The nonsteroidal anti-inflammatory drug tolfenamic acid has been shown to suppress cancer cell growth and tumorigenesis in different cancer models. However, the underlying mechanism by which tolfenamic acid exerts its antitumorigenic effect remains unclear. Previous data from our group and others indicate that tolfenamic acid alters expression of apoptosis- and cell-cycle arrest–related genes in colorectal cancer cells. Here, we show that tolfenamic acid markedly reduced the number of polyps and tumor load in APCmin/+ mice, accompanied with cyclin D1 downregulation in vitro and in vivo. Mechanistically, tolfenamic acid promotes endoplasmic reticulum (ER) stress, resulting in activation of the unfolded protein response (UPR) signaling pathway, of which PERK-mediated phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) induces the repression of cyclin D1 translation. Moreover, the PERK-eIF2α-ATF4 branch of the UPR pathway plays a role in tolfenamic acid-induced apoptosis in colorectal cancer cells, as silencing ATF4 attenuates tolfenamic acid-induced apoptosis. Taken together, these results suggest ER stress is involved in tolfenamic acid-induced inhibition of colorectal cancer cell growth, which could contribute to antitumorigenesis in a mouse model. Cancer Prev Res; 6(12); 1337–47. ©2013 AACR.

Colorectal cancer is the third-leading cause of cancer-related death in the United States (1). Taking nonsteroidal anti-inflammatory drugs (NSAID) has been associated with a reduced risk of colorectal tumorigenesis in epidemiologic studies (2), and a large amount of evidence from cell culture and animal studies has also shown that NSAID treatments can inhibit growth of cancer cells and tumors (3). Many mechanisms have been proposed to elucidate the effect of NSAIDs in antitumorigenesis (4). One such proposed mechanism is through the inhibition of COX-2, which is overexpressed in human tumors and plays a role in carcinogenesis. However, a COX-independent function of NSAIDs also plays an important role in antitumorigenesis and has received much attention since the inhibition of COX-2 activity produces adverse effects (5).

Tolfenamic acid is a conventional NSAID that has been long used for treatment of migraines. Compared with the other NSAIDs, tolfenamic acid exhibits fewer upper gastrointestinal side effects (6). Increasing evidence has shown that tolfenamic acid also suppresses tumorigenesis in several cancer models (7–12), and that this suppression seems to be independent of COX inhibition. For example, suppression of specificity proteins Sp1, Sp3, and Sp4, and their target genes, has been considered to contribute to tolfenamic acid-induced anticancer activity (7). Previous results from our lab also indicate that tolfenamic acid inhibits cell proliferation and induces apoptosis in human colorectal cancer cells, which is in part mediated by the induction of the tumor suppressor proteins NAG-1 and ATF3 (13, 14). These data suggest that tolfenamic acid could exert anticancer activity through various molecular mechanisms, and prompted us to further investigate the additional molecular mechanisms by which tolfenamic acid induces anticancer activity.

Cyclin D1, an oncogenic protein, is often overexpressed in various cancer cells and tumor tissues. The activated cyclin D1-CDK4/6 complex phosphorylates Rb protein and subsequently induces the expression of E2F-target genes that are necessary for DNA synthesis (15). In a recent study, cyclin D1 was also shown to play a role in DNA repair through binding to DNA repair proteins, such as BRCA2 and RAD51 (16), indicating an extra role of cyclin D1. A variety of compounds, including NSAIDs, have been documented to mediate cyclin D1 degradation and cell growth inhibition (17). It has been reported that tolfenamic acid also downregulated cyclin D1 expression in esophageal and breast cancer cells (8, 9); however, the detailed molecular mechanism(s) is not clear. Elucidation of the molecular mechanism(s) underlying cyclin D1 downregulation by tolfenamic acid would be beneficial to further understand the antitumorigenic activity of tolfenamic acid, and to develop novel derivatives, which could exert more pronounced anticancer effects.

Endoplasmic reticulum (ER) stress inhibits cyclin D1 translation and cell-cycle progression (18). Moreover, ER stress initiates apoptosis by activating transcription factor 4 (ATF4)-dependent C/EBP homologous transcription factor (CHOP), apoptosis signal-regulating kinase 1 (ASK1), and caspase-12 (19). Interestingly, two ER stress-inducible genes (EGR1 and ATF3) have been previously identified as the anticancer targets of tolfenamic acid in colon cancer (13, 14). Thus, ER stress could be involved in tolfenamic acid-mediated cancer cell growth suppression. Indeed, some NSAIDs like indomethacin and celecoxib have been shown to trigger ER stress response (20, 21). In contrast, diclofenac blocked ER stress-induced apoptosis in SH-SY5Y cells (22), and pranoprofen also suppressed ER stress-induced glucose-regulated protein 78 (GRP78) and CHOP expression (23). Therefore, how ER stress is involved in tolfenamic acid's effect on cell growth remains unclear and needs to be further elucidated.

In the current studies, we first evaluated the chemopreventive effect of tolfenamic acid in a mouse model of colorectal cancer. As a result, we found that tolfenamic acid inhibited polyp formation in APCmin/+ mice, along with a dramatic decrease in cyclin D1 expression in tumors. Consistently, we have also shown that tolfenamic acid downregulates cyclin D1 expression in different cancer cells in vitro, which is associated with the increase in Rb activity. We further investigated the underlying mechanism, showing that tolfenamic acid repressed cyclin D1 translation through activation of the ER stress-mediated unfolded protein responses (UPR) pathway. In addition, ER stress played a role in tolfenamic acid-induced apoptosis in colorectal cancer cells. Therefore, our data strongly suggest that ER stress response could contribute to the antitumorigenic activity of tolfenamic acid.

Reagents and antibodies

Tolfenamic acid, SC-560, and lactacystin were purchased from Cayman Chemical, and 5,5-dimethyl-3-(3-fluorophenyl)-4-(4-methylsulphonyl)phenyl-2(5H)-furanone (DFU) was described previously (24). All other NSAIDs, cycloheximide, and propidium iodide (PI) were purchased from Sigma-Aldrich. Epoxomicin was obtained from Calbiochem. All other chemicals were purchased from Fisher Scientific. RNase A was purchased from 5PRIME. Antibodies for cyclin D1, ATF4, Bcl-2, and Actin were from Santa Cruz Biotechnology; antibodies for pRb (Ser780), total Rb, p-Cyclin D1 (Thr286), BiP, CHOP, p-eIF2α, total eIF2α, PERK, and PARP were from Cell Signaling Technology; and antibody for HA tag was from GenScript. Control siRNA (#6201) and siRNA for PERK (#9024) were obtained from Cell Signaling Technology. siATF4 (sc-35112) was purchased from Santa Cruz Biotechnology.

Cell culture

Human cancer cell lines (HCT-116, HT-29, SW480, LoVo, Caco-2, A549, PC-3, and AsPC-1) were purchased from American Type Culture Collection (ATCC). ATCC tests the authenticity of these cell lines using short tandem repeat analyses. HCT-116 and HT-29 cells were maintained in McCoy's 5A medium (Bio Whittaker). SW480, A549, PC-3, and AsPC-1 cells were maintained in RPMI-1640 medium (Mediatech). Caco-2 and LoVo cells were kept in Eagle's minimum essential medium and Ham's F12 medium (HyClone), respectively. All cells were cultured in media supplemented with 10% FBS, 100 U/mL penicillin, and 100 mg/mL streptomycin in a 5% CO2 atmosphere at 37°C.

