In several neoplastic diseases, including hepatocellular carcinoma, the expression of P-glycoprotein and cyclooxygenase-2 (COX-2) are often increased and involved in drug resistance and poor prognosis. P-glycoprotein, in addition to drug resistance, blocks cytochrome c release, preventing apoptosis in tumor cells. Because COX-2 induces P-glycoprotein expression, we evaluated the effect of celecoxib, a specific inhibitor of COX-2 activity, on P-glycoprotein–mediated resistance to apoptosis in cell lines expressing multidrug resistant (MDR) phenotype. Experiments were done using MDR-positive and parental cell lines at basal conditions and after exposure to 10 or 50 μmol/L celecoxib. We found that 10 μmol/L celecoxib reduced P-glycoprotein, Bcl-xL, and Bcl-2 expression, and induced translocation of Bax from cytosol to mitochondria and cytochrome c release into cytosol in MDR-positive hepatocellular carcinoma cells. This causes the activation of caspase-3 and increases the number of cells going into apoptosis. No effect was shown on parental drug-sensitive or on MDR-positive hepatocellular carcinoma cells after transfection with MDR1 small interfering RNA. Interestingly, although inhibiting COX-2 activity, 50 μmol/L celecoxib weakly increased the expression of COX-2 and P-glycoprotein and did not alter Bcl-xL and Bcl-2 expression. In conclusion, these results show that relatively low concentrations of celecoxib induce cell apoptosis in MDR cell lines. This effect is mediated by P-glycoprotein and suggests that the efficacy of celecoxib in the treatment of different types of cancer may depend on celecoxib concentration and P-glycoprotein expression. [Cancer Res 2007;67(10):4915–23]

One type of drug resistance to anticancer compounds is mediated by the overexpression of the P-glycoprotein, the product of multidrug resistant (MDR1) gene (1). P-glycoprotein is a membrane-associated glycoprotein and a member of the ATP-binding cassette superfamily of membrane transport proteins (2). P-glycoprotein works like an energy-dependent efflux pump to extrude anticancer drugs and other compounds from tumor cells (3).

Previous studies have shown increased levels of the inducible enzyme cyclooxygenase-2 (COX-2) in various types of carcinoma, including hepatocellular carcinoma (4). Cyclooxygenases (COX-1 and COX-2) are enzymes that catalyze the metabolism of arachidonic acid to prostaglandins and thromboxanes (5). COX-1 is constitutively expressed in many tissues and is responsible for various physiologic functions (68). COX-2 is an immediate-early inducible gene that responds to various stimuli, such as mitogens, cytokines, and growth factors; and is involved in inflammation, regulation of cell growth, differentiation, prevention of apoptosis, tumorigenesis, and angiogenesis (7, 914).

The expression of P-glycoprotein and COX-2 in cancerous tissues varies according to several factors, decreasing with the increase of anaplasia (4, 15). This observation suggests that P-glycoprotein and COX-2 can play a role in early stage of carcinogenesis and in malignity in many organs, liver included, although very few links have been shown between P-glycoprotein and COX-2 pathways. We showed that the development of the MDR phenotype is associated with a constitutive expression of COX-2 and inducible nitric oxide synthase in a human hepatocellular carcinoma cell line and these enzymes may enhance the angiogenic activity of the MDR-positive hepatocellular carcinoma cell line (16, 17). Moreover, a causal link between expression and activity of COX-2 and P-glycoprotein was shown (1821). Cells expressing the MDR phenotype are more resistant to several agents that trigger cell apoptosis compared with parental drug-sensitive cells (22). This phenomenon can be linked to the fact that MDR-positive cells exhibit a block in the release of mitochondrial cytochrome c into the cytosol and, because of this, they seem protected from undergoing apoptosis (23). This feature of MDR-positive cells seems to be dependent on overexpression of Bcl-xL, an antiapoptotic protein that belongs to the Bcl-2 family (23). Bcl-xL and Bcl-2 act on the release of cytochrome c from mitochondria by modulating the activity of the voltage-dependent anion channel (24). Their overexpression blocks cytochrome c release and activation of caspase-3 (25). Moreover, the transfection of human hepatocellular carcinoma cell line PLC/PRF/5 with a plasmid vector containing recombinant bcl-2 makes these cells resistant to doxorubicin-induced cytotoxicity, suggesting a possible link between P-glycoprotein and Bcl-2 family members expression (26).

