, notably Bcl-XL and Bcl-2. It was reported that sulindac suppresses Bcl-XL expression, creating an imbalance between Bcl-XL and Bax, and subsequent mitochondria-mediated apoptosis (42). Moreover, sulindac inhibits Bcl-2 expression (43), further enhancing Bad- and Bax-mediated mitochondria damage, Smac/DIABLO release, and apoptosis (44). Taken together, our results and the published data suggest that sulindac and other NSAIDs induce colorectal cancer cell apoptosis by multiple mitochondrial apoptotic processes as proposed in Fig. 6C. First, NSAIDs suppress PPARδ, thereby reducing 14-3-3ε level and Bad sequestration. Second, NSAIDs inhibit extracellular signal–regulated kinase, thereby blocking Bad phosphorylation (45), and further reducing Bad sequestration leading to excessive Bad translocation to the mitochondria. Lastly, NSAIDs inhibit Bcl-XL and Bcl-2 expression resulting in a reduction in the ratio of antiapoptotic to proapoptotic Bcl-2 family proteins on the mitochondrial membrane with consequent loss of mitochondrial membrane potential, release of proapoptotic factors especially Smac/DIABLO resulting in caspase activation and apoptosis. These processes are interrelated and mutually reinforcing. As PPARδ or 14-3-3ε overexpression rescues apoptosis, the PPARδ/14-3-3ε transcriptional pathway represents a major pathway for colorectal cancer cell survival.

Grant support: NIH (National Heart, Lung, and Blood Institute, HL-50675).

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.

We thank Susan Mitterling and Nathalie Huang for excellent editorial assistance.

1
Gupta RA, Dubois RN. Colorectal cancer prevention and treatment by inhibition of cyclooxygenase-2.
Nat Rev Cancer
2001
;
1
:
11
–21.
2
Thun MJ. Aspirin and gastrointestinal cancer.
Adv Exp Med Biol
1997
;
400A
:
395
–402.
3
Smalley WE, DuBois RN. Colorectal cancer and nonsteroidal anti-inflammatory drugs.
Adv Pharmacol
1997
;
39
:
1
–20.
4
Jacoby RF, Marshall DJ, Newton MA, et al. Chemoprevention of spontaneous intestinal adenomas in the Apc Min mouse model by the nonsteroidal anti-inflammatory drug piroxicam.
Cancer Res
1996
;
56
:
710
–4.
5
Barnes CJ, Lee M. Chemoprevention of spontaneous intestinal adenomas in the adenomatous polyposis coli Min mouse model with aspirin.
Gastroenterology
1998
;
114
:
873
–7.
6
Janne PA, Mayer RJ. Chemoprevention of colorectal cancer.
N Engl J Med
2000
;
342
:
1960
–8.
7
Cruz-Correa M, Hylind LM, Romans KE, Booker SV, Giardiello FM. Long-term treatment with sulindac in familial adenomatous polyposis: a prospective cohort study.
Gastroenterology
2002
;
122
:
641
–5.
8
Dubois RN, Abramson SB, Crofford L, et al. Cyclooxygenase in biology and disease.
FASEB J
1998
;
12
:
1063
–73.
9
Eberhart CE, Coffey RJ, Radhika A, Giardiello FM, Ferrenbach S, DuBois RN. Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas.
Gastroenterology
1994
;
107
:
1183
–8.
10
Sano H, Kawahito Y, Wilder RL, et al. Expression of cyclooxygenase-1 and -2 in human colorectal cancer.
Cancer Res
1995
;
55
:
3785
–9.
11
Piazza GA, Alberts DS, Hixson LJ, et al. Sulindac sulfone inhibits azoxymethane-induced colon carcinogenesis in rats without reducing prostaglandin levels.
Cancer Res
1997
;
57
:
2909
–15.
12
Piazza GA, Rahm AL, Krutzsch M, et al. Antineoplastic drugs sulindac sulfide and sulfone inhibit cell growth by inducing apoptosis.
Cancer Res
1995
;
55
:
3110
–6.