Plasmid, mutagenesis, and transient transfection

The pRcCMV-cyclin D1-HA plasmid was generously provided by Dr. E. Dmitrovsky (Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH; ref. 25). The pCMV-cyclin D1 and ubiquitin (Ub)-HA were gifts from Dr. Richard G. Pestell (Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA). Cyclin D1 transversion from threonine to alanine at the 286 amino acid position was generated using the QuickChange site-directed mutagenesis kit (Strategene) according to the manufacturer's protocol. ATF4 expression construct was purchased from OriGene, and p5xATF6-GL3 luciferase reporter construct (plasmid #11976) was obtained from Addgene (26). Transient transfection was carried out using TransIT-2020 transfection reagent (Mirus Bio LLC) according to the manufacturer's protocol. For luciferase assay, cells were seeded in 12-well plates at a density of 1.0 × 105 cells per well. After transfection with 1 μg p5xATF6-GL3 and pRL-null, cells were treated with dimethyl sulfoxide (DMSO) and tolfenamic acid for 24 hours. Cell lysates were harvested using 1XPassive lysis buffer (Promega), and then were subjected to luciferase activity analysis using DualGlo Luciferase Assay Kit (Promega).

Caspase-3/7 activity assay

Cells were seeded in a 96-well plate at a density of 1.0 × 104 cells per well. Empty vector and ATF4 expression vector were transiently transfected using TransIT-2020 transfection reagent (Mirus Bio LLC) for 24 hours. Caspase-3/7 activity was determined by caspase-Glo 3/7 reagent (Promega). Briefly, 100 μL reagent was added to each well. After incubating the plate at room temperature for 1 hour in the dark, luminescence was measured using a FLX-800 microplate reader (Bio-Tek). Fold change compared with empty vector transfection is represented as the mean ± SD of three wells.

Determination of sub-G1 cells

Cells were seeded in 6-well plates at a density of 3.0 × 105 cells per well in three replicates, and cultured to 60% to 80% confluence. Then, cells were treated with vehicle and tolfenamic acid (50 μmol/L) for 24 hours, then harvested and fixed in 70% ethanol. After being stored at −20°C overnight, the fixed cells were washed with PBS and stained with propidium iodide (PI, 70 μmol/L) solution containing RNase A (1 mg/mL) for 15 minutes at room temperature. The sub-G1 phase was determined by a Beckman Coulter Epixs XL flow cytometer equipped with ModFit LT software.

RNA interference

HCT-116 cells were seeded on 6-well plates at a density of 3.0 × 105 cells per well overnight. Control siRNA, siATF4, or siPERK was transfected at a final concentration of 100 nmol/L using PepMute siRNA & DNA Transfection Reagent (SignaGen) according to the manufacturer's instruction. After 24- (siATF4) or 48-hour (siPERK) transfection, cells were treated with vehicle and tolfenamic acid (50 μmol/L).

RNA isolation, semi-quantitative reverse transcription PCR, and real-time PCR

Total RNA of HCT-116 and SW480 cells treated by DMSO and tolfenamic acid were isolated by an E.Z.N.A Total RNA Kit (Omega Bio-Tek) according to the manufacturer's protocol. Then, RNA (1 μg) was reverse transcribed using a Verso cDNA synthesis Kit (ThermoScientific). PCR was performed using GoTaq Green Master Mix PCR Reaction Mixture (Promega) with primers for human cyclin D1, EGR-1, ATF3, XBP1, and GAPDH as follows: cyclin D1, forward 5′-ATGGAACACCAGCTCCTGTGCTGC-3′ and reverse 5′-TCAGATGTCCACGTCCCGCACGT-3′; EGR-1 forward 5′-CTGCGACATCTGTGGAAGAA-3′ and reverse 5′-TGTCCTGGGAGAAAAGGTTG-3′; ATF3: forward 5′-GTTTGAGGATTTTGCTAACCTGAC-3′, and reverse 5′-AGCTGCAATCTTATTTCTTTCTCGT-3′; XBP1: forward 5′-CCTTGTAGTTGAGAACCAGG-3′, and reverse 5′-GGGGCTTGGTATATATGTGG-3′; GAPDH, forward 5′-GGGCTGCTT TTAACTCTGGT-3′ and reverse 5′-TGGCAGGTTTTTCTAGACGG-3′. Real-Time PCR was performed using iTaq Univesal SYBR Green Supermix (Bio-Rad) with different primer sets for human cyclin D1, EGR-1, or GAPDH as follows: cyclin D1, forward 5′-GGCGGAGGAGAACAAACAGA-3′ and reverse 5′- TGTGAGGCGGTAGTAGGACA-3′; EGR-1 forward 5′-CACCTGACCGCAGAGTCTTT-3′ and reverse 5′-CTGACCAAGCTGAAGAGGGG-3′; GAPDH, forward 5′-GGGAGCCAAAAGGGTCATCA-3′ and reverse 5′-TGATGGCATGGACTGTGGTC-3′. Gene expression levels were calculated and GAPDH was used as a house-keeping gene, using MyiQ thermal cycler (Bio-Rad). The fold changes in mRNA levels were calculated using the ΔΔCt method.

Western blot analysis

Cells were washed with PBS and cell lysates harvested using radioimmunoprecipitation assay buffer (50 mmol/L Tris–HCl pH 7.4, 150 mmol/L NaCl, 1 mmol/L EDTA, 1% Triton X-100, 1% sodium deoxycholate, 0.1% SDS) supplemented with 1× protease inhibitor cocktail solution (Calbiochem) and phosphatase inhibitor (1 mmol/L Na3VO4, 1 mmol/L NaF), and centrifuged at 13,000 × g for 10 minutes at 4°C. Protein concentration was determined by the BCA protein assay (Pierce) using Bovine Serum Albumin as the standard. Protein (30 μg) was mixed with an equal amount of 2× SDS-PAGE sample loading buffer and boiled for 5 minutes. After separation by SDS-PAGE, the proteins were transferred to nitrocellulose membranes (Osmonics). The membranes were incubated with a specific primary antibody in TBS containing 0.05% Tween 20 (TSB-T) and 5% nonfat dry milk at 4°C overnight. After three washes with TBS-T, the blots were incubated with peroxidase-conjugated IgG for 1 hour at room temperature, visualized using ECL (Amersham Biosciences), and quantified by Scion Image Software (Scion Corp.).

Immunoprecipitation

The cells were harvested using lysis buffer (0.025 mol/L Tris, 0.15 mol/L NaCl, 0.001 mol/L EDTA, 1% NP40, 5% Glycerol, pH7.4) containing with 1× protease inhibitor cocktail solution (Calbiochem) and phosphatase inhibitor (1 mmol/L Na3VO4, 1 mmol/L NaF), and then kept on ice for 30 minutes. After being spun down for 10 minutes, the suspension was precleared using protein A/G PLUS-agarose (Santa Cruz Biotechnology) for 30 minutes at 4°C. Protein concentration was determined as described above. Protein lysates (1,000 μg) were incubated with 5 μg primary antibody and IgG control for 1 hour at 4°C, followed by adding 50 μL resuspended protein A/G PLUS-agarose overnight. Immunoprecipitates were collected by centrifuging at 1000 × g for 5 minutes at 4°C. After washing five times with lysis buffer, the pellets were resuspended with 50 μL 2× SDS-PAGE sample loading buffer. The samples were boiled for 5 minutes, and 20 μL of samples were subjected to Western blot analysis.

Animal study

The APCMin/+ mice (C57BL/6background) were purchased from Jackson Laboratory. To investigate the long-term effect of tolfenamic acid on tumor formation, mice aged 6 to 8 weeks were randomly assigned to three groups, and 0, 25, or 50 mg/kg of tolfenamic acid was given by oral gavage with 0.5% methylcellulose (vehicle) every 2 days for 4 weeks. To investigate the short-term effect of tolfenamic acid on tumor formation and protein expression, mice aged 16 to 18 weeks were randomly divided into two groups, and 0 or 50 mg/kg body weight of tolfenamic acid was given by oral gavage with 0.5% methylcellulose once per day for 3 days. At the end of the experiment, the mice were euthanized by overdose of CO2, and the entire intestinal tract was removed, flushed with cold saline, and opened longitudinally. The number and size (diameter) of polyps and tumors were scored blindly under a dissecting microscope as described previously (27, 28). Tumor load was calculated as number of tumors × average diameter. Both normal and neoplastic tumors were kept under RNAlater solution for molecular analysis. All animal research procedures were approved by the University of Tennessee Animal Care and Use Committee and were in accordance with NIH guidelines.