COX-2 is thought to play a role in cell apoptosis, but the signaling pathways affected by COX-2 expression and/or activity are still not fully understood (27). The COX inhibitors, known as coxibs, inhibit the proliferation and induce apoptosis in cultured hepatocellular carcinoma cells, although these inhibitors are known to mediate effects through both COX-dependent and COX-independent mechanisms (28). Celecoxib induces cytochrome c release, activation of caspase-9 and caspase-3 and eventually apoptosis in hepatocellular carcinoma cells (29, 30). However, its mechanism of action should be further explored. It is possible to prevent celecoxib-induced cell death by z-Val Ala Asp (Ome)-fluoromethylketone (z-VAD), a wide-spectrum inhibitor of caspase activity (29). Celecoxib, a well-known inhibitor of COX-2 activity, induces apoptosis also in cells that lack the COX-2 enzyme (31).

Because COX-2 and P-glycoprotein proteins are expressed in hepatocellular carcinoma (16, 32), they could affect the apoptotic pathways and cooperate in the development of the MDR phenotype, at least in human hepatocellular carcinoma. In recent years, inhibitors of COX-2, celecoxib included, have been proposed in the treatment of hepatocellular carcinoma, and in fact, the inhibition of COX-2 was proved to possess antiproliferative action (33).

Based on these observations, this work was done to examine the effect of celecoxib on different cell lines expressing the MDR phenotype. To address this issue, the expressions of P-glycoprotein, COX-2, Bcl-2, and Bcl-xL; the release of mitochondrial cytochrome c; the translocation of Bax to the mitochondria; the percentage of fragmented Hoechst-positive nuclei, of Annexin V–positive cells, and of terminal deoxynucleotidyl transferase (TdT)–mediated nick end labeling (TUNEL)–stained cells; and the cell death were assessed at basal conditions and after exposure to celecoxib in different cell lines.

Cell lines. Experiments were done mainly on a human hepatocellular carcinoma cell line PLC/PRF/5 (34), by using the drug-sensitive clone (P5) and the highly MDR subclone [P1(0.5)], and confirmative experiments were done on human colon cancer drug-sensitive (HT-29) and drug-resistant (HT-29-dx) cells, on human ovarian cancer cells (IGROV-1), and on murine NIH3T3 drug-sensitive (PSI-2) and drug-resistant (PN1A) cells.

The P1(0.5) subclone was developed by serial prolonged exposures to increasing concentrations of doxorubicin (Pharmacia & Upjohn) starting from parental P5 cells, and subsequently cultured in DMEM containing 0.5 μg/mL doxorubicin and 10% fetal bovine serum (FBS), as previously reported (16). The human colon carcinoma cells HT-29 and the subpopulation of doxorubicin-resistant cells (HT29-dx) were kindly donated by Dr. A. Bosia (Department of Genetics, Biology and Biochemistry, University of Turin, Turin, Italy). The HT-29 cells were cultured in RPMI 1640 supplemented with 10% FBS; the HT-29-dx were maintained in RPMI 1640 supplemented with 10% FBS and 34 nmol/L doxorubicin, as described by C. Riganti et al. (35). The human ovarian adenocarcinoma cell line (IGROV-1), kindly provided by Dr. J.H. Schellens (the Netherlands Cancer Institute, Amsterdam, the Netherlands), was maintained in RPMI 1640 supplemented with 10% FBS (36). Two subclones of NIH3T3 cells, one of which is drug-resistant (PN1A) and the other drug-sensitive (PSI-2), were originally provided by J.M. Croop (Pediatric Hematology/Oncology, James Whitcomb Riley Hospital for Children, Indianapolis, IN) and were cultured in DMEM supplemented with 10% FBS; 0.1 μg/mL doxorubicin was added to the PN1A cell medium, as previously described (37). The PN1A cell line was generated by transfection of PSI-2 cells with expression vector pBA-mdr, which contained mouse mdr1 cDNA. All cell lines were cultured at 37°C in an atmosphere containing 5% CO2.

Cell treatment. Because only a part of celecoxib added to medium containing 10% FBS is protein-unbound and hence available for interacting on cells, doxorubicin-free, serum-free medium containing celecoxib was used in all experiments, as described in the details for each experimental condition (38).