13
Shiff SJ, Qiao L, Tsai LL, Rigas B. Sulindac sulfide, an aspirin-like compound, inhibits proliferation, causes cell cycle quiescence, and induces apoptosis in HT-29 colon adenocarcinoma cells.
J Clin Invest
1995
;
96
:
491
–503.
14
He TC, Chan TA, Vogelstein B, Kinzler KW. PPARδ is an APC-regulated target of nonsteroidal anti-inflammatory drugs.
Cell
1999
;
99
:
335
–45.
15
Gupta RA, Tan J, Krause WF, et al. Prostacyclin-mediated activation of peroxisome proliferator-activated receptor δ in colorectal cancer.
Proc Natl Acad Sci U S A
2000
;
97
:
13275
–80.
16
Gupta RA, Wang D, Katkuri S, Wang H, Dey SK, DuBois RN. Activation of nuclear hormone receptor peroxisome proliferator-activated receptor-δ accelerates intestinal adenoma growth.
Nat Med
2004
;
10
:
245
–7.
17
Park BH, Vogelstein B, Kinzler KW. Genetic disruption of PPARδ decreases the tumorigenicity of human colon cancer cells.
Proc Natl Acad Sci U S A
2001
;
98
:
2598
–603.
18
Tan NS, Michalik L, Noy N, et al. Critical roles of PPAR β/δ in keratinocyte response to inflammation.
Genes Dev
2001
;
15
:
3263
–77.
19
Hao CM, Redha R, Morrow J, Breyer MD. Peroxisome proliferator-activated receptor δ activation promotes cell survival following hypertonic stress.
J Biol Chem
2002
;
277
:
21341
–5.
20
Di-Poi N, Tan NS, Michalik L, Wahli W, Desvergne B. Antiapoptotic role of PPARβ in keratinocytes via transcriptional control of the Akt1 signaling pathway.
Mol Cell
2002
;
10
:
721
–33.
21
Liou JY, Lee S, Ghelani D, Matijevic-Aleksic N, Wu KK. Protection of endothelial survival by peroxisome proliferator-activated receptor-δ mediated 14-3-3 upregulation.
Arterioscler Thromb Vasc Biol
2006
;
26
:
1481
–7.
22
Crawford LJ, Walker B, Ovaa H, et al. Comparative selectivity and specificity of the proteasome inhibitors BzLLLCOCHO, PS-341, and MG-132.
Cancer Res
2006
;
66
:
6379
–86.
23
Kim SH, Yoo CI, Kim HT, Park JY, Kwon CH, Kim YK. Activation of peroxisome proliferator-activated receptor-γ (PPARγ) induces cell death through MAPK-dependent mechanism in osteoblastic cells.
Toxicol Appl Pharmacol
2006
;
215
:
198
–207.
24
Shyue SK, Tsai MJ, Liou JY, Willerson JT, Wu KK. Selective augmentation of prostacyclin production by combined prostacyclin synthase and cyclooxygenase-1 gene transfer.
Circulation
2001
;
103
:
2090
–5.
25
Liou JY, Shyue SK, Tsai MJ, Chung CL, Chu KY, Wu KK. Colocalization of prostacyclin synthase with prostaglandin H synthase-1 (PGHS-1) but not phorbol ester-induced PGHS-2 in cultured endothelial cells.
J Biol Chem
2000
;
275
:
15314
–20.
26
Liou JY, Aleksic N, Chen SF, Han TJ, Shyue SK, Wu KK. Mitochondrial localization of cyclooxygenase-2 and calcium-independent phospholipase A2 in human cancer cells: implication in apoptosis resistance.
Exp Cell Res
2005
;
306
:
75
–84.
27
Blankson H, Grotterod EM, Seglen PO. Prevention of toxin-induced cytoskeletal disruption and apoptotic liver cell death by the grapefruit flavonoid, naringin.
Cell Death Differ
2000
;
7
:
739
–46.
28
Datta SR, Dudek H, Tao X, et al. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery.
Cell
1997
;
91
:
231
–41.
29
Zha J, Harada H, Yang E, Jockel J, Korsmeyer SJ. Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L).