Statistical analysis

Statistical analysis was performed using the Student unpaired t test with statistical significance set at *, P < 0.5; **, P < 0.01; and ***, P < 0.001.

Tolfenamic acid suppressed polyp formation in an APCmin/+ mouse model

To evaluate the chemopreventive activity of tolfenamic acid in a mouse model, we examined the number of polyps and tumor load of APCmin/+ mice treated with vehicle and tolfenamic acid. In the first experiment, we treated mice aged 6 to 8 weeks with tolfenamic acid for 4 weeks. As shown in Fig. 1A, tolfenamic acid caused a striking reduction in the total number of polyps and tumor load in a dose-dependent manner. Next, we treated mice at 16 to 18 weeks of age with tolfenamic acid for a short period (3 days) to get a tumor sample and observe the short-term effect of tolfenamic acid treatment on tumorigenicity. As shown in Fig. 1B, tolfenamic acid still significantly reduced the number of polyps and tumor load, compared with the control group. Many genes, including β-catenin and Smad, had altered expression in tissue samples (data not shown). However, the most interesting and consistent gene alteration was cyclin D1 (Fig. 1C). Both cyclin D1 and COX-2 were overexpressed in tumor tissues, which is in agreement with a previous report (29); however, only cyclin D1 was dramatically suppressed in tolfenamic acid-treated tumor samples.

Figure 1.

Tolfenamic acid suppresses colorectal tumorigenesis in a mouse model. A, mice aged 6–8 weeks were randomly divided into three groups, and respectively administered 0.5% methylcellulose, 25 mg/kg body weight, or 50 mg/kg body weight of tolfenamic acid (TA) every other day for 4 weeks (total 14 times). The number of polyps and tumor load were calculated as described in Materials and Methods. Data indicate mean ± SD of all experiments. Results were considered statistically significant at ***, P < 0.001, compared to group 1. B, mice aged 16–18 weeks were randomly divided into two groups, and respectively treated with 0.5% methylcellulose or 50 mg/kg body weight tolfenamic acid once per day for 3 days (total three times). The number of polyps and tumor load were calculated, and the data are expressed as mean ± SD of all experiments. Results were considered statistically significant at **, P < 0.01, compared to group 1. C, proteins prepared from tissues of mice at age 16–18 weeks fed vehicle or 50 mg/kg body weight tolfenamic acid were subjected to Western blot analysis for COX-2, cyclin D1, and actin expression.

Figure 1.

Tolfenamic acid suppresses colorectal tumorigenesis in a mouse model. A, mice aged 6–8 weeks were randomly divided into three groups, and respectively administered 0.5% methylcellulose, 25 mg/kg body weight, or 50 mg/kg body weight of tolfenamic acid (TA) every other day for 4 weeks (total 14 times). The number of polyps and tumor load were calculated as described in Materials and Methods. Data indicate mean ± SD of all experiments. Results were considered statistically significant at ***, P < 0.001, compared to group 1. B, mice aged 16–18 weeks were randomly divided into two groups, and respectively treated with 0.5% methylcellulose or 50 mg/kg body weight tolfenamic acid once per day for 3 days (total three times). The number of polyps and tumor load were calculated, and the data are expressed as mean ± SD of all experiments. Results were considered statistically significant at **, P < 0.01, compared to group 1. C, proteins prepared from tissues of mice at age 16–18 weeks fed vehicle or 50 mg/kg body weight tolfenamic acid were subjected to Western blot analysis for COX-2, cyclin D1, and actin expression.

Close modal

Tolfenamic acid downregulated cyclin D1 expression in cancer cells

As cyclin D1 acts as a pro-oncogenic factor, it is not surprising that colorectal cancer cells harbor overexpressed cyclin D1 (Fig. 2A), and its downregulation may contribute to the antiproliferative effect of NSAIDs. As it has been documented that NSAIDs differ in their ability to suppress cyclin D1 expression or cell proliferation (30), we treated SW480 cells for 24 hours with the same dose (50 μmol/L) of different NSAIDs: conventional (diclofenac, ibuprofen, aspirin, naproxen, piroxicam, tolfenamic acid), COX-2 selective (SC-236, DFU), or COX-1 selective (SC-560). Tolfenamic acid and SC-560 completely decreased cyclin D1 expression, suggesting both of them were the most potent agents of those we tested with respect to cyclin D1 downregulation (Fig. 2B). As we have shown that tolfenamic acid can suppress cyclin D1 in an in vivo model (Fig. 1), we selected this compound for subsequent studies. Tolfenamic acid caused rapid cyclin D1 downregulation in a dose- and time-dependent manner, and the decrease occurred within 1 hour (Fig. 2C). To further evaluate the effect of tolfenamic acid on other cancer cells, we treated colorectal cancer cells (HCT-116 and Caco-2), prostate cancer cells (PC-3), pancreatic cancer cells (AsPC-1), and lung cancer cells (A549) with tolfenamic acid. As shown in Fig. 2D, tolfenamic acid dose-dependently downregulated cyclin D1 expression in different cancer cells.

Figure 2.

Tolfenamic acid downregulates cyclin D1 in cancer cells. A, endogenous cyclin D1 expression in different colorectal cancer cells. Western blot analysis was carried out for analyzing expression of cyclin D1 and actin in the indicated cell lines. B, SW480 cells were treated with various NSAIDs at 50 μmol/L for 24 hours. The cell lysates were harvested and subjected to Western blot analysis for cyclin D1 and actin. C, SW480 cells were treated with 0, 5, 10, 25, or 50 μmol/L tolfenamic acid for 24 hours (left) and 50 μmol/L tolfenamic acid for 0, 1, 3, 6, 12, or 24 hours (right). Cell lysates were analyzed by Western blot using cyclin D1 and actin antibodies. D, colorectal (HCT-116, Caco-2), prostate (PC-3), pancreatic (AsPC-1), and lung (A549) cancer cells were treated with 0, 5, 25, or 50 μmol/L of tolfenamic acid for 24 hours. Western blot analysis was carried out for analyzing cyclin D1 and actin.

Figure 2.

Tolfenamic acid downregulates cyclin D1 in cancer cells. A, endogenous cyclin D1 expression in different colorectal cancer cells. Western blot analysis was carried out for analyzing expression of cyclin D1 and actin in the indicated cell lines. B, SW480 cells were treated with various NSAIDs at 50 μmol/L for 24 hours. The cell lysates were harvested and subjected to Western blot analysis for cyclin D1 and actin. C, SW480 cells were treated with 0, 5, 10, 25, or 50 μmol/L tolfenamic acid for 24 hours (left) and 50 μmol/L tolfenamic acid for 0, 1, 3, 6, 12, or 24 hours (right). Cell lysates were analyzed by Western blot using cyclin D1 and actin antibodies. D, colorectal (HCT-116, Caco-2), prostate (PC-3), pancreatic (AsPC-1), and lung (A549) cancer cells were treated with 0, 5, 25, or 50 μmol/L of tolfenamic acid for 24 hours. Western blot analysis was carried out for analyzing cyclin D1 and actin.