Western blot analysis. Cells were seeded in complete medium for 24 h and then exposed to doxorubicin-free, serum-free medium containing 10 or 50 μmol/L celecoxib (a kind gift of Prof. E. Masini, Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy). After 3, 6, and 24 h, total proteins were extracted and evaluated by Western blot analysis, as previously described (16), using each of the following primary antibodies: anti–COX-2 polyclonal antibody (Cayman Chemical); anti–P-glycoprotein (C219) monoclonal antibody (Calbiochem); anti-Bax polyclonal antibody, anti–Bcl-2 monoclonal antibody, anti–Bcl-xL monoclonal antibody, and anti–cytochrome c polyclonal antibody (Oncogene Research Products); anti–caspase-3 (E8) monoclonal antibody (Santa Cruz Biotechnology); anti–β-actin monoclonal antibody (Sigma Chemical Co.).

Small interfering RNA transfection. Positive control GAPDH small interfering RNA (siRNA), siCONTROL nontargeting siRNA and a pool of four different double-stranded RNA oligonucleotides directed against MDR1 (siGENOME SMARTpool, human ABCB1) were purchased from Dharmacon. P1(0.5) cells were seeded in 60-mm dishes to reach 30% to 50% confluency after 24 h of incubation, and transfected with a total of 100 nmol/L siRNA using LipofectAMINE 2000 (Invitrogen Life Technologies, Inc.) in antibiotic-free medium, according to the manufacturer's instructions. Forty-eight hours after transfection, total RNA was extracted for reverse transcription-PCR (RT-PCR) analysis, and 72 h after transfection, total proteins were extracted for Western blot analysis.

Reverse transcription-PCR. Total RNA was extracted from cells with TRIzol reagent (Invitrogen Life Technologies) and quantified by absorbance spectroscopy. The reverse transcription reaction was done by heating for 5 min at 65°C 5 ng of total RNA, 250 ng of random primer, and 1 mmol/L deoxyribonucleoside triphosphate (dNTP). After cooling at 4°C, the mixture was incubated for 50 min at 37°C in the reverse transcription reaction mixture (10 mmol/L DTT, 20 units RNAsin; Invitrogen), and 200 units of Moloney murine leukemia virus reverse transcriptase (Invitrogen Life Technologies). Samples were then heated at 70°C for 15 min. Five microliters of the synthesized cDNA was then amplified in a 55 μL reaction mixture containing Taq polymerase buffer, 1 mmol/L dNTP, 2.5 mmol/L MgCl2, 0.5 μmol/L of each primer, and 0.5 units Taq polymerase (Invitrogen Life Technologies). The primers used were as follows: for MDR1, 5′-ATATCAGCAGCCCACATCAT-3′ and 5′-GAAGCACTGGGATGTCCGGT-3′; for GAPDH, 5′-GCCAAAAGGGTCATCATCTC-3′ and 5′-GTAGAGGCAGGGATGATGTTC-3′ (39). Amplification cycles were 94°C for 3 min, then 33 cycles at 94°C for 1 min, 58°C for 1 min, 72°C for 1.5 min, followed by 72°C for 15 min. Aliquots of the PCR product were electrophoresed on 1.5% agarose gels, and PCR fragments were visualized by ethidium bromide staining (39).

Preparation of mitochondrial fractions. Cells were washed twice with ice-cold PBS, trypsinized, and collected by centrifugation; the pellet was suspended in 500 μL of ice-cold buffer A [20 mmol/L HEPES (pH 7.5), 1.5 mmol/L MgCl2, 10 mmol/L KCl, 1 mmol/L EDTA, 1 mmol/L EGTA, 1 mmol/L DTT, 0.1 mmol/L phenylmethylsulfonyl fluoride, and 10 μg/mL each leupeptin, aprotinin, and pepstatin A] containing 250 mmol/L sucrose.

Unbroken cells, large plasma membrane pieces, and nuclei were removed by centrifuging the homogenates at 1,000 × g at 4°C for 10 min. The resulting supernatant was subjected to 10,000 × g centrifugation at 4°C for 20 min. The pellet fraction (i.e., mitochondria) was first washed with the above buffer A containing sucrose and then solubilized in 50 μL of TNC buffer [10 mmol/L Tris acetate (pH 8.0), 0.5% Igepal CA-630, 5 mmol/L CaCl2]. The supernatant was recentrifuged at 100,000 × g (4°C, 1 h) to generate cytosol.