Cell
1996
;
87
:
619
.
30
Fu H, Subramanian RR, Masters SC. 14-3-3 proteins: structure, function, and regulation.
Annu Rev Pharmacol Toxicol
2000
;
40
:
617
–47.
31
Tzivion G, Avruch J. 14-3-3 proteins: active cofactors in cellular regulation by serine/threonine phosphorylation.
J Biol Chem
2002
;
277
:
3061
–4.
32
Zhou XM, Liu Y, Payne G, Lutz RJ, Chittenden T. Growth factors inactivate the cell death promoter BAD by phosphorylation of its BH3 domain on Ser155.
J Biol Chem
2000
;
275
:
25046
–51.
33
Aberle H, Schwartz H, Kemler R. Cadherin-catenin complex: protein interactions and their implications for cadherin function.
J Cell Biochem
1996
;
61
:
514
–23.
34
Thompson WJ, Piazza GA, Li H, et al. Exisulind induction of apoptosis involves guanosine 3′,5′-cyclic monophosphate phosphodiesterase inhibition, protein kinase G activation, and attenuated β-catenin.
Cancer Res
2000
;
60
:
3338
–42.
35
Maier TJ, Janssen A, Schmidt R, Geisslinger G, Grosch S. Targeting the β-catenin/APC pathway: a novel mechanism to explain the cyclooxygenase-2-independent anticarcinogenic effects of celecoxib in human colon carcinoma cells.
FASEB J
2005
;
19
:
1353
–5.
36
Chiu CH, McEntee MF, Whelan J. Sulindac causes rapid regression of preexisting tumors in Min/+ mice independent of prostaglandin biosynthesis.
Cancer Res
1997
;
57
:
4267
–73.
37
Grosch S, Maier TJ, Schiffmann S, Geisslinger G. Cyclooxygenase-2 (COX-2)-independent anticarcinogenic effects of selective COX-2 inhibitors.
J Natl Cancer Inst
2006
;
98
:
736
–47.
38
Hanif R, Pittas A, Feng Y, et al. Effects of nonsteroidal anti-inflammatory drugs on proliferation and on induction of apoptosis in colon cancer cells by a prostaglandin-independent pathway.
Biochem Pharmacol
1996
;
52
:
237
–45.
39
Elder DJ, Halton DE, Hague A, Paraskeva C. Induction of apoptotic cell death in human colorectal carcinoma cell lines by a cyclooxygenase-2 (COX-2)-selective nonsteroidal anti-inflammatory drug: independence from COX-2 protein expression.
Clin Cancer Res
1997
;
3
:
1679
–83.
40
Castellone MD, Teramoto H, Williams BO, Druey KM, Gutkind JS. Prostaglandin E2 promotes colon cancer cell growth through a Gs-axin-β-catenin signaling axis.
Science
2005
;
310
:
1504
–9.
41
Lu D, Cottam HB, Corr M, Carson DA. Repression of β-catenin function in malignant cells by nonsteroidal antiinflammatory drugs.
Proc Natl Acad Sci U S A
2005
;
102
:
18567
–71.
42
Zhang L, Yu J, Park BH, Kinzler KW, Vogelstein B. Role of BAX in the apoptotic response to anticancer agents.
Science
2000
;
290
:
989
–92.
43
Richter M, Weiss M, Weinberger I, Furstenberger G, Marian B. Growth inhibition and induction of apoptosis in colorectal tumor cells by cyclooxygenase inhibitors.
Carcinogenesis
2001
;
22
:
17
–25.
44
Kohli M, Yu J, Seaman C, et al. SMAC/Diablo-dependent apoptosis induced by nonsteroidal antiinflammatory drugs (NSAIDs) in colon cancer cells.
Proc Natl Acad Sci U S A
2004
;
101
:
16897
–902.
45
Rice PL, Washington M, Schleman S, Beard KS, Driggers LJ, Ahnen DJ. Sulindac sulfide inhibits epidermal growth factor-induced phosphorylation of extracellular-regulated kinase 1/2 and Bad in human colon cancer cells.
Cancer Res
2003
;
63
:
616
–20.