Close modal

Tolfenamic acid affected phosphorylation of Rb protein

The cyclin D1-CDK4/6 complex phosphorylates and inactivates Rb, facilitating the G1–S phase transition. Specifically, cyclin D1 is required for phosphorylating the Ser780 amino acid position of Rb (31). We observed that tolfenamic acid decreased phosphorylated Rb at Ser780 in both SW480 and HCT-116 cells (Fig. 3A). To further investigate whether cyclin D1 plays a role in the loss of Rb phosphorylation by tolfenamic acid, we transiently transfected cyclin D1 expression vector containing an alanine for threonine substitution at codon 286 (cyclin D1-HA [T286A]), which is more stable than the wild-type form (32). Overexpression of the stable form of cyclin D1 restored phosphorylated Rb at Ser780, suggesting that cyclin D1 downregulation by tolfenamic acid, at least in part, facilitates dephosphorylation of Rb at Ser780 (Fig. 3B).

Figure 3.

Tolfenamic acid (TA) activates Rb via dephosphorylation. A, HCT-116 and SW480 cells were treated with 50 μmol/L tolfenamic acid for 0, 1, 3, 6, 12, or 24 hours. Cell lysates were harvested and analyzed by Western blot for phospho-Rb (Ser780) and total Rb. B, HCT-116 cells were transiently transfected with Mock and cyclin D1-HA (T286A) for 24 hours, followed by treatment with 50 μmol/L tolfenamic acid for 0, 6, 12, or 24 hours. Western blot analysis was performed for phosphor-Rb (Ser780), total Rb, HA, and actin.

Figure 3.

Tolfenamic acid (TA) activates Rb via dephosphorylation. A, HCT-116 and SW480 cells were treated with 50 μmol/L tolfenamic acid for 0, 1, 3, 6, 12, or 24 hours. Cell lysates were harvested and analyzed by Western blot for phospho-Rb (Ser780) and total Rb. B, HCT-116 cells were transiently transfected with Mock and cyclin D1-HA (T286A) for 24 hours, followed by treatment with 50 μmol/L tolfenamic acid for 0, 6, 12, or 24 hours. Western blot analysis was performed for phosphor-Rb (Ser780), total Rb, HA, and actin.

Close modal

Tolfenamic acid inhibited cyclin D1 at the translational regulation in colorectal cancer cells

To elucidate the molecular mechanism by which tolfenamic acid downregulated cyclin D1 expression in colorectal cancer cells, we treated HCT-116 and SW480 cells with tolfenamic acid for 1 hour and examined the expression of cyclin D1 transcripts. As shown in Fig. 4A, cyclin D1 transcript levels were not altered by tolfenamic acid within 1 hour, although there was a dramatic increase in EGR-1 and ATF3 mRNA expression as a control (13, 14). Given cyclin D1 is a short-lived protein, we examined whether tolfenamic acid triggered the cyclin D1 degradation. Results in Fig. 4B and C indicate that neither endogenous nor exogenous cyclin D1 stability was affected by tolfenamic acid. Furthermore, treatment of inhibitors responsible for cyclin D1 degradation did not restore cyclin D1 expression in the presence of tolfenamic acid (Fig. 4D and Supplementary Fig. S1). In addition, tolfenamic acid did not affect Thr286 phosphorylation nor ubiquitination of cyclin D1, which has been well documented to be associated with cyclin D1 degradation (Fig. 4E and Supplementary Fig. S2). Taking these results together, we found that tolfenamic acid did not affect transcription nor protein degradation pathway of cyclin D1; rather tolfenamic acid may inhibit cyclin D1 translation initiation.

Figure 4.

Tolfenamic acid suppresses cyclin D1 translation. A, HCT-116 and SW480 cells were treated with 0, 5, 25, or 50 μmol/L of tolfenamic acid for 1 hour. Semi-quantitative RT-PCR was carried out for analyzing cyclin D1, EGR-1, ATF3, and GAPDH mRNA expression using specific primers as described in Materials and Methods. Real-time PCR was also performed as described in Materials and Methods (right). B, SW480 cells were treated with 0, 10, or 25 μmol/L of tolfenamic acid for 1 hour, followed by incubation with 10 μg/mL cycloheximide (CHX) for the indicated times. Cell lysates were subjected to Western blot using cyclin D1 and actin antibodies. C, cyclin D1-HA construct was transiently transfected into HCT-116 cells along with GFP expression vector serving as a transfection control for 24 hours. Cells were treated with DMSO and 50 μmol/L tolfenamic acid for 1 hours, followed by incubation with 10 μg/mL cycloheximide (CHX) for the indicated times. Cell lysates were analyzed using HA (cyclin D1) and GFP antibodies. D, SW480 cells were treated with DMSO, lactacystin (10 μmol/L), epoxomicin (1 μmol/L), NH4Cl (20 mmol/L), z-VAD-fmk (50 μmol/L), calpeptin (50 μmol/L), leupeptin (200 μmol/L), KU55933 (10 μmol/L), or staurosporine (2 μmol/L) for 1 hour, followed by incubation with 50 μmol/L of tolfenamic acid for 1 hour, as indicated. Cell lysates were analyzed for cyclin D1 and actin using Western blot analysis. E, the pCMV-cyclin D1 and Ub-HA plasmids were cotransfected into HCT-116 cells for 24 hours. Cell lysates were immunoprecipitated using either cyclin D1 antibody (IP: cyclin D1) or nonspecific IgG antibody (IP: IgG). Cyclin D1 ubiquitination, phosphorylation of cyclin D1, and total amount of cyclin D1 were determined with the indicated antibody.

Figure 4.

Tolfenamic acid suppresses cyclin D1 translation. A, HCT-116 and SW480 cells were treated with 0, 5, 25, or 50 μmol/L of tolfenamic acid for 1 hour. Semi-quantitative RT-PCR was carried out for analyzing cyclin D1, EGR-1, ATF3, and GAPDH mRNA expression using specific primers as described in Materials and Methods. Real-time PCR was also performed as described in Materials and Methods (right). B, SW480 cells were treated with 0, 10, or 25 μmol/L of tolfenamic acid for 1 hour, followed by incubation with 10 μg/mL cycloheximide (CHX) for the indicated times. Cell lysates were subjected to Western blot using cyclin D1 and actin antibodies. C, cyclin D1-HA construct was transiently transfected into HCT-116 cells along with GFP expression vector serving as a transfection control for 24 hours. Cells were treated with DMSO and 50 μmol/L tolfenamic acid for 1 hours, followed by incubation with 10 μg/mL cycloheximide (CHX) for the indicated times. Cell lysates were analyzed using HA (cyclin D1) and GFP antibodies. D, SW480 cells were treated with DMSO, lactacystin (10 μmol/L), epoxomicin (1 μmol/L), NH4Cl (20 mmol/L), z-VAD-fmk (50 μmol/L), calpeptin (50 μmol/L), leupeptin (200 μmol/L), KU55933 (10 μmol/L), or staurosporine (2 μmol/L) for 1 hour, followed by incubation with 50 μmol/L of tolfenamic acid for 1 hour, as indicated. Cell lysates were analyzed for cyclin D1 and actin using Western blot analysis. E, the pCMV-cyclin D1 and Ub-HA plasmids were cotransfected into HCT-116 cells for 24 hours. Cell lysates were immunoprecipitated using either cyclin D1 antibody (IP: cyclin D1) or nonspecific IgG antibody (IP: IgG). Cyclin D1 ubiquitination, phosphorylation of cyclin D1, and total amount of cyclin D1 were determined with the indicated antibody.

Close modal

Tolfenamic acid promoted ER stress response

To further understand the cellular signaling pathways involved in tolfenamic acid-mediated cyclin D1 translation, we investigated the effect of tolfenamic acid on the mTOR pathway, which is mainly responsible for protein translation. However, we did not observe that tolfenamic acid suppressed this pathway (data not shown). Another important protein translation control is mediated by eIF2α, phosphorylation of which can lead to nearly global translation inhibition. As ER stress usually induces phosphorylated eIF2α, we therefore asked whether tolfenamic acid promoted ER stress in colorectal cancer cells. The ER stress-activated UPR pathway consists of a large variety of downstream responses, including ATF6-mediated transcriptional activation, splicing of x-box-binding protein-1(XBP-1) mRNA, and induction of target gene expression, such as BiP, ATF4, and CHOP (33). As expected, treatment of HCT-116 and SW480 cells with tolfenamic acid resulted in the induction of ATF6 transcriptional activity, XBP-1 mRNA splicing, and expression of specific genes (BiP, ATF4, and CHOP) in a time-dependent manner (Fig. 5A–C), indicating tolfenamic acid could be a potential activator of ER stress.