Immunofluorescence analysis of cytochrome c release. Cells were plated onto a coverslip and were grown in culture medium containing 10% FBS. After 24 h, cells were treated with 10 μmol/L celecoxib in doxorubicin-free, serum-free medium for 3 h and then were fixed with paraformaldehyde (3.7% w/v), permeabilized with 0.05% (v/v) Triton X-100 for 10 min, blocked with 10% (w/v) normal goat serum, then incubated overnight with primary antibody at an appropriate dilution (1:500). The purified anti–cytochrome c monoclonal antibody was obtained from BD Biosciences PharMingen (BD Biosciences). After washing, samples were incubated with FITC-conjugated secondary antibodies (Molecular Probes) at room temperature in the dark. FITC-labeled cells were analyzed by fluorescence Nikon microscope using standard fluorescein filters (excitation 488 nm).

Hoechst assay. Nuclear morphology was assessed using Hoechst staining. Cells were plated onto coverslip and were grown in complete medium. After 24 h at 37°C, the cells were rinsed to remove the detached cells, and then exposed to doxorubicin-free, serum-free medium containing 10 μmol/L celecoxib or 10 μmol/L verapamil or 100 or 200 μmol/L taurolithocholic acid for 24 h. Afterward, the supernatant was removed and cells were washed twice with PBS. Then, the cells were fixed for 20 min at room temperature in freshly prepared 3.7% paraformaldehyde in PBS, then rinsed thrice with PBS. After that, 0.5% Triton X-100 was added to the sample and left for 5 min at room temperature. Cells were washed twice with PBS and stained for 30 min at 37°C in Hoechst 33258 1 μg/mL (Sigma Chemical); then, they were rinsed with PBS and examined with a fluorescence microscope (UV light). Fragmented nuclei were expressed as percentage of fragmented Hoechst-positive nuclei versus the total Hoechst-positive nuclei.

Annexin V assay. Drug-sensitive and drug-resistant cells were plated in 60-mm Petri dishes and grown in DMEM supplemented with 10% FBS. The next day, cells were treated with 10 μmol/L celecoxib in doxorubicin-free, serum-free medium for 24 h. Afterward, both floating and attached cells were collected by brief trypsinization. Cells were stained with an Annexin V–Fluos staining kit (Roche Applied Science), and apoptosis was evaluated by using the FACScan flow cytometer (Becton Dickinson).

TUNEL assay. DNA fragmentation during apoptosis was detected by the TUNEL technique using an apoptosis detection kit. Briefly, drug-sensitive and drug-resistant cells were plated in 60-mm Petri dishes and grown in DMEM supplemented with 10% FBS. The next day, cells were treated with 10 μmol/L celecoxib in doxorubicin-free, serum-free medium for 24 h. Cells were fixed in 10% (v/v) formalin and, subsequently, a 250 μL TUNEL reaction mixture containing biotinylated nucleotide and TdT was added to cells for 60 min at 37°C in the dark. For negative control, TdT was omitted from the reaction mixture. The reaction was analyzed by Nikon fluorescent microscope.

Cell death assay. P5 and P1(0.5) cells were plated in 60-mm Petri dishes and grown in DMEM supplemented with 10% FBS. On the next day, cells were treated with 10 μmol/L celecoxib in doxorubicin-free, serum-free medium; moreover, 1 h before the addition of celecoxib, cells were preincubated with a cell permeable pan-caspase-inhibitor z-VAD at 20 μmol/L (Sigma). After 6, 12, 18, and 24 h, the dead cells were identified using the trypan blue staining test. Adherent cells were detached, resuspended in DMEM, and both adherent and suspended cells were mixed with an equal volume of 0.04% (w/v) trypan blue. The percentage of dead cells stained was determined by counting.

Statistical analysis. The significance of the differences between groups was determined by ANOVA followed by the Bonferroni post hoc test, using GraphPad Prism version 4.0 for Windows (GraphPad Software). P < 0.05 was considered significant.