Figure 5.

Tolfenamic acid promotes ER stress response. A, HCT-116 and SW480 cells were transiently transfected with p5×ATF6-GL3 and pRL-null for 24 hours, followed by treatment with 0, 5, 25, or 50 μmol/L of tolfenamic acid for 24 hours. Luciferase activity was measured as described in Materials and Methods. RLU, relative luciferase unit. Results were considered statistically significant at *, P < 0.5; **, P < 0.01; and ***, P < 0.001, compared with DMSO. B, HCT-116 and SW480 cells were treated with 0, 5, 25, or 50 μmol/L of tolfenamic acid for 24 hours. Reverse transcription-PCR was carried out for analyzing XBP-1 and GAPDH mRNA expression using specific primers as described in Materials and Methods. C, HCT-116 and SW480 cells were treated with 50 μmol/L of tolfenamic acid for 0, 1, 3, 6, 12, or 24 hours. The cell lysates were subjected to Western blot analysis for analyzing BiP, ATF4, CHOP, and actin expression.

Figure 5.

Tolfenamic acid promotes ER stress response. A, HCT-116 and SW480 cells were transiently transfected with p5×ATF6-GL3 and pRL-null for 24 hours, followed by treatment with 0, 5, 25, or 50 μmol/L of tolfenamic acid for 24 hours. Luciferase activity was measured as described in Materials and Methods. RLU, relative luciferase unit. Results were considered statistically significant at *, P < 0.5; **, P < 0.01; and ***, P < 0.001, compared with DMSO. B, HCT-116 and SW480 cells were treated with 0, 5, 25, or 50 μmol/L of tolfenamic acid for 24 hours. Reverse transcription-PCR was carried out for analyzing XBP-1 and GAPDH mRNA expression using specific primers as described in Materials and Methods. C, HCT-116 and SW480 cells were treated with 50 μmol/L of tolfenamic acid for 0, 1, 3, 6, 12, or 24 hours. The cell lysates were subjected to Western blot analysis for analyzing BiP, ATF4, CHOP, and actin expression.

Close modal

Cyclin D1 suppression and ATF4 induction by tolfenamic acid are mediated by PERK-eIF2α pathway

Given that ER stress would activate the kinase PERK that resides in the ER lumen, resulting in the phosphorylation of eIF2α and subsequently the repression of cyclin D1 translation (18), we examined the relationship of PERK/eIF2α signaling and cyclin D1 expression in the presence of tolfenamic acid. As shown in Fig. 6A, tolfenamic acid caused rapid phosphorylation of eIF2α coupled with a dramatic decrease of cyclin D1 expression in a time-dependent manner. In addition, thapsigargin, a well-known activator of ER stress had an effect on cyclin D1 expression similar to that of tolfenamic acid (Fig. S3). Moreover, silence of PERK using specific siRNA slightly restored cyclin D1 expression in the presence of tolfenamic acid (Fig. 6B), suggesting the PERK-eIF2α pathway was involved in tolfenamic acid-mediated cyclin D1 downregulation. The UPR is a prosurvival response to allow cells to remove unfolded proteins and re-establish homeostasis. However, if the primary stimuli causing unfolded proteins are persistent and the stress cannot be resolved, signaling switches from prosurvival to proapoptosis (34). Of the UPR signaling pathway branches, PERK-eIF2α-ATF4 upregulates CHOP expression, which subsequently induces apoptosis in part through downregulating BCL-2 gene expression (19). As shown in Fig. 6B, tolfenamic acid-induced CHOP expression is mediated by the PERK-eIF2α-ATF4 axis. Consistent with CHOP upregulation, tolfenamic acid also downregulated Bcl-2 expression accompanied with the induction of apoptosis as measured by cleaved PARP and the percentage of sub-G1 fraction cells (Fig. 6C). In addition, overexpression of ATF4 increased caspase-3/7 activity, supporting the previously documented proapoptotic role of ATF4 in ER stress (35, 36; Fig. 6D). Finally, knockout of ATF4 using specific siRNA blocked tolfenamic acid-induced CHOP expression and subsequent Bcl-2 downregulation, and attenuated apoptosis as measured by cleaved PARP (Fig. 6E). These data indicated tolfenamic acid caused cell apoptosis via, at least in part, the PERK-eIF2α-ATF4 pathway in colorectal cancer cells.

Figure 6.

Tolfenamic acid affects cyclin D1 and ATF4 expression via PERK-eIF2α axis. A, HCT-116 cells were treated with 50 μmol/L tolfenamic acid for the indicated times. Western blot analysis was performed for analysis of phosphorylated eIF2α, total eIF2α, cyclin D1, and actin expression. B, knockout of PERK was performed as described in Materials and Methods in HCT-116 cells, followed by incubating with 50 μmol/L tolfenamic acid for 24 hours, as indicated. Cell lysates were analyzed for PERK, Cyclin D1, ATF4, CHOP, and actin expression. C, HCT-116 cells were treated with 50 μmol/L tolfenamic acid for the indicated times. Western blot analysis was carried out using Bcl-2, PARP, and actin antibodies. The proportion of the sub-G1 phase of cells treated with 50 μmol/L tolfenamic acid for 24 hours was determined by flow cytometric analysis as described in Materials and Methods. The data are expressed as mean ± SD of three replicates. Results were considered statistically significant at **, P < 0.01, compared with DMSO. D, The ATF4 construct was transfected into HCT-116 cells. Transfection efficiency was examined by Western blot using ATF4 antibody. Caspase-3/7 activity was measured as described in Materials and Methods, and the data are expressed as mean ± SD of three wells. Results were considered statistically significant at **, P < 0.01, compared with EV. E, HCT-116 cells were transfected with siRNA against ATF4 for 24 h, followed by treatment with 50 μmol/L tolfenamic acid for 24 hours, as indicated. Cell lysates were subjected to Western blot analysis using ATF4, CHOP, Bcl-2, PARP, and actin antibodies.

Figure 6.

Tolfenamic acid affects cyclin D1 and ATF4 expression via PERK-eIF2α axis. A, HCT-116 cells were treated with 50 μmol/L tolfenamic acid for the indicated times. Western blot analysis was performed for analysis of phosphorylated eIF2α, total eIF2α, cyclin D1, and actin expression. B, knockout of PERK was performed as described in Materials and Methods in HCT-116 cells, followed by incubating with 50 μmol/L tolfenamic acid for 24 hours, as indicated. Cell lysates were analyzed for PERK, Cyclin D1, ATF4, CHOP, and actin expression. C, HCT-116 cells were treated with 50 μmol/L tolfenamic acid for the indicated times. Western blot analysis was carried out using Bcl-2, PARP, and actin antibodies. The proportion of the sub-G1 phase of cells treated with 50 μmol/L tolfenamic acid for 24 hours was determined by flow cytometric analysis as described in Materials and Methods. The data are expressed as mean ± SD of three replicates. Results were considered statistically significant at **, P < 0.01, compared with DMSO. D, The ATF4 construct was transfected into HCT-116 cells. Transfection efficiency was examined by Western blot using ATF4 antibody. Caspase-3/7 activity was measured as described in Materials and Methods, and the data are expressed as mean ± SD of three wells. Results were considered statistically significant at **, P < 0.01, compared with EV. E, HCT-116 cells were transfected with siRNA against ATF4 for 24 h, followed by treatment with 50 μmol/L tolfenamic acid for 24 hours, as indicated. Cell lysates were subjected to Western blot analysis using ATF4, CHOP, Bcl-2, PARP, and actin antibodies.

Close modal

We and others have previously reported that tolfenamic acid exhibits anticancer activity via different molecular targets, including Sps (7, 37), ATF2/ATF3 (14), NAG-1 (13), NF-κB (38), and Smads (39). In the present study, we further confirmed that tolfenamic acid is a powerful chemopreventive or therapeutic agent against colorectal cancer in vitro and in vivo, and that tolfenamic acid-mediated growth inhibition might be associated with ER stress response, especially the PERK-eIF2α axis, which resulted in downregulation of prosurvival proteins like cyclin D1, and upregulation of proapoptotic proteins like CHOP. In addition, we found that tolfenamic acid markedly downregulated cyclin D1 in tumors and a large variety of cell lines, and that this downregulation partially contributed to the loss of phosphorylated Rb. Given the key role of cyclin D1 in G1–S phase transition, downregulation of cyclin D1 by tolfenamic acid could induce cell-cycle arrest. Indeed, there are several reports showing that treatment with tolfenamic acid results in G0–G1 cell-cycle arrest (11, 40, 41). On the other hand, we also provided direct evidence that tolfenamic acid induced apoptosis via activation of the PERK-eIF2α-ATF4 pathway.

It has been suggested that tolfenamic acid suppresses erbB2 and Sp transcription factors, which could result in the suppression of cyclin D1 expression (8, 9). However, our data clearly indicate that tolfenamic acid-induced cyclin D1 downregulation is an erbB2- and Sp-independent pathway; although tolfenamic acid did not affect the cyclin D1 transcription, it significantly decreased protein expression (Figs. 3 and 4A), and tolfenamic acid-induced cyclin D1 downregulation occurred much earlier than tolfenamic acid-mediated Sp downregulation (1 vs. 24 hours) (unpublished data). These results further indicate that tolfenamic acid-mediated cyclin D1 downregulation is independent of transcription factors such as Sp1 and erbB2, and even other transcription factors β-catenin or NF-κB.

Interestingly, we found that cyclin D1 degradation was not enhanced in the presence of tolfenamic acid (Fig. 4B–E). On the basis of our data, we suggest that tolfenamic acid could be able to suppress cyclin D1 translation, at least in colorectal cancer cells. Tolfenamic acid triggered an ER stress response, resulting in the phosphorylation of eIF2α, which leads to the repression of protein translation through limiting the delivery of initiator met-tRNAiMet to translation machinery (42). After exposure of cells to ER stress, PERK is mainly responsible for phosphorylating eIF2α, which mediates cyclin D1 translation repression (43). Indeed, silencing PERK in HCT-116 cells partially restored cyclin D1 expression in the presence of tolfenamic acid. Because we could not completely block the expression of PERK (Fig. 6B), it is likely that remaining PERK activity contributed to the loss of cyclin D1 in HCT-116 cells treated with tolfenamic acid. In addition, it is possible that other kinases would be able to phosphorylate eIF2α in the absence of PERK, resulting in cyclin D1 repression (43). Therefore, ER stress-sensing pathways are very complicated, and the detail pathways need to be elucidated.

Accumulating evidence suggests that the activation of UPR exerts contradictory consequences to tumor development. There is some evidence that UPR signaling contributes to tumor growth through increasing tumor cell tolerance to hypoxia (44, 45). On the other hand, various compounds harboring anticancer activity have been shown to induce ER stress-mediated cell-cycle arrest and apoptosis (46–49). Our data indicate that tolfenamic acid exerts anticancer activity, in part dependent on the activation of the UPR signaling pathway. Similar findings were previously reported in cells treated with the COX-2 selective inhibitor celecoxib and its analog (21, 50), suggesting tolfenamic acid and celecoxib might exhibit comparable antineoplastic activity in a COX-independent manner.

We have identified EGR1 and ATF3 as mediators of tolfenamic acid-induced apoptosis in colorectal cancer cells (13, 14). Interestingly, both of them are inducible under ER stress (51, 52). Tolfenamic acid might transcriptionally upregulate expression of those two proapoptotic genes through regulation of ATF4 and CHOP, thereby causing apoptosis. In addition, tolfenamic acid also activated ER stress-associated kinases (JNK) in colorectal cancer cells, which would trigger apoptosis as well (14). Although Sp transcriptional factors played an important role in tolfenamic acid-mediated cell growth inhibition, that specific inhibition is likely to occur at a later stage. Here, we provided evidence that ER stress response was an early event of treatment with tolfenamic acid, whereas Sp downregulation is later event affected by tolfenamic acid in antitumorigenesis.

We also reported here that tolfenamic acid dramatically reduced the intestinal tumor load and polyp number in an APCMin/+ mouse model. Although we treated older mice with tolfenamic acid for only 3 days, the number of polyps and tumor load were still significantly decreased. The results described here extend the involvement of cyclin D1 to the development of benign lesions in the APCMin/+ model of intestinal neoplasia. The potential relevance of our results to human familial and spontaneous colon cancer is provocative. Cyclin D1 overexpression has been demonstrated in adenomas from familial adenomatous polyposis patients (53), and more than 60% of spontaneous human colonic adenocarcinomas exhibit APC mutations (54). Our data are consistent with the recent report that tolfenamic acid exhibits anticancer activity, as assessed by nude mice experiments (37). Along with the fact that each NSAID exhibits distinctive action in anticancer activity, tolfenamic acid has great potential to be translated to the clinic for chemopreventive or therapeutic intervention of cancer. Moreover, our findings suggest that tolfenamic acid would be a very powerful and effective chemopreventive agent for colorectal cancer development, and inactivation of cyclin D1 would provide one mechanism to support tolfenamic acid effects on chemoprevention.

In conclusion, the current study provides further evidence for the antitumorigenic effect of tolfenamic acid on colorectal cancer and elucidates the underlying molecular mechanisms of its action. Together with previous reports, our results suggest tolfenamic acid would be a promising chemopreventive agent for cancer. Novel tolfenamic acid derivatives that show similar safety profiles and enhanced anticancer effects would be attractive in ongoing investigations.

No potential conflicts of interest were disclosed

Conception and design: X. Zhang, S.-H. Lee, K.-W. Min, S.J. Baek

Development of methodology: M.F. McEntee, S.J. Baek

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M.F. McEntee

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): X. Zhang, S.-H. Lee, M.F. McEntee, S.J. Baek

Writing, review, and/or revision of the manuscript: X. Zhang, K.-W. Min, M.F. McEntee, S.J. Baek

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): K.-W. Min, M.F. McEntee, J. Jeong, S.J. Baek

Study supervision: Q. Li, S.J. Baek

The authors thank Misty Bailey for her critical reading of this manuscript.

This work was supported by The University of Tennessee Center of Excellence in Livestock Diseases and Human Health grant (to S.J. Baek), NIH grant R01CA108975 (to S.J. Baek), American Cancer Society grant (RSG-11-133-01-CCE; to S.-H. Lee), and a grant from the Ministry of Agriculture of the People's Republic of China (2011ZX08008-003; to Q. Li). Financial support for X. Zhang was provided by the Program in Organizational or Personal Cooperation with Foreign Counterparts (2010630161), China Scholarship Council, China.