Celecoxib modifies P-glycoprotein expression in MDR-positive cells. First, we confirmed that P1(0.5) cells have markedly higher expression of P-glycoprotein and COX-2 compared with parental cells P5 (Fig. 1A,, i). To evaluate whether COX-2 activity induced the expression of P-glycoprotein in our model, we determined whether the selective inhibition of COX-2 activity by celecoxib modified P-glycoprotein and/or COX-2 expression in P1(0.5) and in parental P5 cells. The levels of P-glycoprotein and COX-2 expression in P1(0.5) cells cultured in a doxorubicin-free, serum-free medium for 3, 6, and 24 h were not modified (Fig. 1A,, ii). Western blot analysis clearly showed that the P-glycoprotein expression was reduced in P1(0.5) cells treated for 3 h with 10 μmol/L celecoxib and the reduced expression persisted at least up to 24 h, whereas COX-2 expression was not altered (Fig. 1B,, i and ii). On the contrary, cell exposure to a 5-fold higher concentration of celecoxib (50 μmol/L) weakly increased COX-2 expression at 6 h and P-glycoprotein expression at 24 h in P1(0.5) cells (Fig. 1C). P-glycoprotein and COX-2 proteins were not expressed in P5 parental cells at basal conditions and their exposure to celecoxib did not change the expression of these proteins (Fig. 1A,, i, B, i, and C). Because it is known that COX-2 induces P-glycoprotein expression but it is unknown whether P-glycoprotein can modify COX-2 expression, we evaluated the effect of inhibiting the expression of MDR1 by siRNA on COX-2 expression. The MDR1 siRNA reduced the levels of MDR1 mRNA (Fig. 1D,, i) and abolished the expression of P-glycoprotein (Fig. 1D,, ii). siRNA targeting GAPDH strongly reduced GAPDH transcript levels (data not shown). The inhibition of P-glycoprotein expression was not observed using the nonspecific control siRNA or the mock transfection control (data not shown). MDR1 siRNA did not affect the expression of COX-2 in P1(0.5) cells (Fig. 1D , ii). The inhibitory effect of celecoxib on P-glycoprotein expression was also evaluated in different cell lines (Supplementary Fig. S1). The levels of P-glycoprotein in HT-29-dx, IGROV-1, and PN1A cells cultured in a doxorubicin-free, serum-free medium for 3, 6, and 24 h were not modified (data not shown). The expression of P-glycoprotein was reduced in HT-29-dx after 3, 6, and 24 h of treatment with 10 μmol/L celecoxib (Supplementary Fig. S1A, i and iii). On the contrary, under the same experimental conditions, the P-glycoprotein levels were not affected in parental HT-29 cells (Supplementary Fig. S1A, i and ii). As shown in Supplementary Fig. S1A, iv, celecoxib reduced P-glycoprotein expression only after 6 h in IGROV-1 cells. Moreover, P-glycoprotein expression was reduced in mdr1 cDNA transfected PN1A cells after treatment with 10 μmol/L celecoxib for 3, 6, and 24 h (Supplementary Fig. S1C, i and iii). No changes in the expression of P-glycoprotein were observed in parental PSI-2 cells treated with celecoxib (Supplementary Fig. S1C, i and ii).

Figure 1.

Effect of celecoxib on COX-2 and P-glycoprotein expression in P5 and P1(0.5) cells. A, i, Western blot analysis of P-glycoprotein (P-gp) and COX-2 expression in P5 and P1(0.5) cells at basal conditions. ii, Western blot analysis of P-glycoprotein and COX-2 expression in P1(0.5) cells cultured at basal conditions (Cont) or in doxorubicin (DOX)-free, serum-free medium for 3, 6, and 24 h. B, i, P-glycoprotein and COX-2 expression in P5 and P1(0.5) cells at basal conditions and after treatment with 10 μmol/L celecoxib (CLX) for 3, 6, and 24 h. ii, results of densitometric analysis in P1(0.5) cells are reported in the graph as percentage of control band density. C, P-glycoprotein and COX-2 expression in P5 and P1(0.5) cells at basal condition and after treatment with 50 μmol/L celecoxib for 3, 6, and 24 h. D, i, effect of the MDR1 siRNA on MDR1 mRNA expression evaluated by RT-PCR analysis at basal conditions and after 72 h transfection with MDR1 siRNA in P1(0.5) cells. SM, 123-bp size marker (Invitrogen Life Technologies). ii, P-glycoprotein and COX-2 expression at basal conditions, after 72 h transfection with MDR1 siRNA and after treatment with 10 μmol/L celecoxib for 3 and 6 h following 72 h transfection (MDR1 siRNA + celecoxib 10 μmol/L) in P1(0.5) cells. β-Actin was used in all Western blot analyses as a protein loading control. Three separate experiments were done and one representative of all is shown.

Figure 1.