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.
Jemal
A
,
Siegel
R
,
Ward
E
,
Hao
Y
,
Xu
J
,
Thun
MJ
. 
Cancer statistics, 2009
.
CA Cancer J Clin
2009
;
59
:
225
49
.
2.
Kraus
S
,
Arber
N
. 
Cancer: do aspirin and other NSAIDs protect against colorectal cancer?
Nat Rev Gastroenterol Hepatol
2011
;
8
:
125
6
.
3.
Mann
JR
,
Backlund
MG
,
DuBois
RN
. 
Mechanisms of disease: inflammatory mediators and cancer prevention
.
Nat Clin Pract Oncol
2005
;
2
:
202
10
.
4.
Wang
X
,
Baek
SJ
,
Eling
T
. 
COX inhibitors directly alter gene expression: role in cancer prevention?
Cancer Metastasis Rev
2011
;
30
:
641
57
.
5.
Jones
MK
,
Wang
H
,
Peskar
BM
,
Levin
E
,
Itani
RM
,
Sarfeh
IJ
, et al
Inhibition of angiogenesis by nonsteroidal anti-inflammatory drugs: insight into mechanisms and implications for cancer growth and ulcer healing
.
Nat Med
1999
;
5
:
1418
23
.
6.
Hansen
PE
. 
Tolfenamic acid in acute and prophylactic treatment of migraine: a review
.
Pharmacol Toxicol
1994
;
75
:
81
2
.
7.
Abdelrahim
M
,
Baker
CH
,
Abbruzzese
JL
,
Safe
S
. 
Tolfenamic acid and pancreatic cancer growth, angiogenesis, and Sp protein degradation
.
J Natl Cancer Inst
2006
;
98
:
855
68
.
8.
Papineni
S
,
Chintharlapalli
S
,
Abdelrahim
M
,
Lee
SO
,
Burghardt
R
,
Abudayyeh
A
, et al
Tolfenamic acid inhibits esophageal cancer through repression of specificity proteins and c-Met
.
Carcinogenesis
2009
;
30
:
1193
201
.
9.
Liu
X
,
Abdelrahim
M
,
Abudayyeh
A
,
Lei
P
,
Safe
S
. 
The nonsteroidal anti-inflammatory drug tolfenamic acid inhibits BT474 and SKBR3 breast cancer cell and tumor growth by repressing erbB2 expression
.
Mol Cancer Ther
2009
;
8
:
1207
17
.
10.
Colon
J
,
Basha
MR
,
Madero-Visbal
R
,
Konduri
S
,
Baker
CH
,
Herrera
LJ
, et al
Tolfenamic acid decreases c-Met expression through Sp proteins degradation and inhibits lung cancer cells growth and tumor formation in orthotopic mice
.
Invest New Drug
2011
;
29
:
41
51
.
11.
Sankpal
UT
,
Abdelrahim
M
,
Connelly
SF
,
Lee
CM
,
Madero-Visbal
R
,
Colon
J
, et al
Small molecule tolfenamic acid inhibits PC-3 cell proliferation and invasion in vitro, and tumor growth in orthotopic mouse model for prostate cancer
.
Prostate
2012
;
72
:
1648
58
.
12.
Kang
SU
,
Shin
YS
,
Hwang
HS
,
Baek
SJ
,
Lee
SH
,
Kim
CH
. 
Tolfenamic acid induces apoptosis and growth inhibition in head and neck cancer: involvement of NAG-1 expression
.
PLoS ONE
2012
;
7
:
e34988
.
13.
Lee
SH
,
Bahn
JH
,
Choi
CK
,
Whitlock
NC
,
English
AE
,
Safe
S
, et al
ESE-1/EGR-1 pathway plays a role in tolfenamic acid-induced apoptosis in colorectal cancer cells
.
Mol Cancer Ther
2008
;
7
:
3739
50
.
14.
Lee
SH
,
Bahn
JH
,
Whitlock
NC
,
Baek
SJ
. 
Activating transcription factor 2 (ATF2) controls tolfenamic acid-induced ATF3 expression via MAP kinase pathways
.
Oncogene
2010
;
29
:
5182
92
.
15.
Musgrove
EA
,
Caldon
CE
,
Barraclough
J
,
Stone
A
,
Sutherland
RL
. 
Cyclin D as a therapeutic target in cancer
.
Nat Rev Cancer
2011
;
11
:
558
72
.
16.
Jirawatnotai
S
,
Hu
Y
,
Michowski
W
,
Elias
JE
,
Becks
L
,
Bienvenu
F
, et al
A function for cyclin D1 in DNA repair uncovered by protein interactome analyses in human cancers
.
Nature
2011
;
474
:
230
4
.
17.
Alao
JP
. 
The regulation of cyclin D1 degradation: roles in cancer development and the potential for therapeutic invention
.
Mol Cancer
2007
;
6
:
24
.
18.
Brewer
JW
,
Hendershot
LM
,
Sherr
CJ
,
Diehl
JA
. 
Mammalian unfolded protein response inhibits cyclin D1 translation and cell-cycle progression
.
Proc Natl Acad Sci USA
1999
;
96
:
8505
10
.
19.
Szegezdi
E
,
Logue
SE
,
Gorman
AM
,
Samali
A
. 
Mediators of endoplasmic reticulum stress-induced apoptosis
.
EMBO Rep
2006
;
7
:
880
5
.
20.
Tsutsumi
S
,
Gotoh
T
,
Tomisato
W
,
Mima
S
,
Hoshino
T
,
Hwang
HJ
, et al
Endoplasmic reticulum stress response is involved in nonsteroidal anti-inflammatory drug-induced apoptosis
.
Cell Death Differ
2004
;
11
:
1009
16
.
21.
Tsutsumi
S
,
Namba
T
,
Tanaka
KI
,
Arai
Y
,
Ishihara
T
,
Aburaya
M
, et al
Celecoxib upregulates endoplasmic reticulum chaperones that inhibit celecoxib-induced apoptosis in human gastric cells
.
Oncogene
2006
;
25
:
1018
29
.
22.
Yamazaki
T
,
Muramoto
M
,
Oe
T
,
Morikawa
N
,
Okitsu
O
,
Nagashima
T
, et al
Diclofenac, a non-steroidal anti-inflammatory drug, suppresses apoptosis induced by endoplasmic reticulum stresses by inhibiting caspase signaling
.
Neuropharmacology
2006
;
50
:
558
67
.
23.
Hosoi
T
,
Sasaki
M
,
Baba
S
,
Ozawa
K
. 
Effect of pranoprofen on endoplasmic reticulum stress in the primary cultured glial cells
.
Neurochem Int
2009
;
54
:
1
6
.
24.
Baek
SJ
,
Kim
JS
,
Moore
SM
,
Lee
SH
,
Martinez
J
,
Eling
TE
. 
Cyclooxygenase inhibitors induce the expression of the tumor suppressor gene EGR-1, which results in the up-regulation of NAG-1, an antitumorigenic protein
.
Mol Pharmacol
2005
;
67
:
356
64
.
25.
Feng
Q
,
Sekula
D
,
Muller
R
,
Freemantle
SJ
,
Dmitrovsky
E
. 
Uncovering residues that regulate cyclin D1 proteasomal degradation
.
Oncogene
2007
;
26
:
5098
106
.
26.
Wang
Y
,
Shen
J
,
Arenzana
N
,
Tirasophon
W
,
Kaufman
RJ
,
Prywes
R
. 
Activation of ATF6 and an ATF6 DNA binding site by the endoplasmic reticulum stress response
.
J Biol Chem
2000
;
275
:
27013
20
.
27.
Baek
SJ
,
Okazaki
R
,
Lee
SH
,
Martinez
J
,
Kim
JS
,
Yamaguchi
K
, et al
Nonsteroidal anti-inflammatory drug-activated gene-1 over expression in transgenic mice suppresses intestinal neoplasia
.
Gastroenterology
2006
;
131
:
1553
60
.
28.
Sukhthankar
M
,
Yamaguchi
K
,
Lee
SH
,
McEntee
MF
,
Eling
TE
,
Hara
Y
, et al
A green tea component suppresses posttranslational expression of basic fibroblast growth factor in colorectal cancer
.
Gastroenterology
2008
;
134
:
1972
80
.
29.
Wong
NACS
,
Pignatelli
M
. 
β-catenin—A linchpin in colorectal carcinogenesis?
Am J Pathol
2002
;
160
:
389
401
.
30.
Takada
Y
,
Bhardwaj
A
,
Potdar
P
,
Aggarwal
BB
. 
Nonsteroidal anti-inflammatory agents differ in their ability to suppress NF-kappaB activation, inhibition of expression of cyclooxygenase-2 and cyclin D1, and abrogation of tumor cell proliferation
.
Oncogene
2004
;
23
:
9247
58
.
31.
Geng
Y
,
Yu
Q
,
Sicinska
E
,
Das
M
,
Bronson
RT
,
Sicinski
P
. 
Deletion of the p27Kip1 gene restores normal development in cyclin D1-deficient mice
.
Proc Natl Acad Sci USA
2001
;
98
:
194
9
.
32.
Diehl
JA
,
Zindy
F
,
Sherr
CJ
. 
Inhibition of cyclin D1 phosphorylation on threonine-286 prevents its rapid degradation via the ubiquitin-proteasome pathway
.
Gene Dev
1997
;
11
:
957
72
.
33.
Zhang
K
,
Kaufman
RJ
. 
Protein folding in the endoplasmic reticulum and the unfolded protein response
.
Handb Exp Pharmacol
2006
;
172
:
69
91
.
34.
Kim
I
,
Xu
W
,
Reed
JC
. 
Cell death and endoplasmic reticulum stress: disease relevance and therapeutic opportunities
.
Nat Rev Drug Discov
2008
;
7
:
1013
30
.
35.
Armstrong
JL
,
Flockhart
R
,
Veal
GJ
,
Lovat
PE
,
Redfern
CP
. 
Regulation of endoplasmic reticulum stress-induced cell death by ATF4 in neuroectodermal tumor cells
.
J Biol Chem
2010
;
285
:
6091
100
.
36.
Carracedo
A
,
Gironella
M
,
Lorente
M
,
Garcia
S
,
Guzman
M
,
Velasco
G
, et al
Cannabinoids induce apoptosis of pancreatic tumor cells via endoplasmic reticulum stress-related genes
.
Cancer Res
2006
;
66
:
6748
55
.
37.
Pathi
S
,
Li
X
,
Safe
S
. 
Tolfenamic acid inhibits colon cancer cell and tumor growth and induces degradation of specificity protein (Sp) transcription factors
.
Mol Carcinog
2013 May 14
.
[Epub ahead of print]
.
38.
Jeong
JB
,
Yang
X
,
Clark
R
,
Choi
J
,
Baek
SJ
,
Lee
SH
. 
A mechanistic study of the proapoptotic effect of tolfenamic acid: involvement of NF-kappaB activation
.
Carcinogenesis
2013
;
34
:
2350
60
.
39.
Zhang
X
,
Min
KW
,
Liggett
J
,
Baek
SJ
. 
Disruption of the transforming growth factor-beta (TGF-beta) pathway by tolfenamic acid via the ERK MAP kinase pathway
.
Carcinogenesis
2013 July 16
.
[Epub ahead of print]
.
40.
Eslin
D
,
Sankpal
UT
,
Lee
C
,
Sutphin
RM
,
Maliakal
P
,
Currier
E
, et al
Tolfenamic acid inhibits neuroblastoma cell proliferation and induces apoptosis: a novel therapeutic agent for neuroblastoma
.
Mol Carcinog
2013
;
52
:
377
86
.
41.
Basha
R
,
Ingersoll
SB
,
Sankpal
UT
,
Ahmad
S
,
Baker
CH
,
Edwards
JR
, et al
Tolfenamic acid inhibits ovarian cancer cell growth and decreases the expression of c-Met and survivin through suppressing specificity protein transcription factors
.
Gynecol Oncol
2011
;
122
:
163
70
.
42.
Harding
HP
,
Zhang
Y
,
Ron
D
. 
Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase
.
Nature
1999
;
397
:
271
4
.
43.
Hamanaka
RB
,
Bennett
BS
,
Cullinan
SB
,
Diehl
JA
. 
PERK and GCN2 contribute to eIF2alpha phosphorylation and cell cycle arrest after activation of the unfolded protein response pathway
.
Mol Biol Cell
2005
;
16
:
5493
501
.
44.
Romero-Ramirez
L
,
Cao
H
,
Nelson
D
,
Hammond
E
,
Lee
AH
,
Yoshida
H
, et al
XBP1 is essential for survival under hypoxic conditions and is required for tumor growth
.
Cancer Res
2004
;
64
:
5943
7
.
45.
Bi
M
,
Naczki
C
,
Koritzinsky
M
,
Fels
D
,
Blais
J
,
Hu
N
, et al
ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth
.
EMBO J
2005
;
24
:
3470
81
.
46.
Lee
AH
,
Iwakoshi
NN
,
Anderson
KC
,
Glimcher
LH
. 
Proteasome inhibitors disrupt the unfolded protein response in myeloma cells
.
Proc Natl Acad Sci USA
2003
;
100
:
9946
51
.
47.
Nawrocki
ST
,
Carew
JS
,
Dunner
K
 Jr
,
Boise
LH
,
Chiao
PJ
,
Huang
P
, et al
Bortezomib inhibits PKR-like endoplasmic reticulum (ER) kinase and induces apoptosis via ER stress in human pancreatic cancer cells
.
Cancer Res
2005
;
65
:
11510
9
.
48.
Nawrocki
ST
,
Carew
JS
,
Pino
MS
,
Highshaw
RA
,
Dunner
K
 Jr
,
Huang
P
, et al
Bortezomib sensitizes pancreatic cancer cells to endoplasmic reticulum stress-mediated apoptosis
.
Cancer Res
2005
;
65
:
11658
66
.
49.
Stolfi
C
,
Sarra
M
,
Caruso
R
,
Fantini
MC
,
Fina
D
,
Pellegrini
R
, et al
Inhibition of colon carcinogenesis by 2-methoxy-5-amino-N-hydroxybenzamide, a novel derivative of mesalamine
.
Gastroenterology
2010
;
138
:
221
30
.
50.
Pyrko
P
,
Kardosh
A
,
Liu
YT
,
Soriano
N
,
Xiong
W
,
Chow
RH
, et al
Calcium-activated endoplasmic reticulum stress as a major component of tumor cell death induced by 2,5-dimethyl-celecoxib, a non-coxib analogue of celecoxib
.
Mol Cancer Ther
2007
;
6
:
1262
75
.
51.
Jiang
HY
,
Wek
SA
,
McGrath
BC
,
Lu
D
,
Hai
T
,
Harding
HP
, et al
Activating transcription factor 3 is integral to the eukaryotic initiation factor 2 kinase stress response
.
Mol Cell Biol
2004
;
24
:
1365
77
.
52.
Reimertz
C
,
Kogel
D
,
Rami
A
,
Chittenden
T
,
Prehn
JH
. 
Gene expression during ER stress-induced apoptosis in neurons: induction of the BH3-only protein Bbc3/PUMA and activation of the mitochondrial apoptosis pathway
.
J Cell Biol
2003
;
162
:
587
97
.
53.
Zhang
T
,
Nanney
LB
,
Luongo
C
,
Lamps
L
,
Heppner
KJ
,
DuBois
RN
, et al
Concurrent overexpression of cyclin D1 and cyclin-dependent kinase 4 (Cdk4) in intestinal adenomas from multiple intestinal neoplasia (Min) mice and human familial adenomatous polyposis patients
.
Cancer Res
1997
;
57
:
169
75
.
54.
Powell
SM
,
Petersen
GM
,
Krush
AJ
,
Booker
S
,
Jen
J
,
Giardiello
FM
, et al
Molecular diagnosis of familial adenomatous polyposis
.
N Engl J Med
1993
;
329
:
1982
7
.