Effect of celecoxib on COX-2 and P-glycoprotein expression in P5 and P1(0.5) cells. A, i, Western blot analysis of P-glycoprotein (P-gp) and COX-2 expression in P5 and P1(0.5) cells at basal conditions. ii, Western blot analysis of P-glycoprotein and COX-2 expression in P1(0.5) cells cultured at basal conditions (Cont) or in doxorubicin (DOX)-free, serum-free medium for 3, 6, and 24 h. B, i, P-glycoprotein and COX-2 expression in P5 and P1(0.5) cells at basal conditions and after treatment with 10 μmol/L celecoxib (CLX) for 3, 6, and 24 h. ii, results of densitometric analysis in P1(0.5) cells are reported in the graph as percentage of control band density. C, P-glycoprotein and COX-2 expression in P5 and P1(0.5) cells at basal condition and after treatment with 50 μmol/L celecoxib for 3, 6, and 24 h. D, i, effect of the MDR1 siRNA on MDR1 mRNA expression evaluated by RT-PCR analysis at basal conditions and after 72 h transfection with MDR1 siRNA in P1(0.5) cells. SM, 123-bp size marker (Invitrogen Life Technologies). ii, P-glycoprotein and COX-2 expression at basal conditions, after 72 h transfection with MDR1 siRNA and after treatment with 10 μmol/L celecoxib for 3 and 6 h following 72 h transfection (MDR1 siRNA + celecoxib 10 μmol/L) in P1(0.5) cells. β-Actin was used in all Western blot analyses as a protein loading control. Three separate experiments were done and one representative of all is shown.

Close modal

Celecoxib modifies Bcl-xL and Bcl-2 expression in MDR-positive cells. To examine a possible association between P-glycoprotein expression and resistance to mitochondrial-related apoptosis, the expression of Bcl-xL, Bcl-2, and Bax proteins was determined in parental P5 and in MDR- and COX-2–positive P1(0.5) cells. Western blot analysis showed that antiapoptotic proteins Bcl-xL and Bcl-2 were more expressed in P1(0.5) cells than in parental P5 cells at basal conditions. On the contrary, the expression of Bax, the proapoptotic member of Bcl-2 family, was higher in parental P5 than in P1(0.5) cells (Fig. 2A,, i). The levels of Bcl-xL, Bcl-2, and Bax expression in P1(0.5) cells cultured in doxorubicin-free, serum-free medium up to 24 h were unmodified (Fig. 2A,, ii). Interestingly, 10 μmol/L celecoxib strongly reduced Bcl-xL expression after 3 and 6 h and weakly after 24 h (Fig. 2B,, i and ii); celecoxib also reduced Bcl-2 expression at 6 h in P1(0.5) cells (Fig. 2B,, i and ii). On the contrary, 50 μmol/L celecoxib did not alter the expression of these proteins in P1(0.5) cells (data not shown). No significant changes in Bcl-xL or Bcl-2 expression were observed in parental P5 cells (Fig. 2B , i).

Figure 2.

Effect of celecoxib on the expression of proapoptotic and antiapoptotic proteins in P5 and P1(0.5) cells. A, i, Western blot analysis of Bcl-xL, Bcl-2, and Bax expression in P5 and P1(0.5) cells at basal conditions. ii, Western blot analysis of Bcl-xL, Bcl-2, and Bax expression in P1(0.5) cells cultured at basal conditions or in doxorubicin-free, serum-free medium for 3, 6, and 24 h. B, i, Bcl-xL and Bcl-2 expression in P5 and P1(0.5) cells at basal condition and after treatment with 10 μmol/L celecoxib for 3, 6, and 24 h. ii, results of densitometric analysis in P1(0.5) cells are reported in the graph as percentage of control band density. C, P-glycoprotein, Bcl-xL, and Bcl-2 expression at basal conditions, after 72 h transfection with MDR1 siRNA, and after treatment with 10 μmol/L celecoxib for 3 and 6 h following 72 h transfection (MDR1 siRNA + celecoxib 10 μmol/L) in P1(0.5) cells. β-Actin was used in all Western blot analyses as a protein loading control. Three separate experiments were done and one representative of all is shown.

Figure 2.

Effect of celecoxib on the expression of proapoptotic and antiapoptotic proteins in P5 and P1(0.5) cells. A, i, Western blot analysis of Bcl-xL, Bcl-2, and Bax expression in P5 and P1(0.5) cells at basal conditions. ii, Western blot analysis of Bcl-xL, Bcl-2, and Bax expression in P1(0.5) cells cultured at basal conditions or in doxorubicin-free, serum-free medium for 3, 6, and 24 h. B, i, Bcl-xL and Bcl-2 expression in P5 and P1(0.5) cells at basal condition and after treatment with 10 μmol/L celecoxib for 3, 6, and 24 h. ii, results of densitometric analysis in P1(0.5) cells are reported in the graph as percentage of control band density. C, P-glycoprotein, Bcl-xL, and Bcl-2 expression at basal conditions, after 72 h transfection with MDR1 siRNA, and after treatment with 10 μmol/L celecoxib for 3 and 6 h following 72 h transfection (MDR1 siRNA + celecoxib 10 μmol/L) in P1(0.5) cells. β-Actin was used in all Western blot analyses as a protein loading control. Three separate experiments were done and one representative of all is shown.

Close modal

To test the role of P-glycoprotein in the process of mitochondrial apoptosis induction, we did siRNA experiments in P1(0.5) cells. The MDR1 siRNA reduced the expression of antiapoptotic proteins Bcl-xL and Bcl-2, compared with basal conditions, whereas 10 μmol/L celecoxib did not modify the Bcl-xL and Bcl-2 expression in P1(0.5) cells transfected with MDR1 siRNA (Fig. 2C).

Because P-glycoprotein activity may affect mitochondrial apoptosis induction, we assessed the effect of verapamil and taurolithocholic acid, two inhibitors of P-glycoprotein activity (40), on P-glycoprotein, procaspase-3, Bcl-xL, Bcl-2, and Bax expression in MDR-positive cells. As shown in Supplementary Fig. S2A and B, the treatment with 10 μmol/L verapamil or 100 or 200 μmol/L taurolithocholic acid for 3, 6, or 24 h did not modify the expression of these proteins in P1(0.5) cell line. Moreover, the two inhibitors of P-glycoprotein activity did not alter the percentage of fragmented Hoechst-positive nuclei (Supplementary Fig. S2C).

Celecoxib induces Bax translocation and mitochondrial cytochrome c release in MDR-positive cells. The expression of Bax did not change in P5 and P1(0.5) cells after exposure to 10 μmol/L celecoxib (Fig. 3A). However, a different subcellular localization of Bax between cytosolic and mitochondrial fractions was observed in P1(0.5) cells after their exposure to 10 μmol/L celecoxib for 3, 6, or 24 h. As shown in Fig. 3B, a partial translocation of Bax to mitochondria was observed after 3 and 6 h of treatment with 10 μmol/L celecoxib. The subcellular localization of Bax did not change in parental P5 cells after exposure to celecoxib (Fig. 3B).

Figure 3.

Effect of celecoxib on the expression and subcellular localization of Bax protein in P5 and P1(0.5) cells. A, Western blot analysis of Bax expression in whole-cell lysates of P5 and P1(0.5) cells at basal conditions and after treatment with 10 μmol/L celecoxib for 3, 6, and 24 h. β-Actin was used as a protein loading control; three separate experiments were done and one representative of all is shown. B, Western blot analysis for subcellular localization of Bax protein expression in mitochondrial and cytosolic fractions obtained from P5 and P1(0.5) cells at basal conditions and after treatment with 10 μmol/L celecoxib for 3, 6, and 24 h; results of densitometric analysis are reported in the graph as percentage of control band density. Three separate experiments were done and one representative of all is shown.

Figure 3.

Effect of celecoxib on the expression and subcellular localization of Bax protein in P5 and P1(0.5) cells. A, Western blot analysis of Bax expression in whole-cell lysates of P5 and P1(0.5) cells at basal conditions and after treatment with 10 μmol/L celecoxib for 3, 6, and 24 h. β-Actin was used as a protein loading control; three separate experiments were done and one representative of all is shown. B, Western blot analysis for subcellular localization of Bax protein expression in mitochondrial and cytosolic fractions obtained from P5 and P1(0.5) cells at basal conditions and after treatment with 10 μmol/L celecoxib for 3, 6, and 24 h; results of densitometric analysis are reported in the graph as percentage of control band density. Three separate experiments were done and one representative of all is shown.

Close modal

To test the hypothesis that 10 μmol/L celecoxib could restore mitochondrial cytochrome c release into the cytosol, MDR- and COX-2–positive cells were treated with 10 or 50 μmol/L celecoxib for 3, 6, and 24 h, then cells were fractioned and cytosolic and mitochondrial cytochrome c were analyzed by Western blot analysis. As shown in Fig. 4A, cytochrome c was released into the cytosol following treatment of P1(0.5) cells with 10 μmol/L celecoxib for 3, 6, and, especially, 24 h. On the contrary, cells treated with a higher concentration of celecoxib (50 μmol/L) did not release cytochrome c from mitochondria into cytosol (Fig. 4A). Immunofluorescence analysis on P5 and P1(0.5) cells treated for 3 h with 10 μmol/L celecoxib confirmed results of Western blot analysis, showing the effect of celecoxib on cytochrome c release into cytosol only in P1(0.5) cells (Fig. 4B). On the contrary, no changes in mitochondrial cytochrome c release were observed when parental P5 cells were treated with celecoxib (Fig. 4A and B).

Figure 4.

Effect of celecoxib on cytochrome c release into cytosol in P5 and P1(0.5) cells. A, Western blot analysis of cytochrome c protein expression in mitochondrial and cytosolic fractions obtained from P5 and P1(0.5) cells at basal conditions and after treatment with 10 or 50 μmol/L celecoxib for 3, 6, and 24 h; results of densitometric analysis are reported in the graph as percentage of control band density; three separate experiments were done and one representative of all is shown. B, immunofluorescence analysis of cytochrome c release into cytosol in P5 and P1(0.5) cells cultured for 3 h in doxorubicin-free, serum-free medium (Control) or treated with 10 μmol/L celecoxib for 3 h. Cells labeled with cytochrome c antibody were visualized by using a fluorescence microscope (original magnification, ×40); the images are representative of five separate experiments.

Figure 4.

Effect of celecoxib on cytochrome c release into cytosol in P5 and P1(0.5) cells. A, Western blot analysis of cytochrome c protein expression in mitochondrial and cytosolic fractions obtained from P5 and P1(0.5) cells at basal conditions and after treatment with 10 or 50 μmol/L celecoxib for 3, 6, and 24 h; results of densitometric analysis are reported in the graph as percentage of control band density; three separate experiments were done and one representative of all is shown. B, immunofluorescence analysis of cytochrome c release into cytosol in P5 and P1(0.5) cells cultured for 3 h in doxorubicin-free, serum-free medium (Control) or treated with 10 μmol/L celecoxib for 3 h. Cells labeled with cytochrome c antibody were visualized by using a fluorescence microscope (original magnification, ×40); the images are representative of five separate experiments.

Close modal

Effect of celecoxib on apoptosis and cell death of MDR-positive and MDR-negative cells. Because Bax translocation from cytosol to the mitochondria and the mitochondrial cytochrome c release into the cytosol play a crucial role in the induction of cellular apoptosis, the expression of the precursor form of caspase-3 (procaspase-3) was evaluated by Western blot analysis. Moreover, the percentage of fragmented nuclei, estimated by histochemical nuclear staining with Hoechst 33258, the percentage of Annexin V– and TUNEL-positive cells were evaluated in P5 and P1(0.5) cell lines treated with celecoxib. Evident activation of caspase-3 was shown by degradation of its precursor form after treatment with 10 μmol/L celecoxib at 3 and 6 h in P1(0.5) cells (Fig. 5A), at 3, 6, and 24 h in HT-29-dx cells and at 3 and 6 h in IGROV-1 cells (Supplementary Fig. S1A, iii and iv). On the contrary, the procaspase-3 levels were not affected in parental P5 and HT-29 cells (Fig. 5A and Supplementary Fig. S1A ii). In addition, treatment with 50 μmol/L celecoxib for 3, 6, or 24 h did not modify the procaspase-3 levels and the percentage of cells with fragmented Hoechst-positive nuclei in P1(0.5) cells (Fig. 5A and C).

Figure 5.

Effect of celecoxib on apoptosis in P5 and P1(0.5) cells. A, Western blot analysis of procaspase-3 levels in P5 and P1(0.5) cells at basal conditions and after treatment with 10 or 50 μmol/L celecoxib for 3, 6, and 24 h; results of densitometric analysis are reported in the graph as percentage of control band density. β-Actin was used as a protein loading control; three separate experiments were done and one representative of all is shown. B, P-glycoprotein and procaspase-3 expression at basal conditions, after 72 h transfection with MDR1 siRNA and after treatment with 10 μmol/L celecoxib for 3 and 6 h following 72 h transfection (MDR1 siRNA + celecoxib 10 μmol/L) in P1(0.5) cells. β-Actin was used as a protein loading control; three separate experiments were done and one representative is shown. C, apoptosis of P5 and P1(0.5) cells cultured for 24 h in doxorubicin-free, serum-free medium or treated for 24 h with 10 or 50 μmol/L celecoxib. Apoptotic nuclei were visualized by Hoechst staining and identified by using a fluorescence microscope (original magnification, ×40). Apoptosis was expressed as a percentage of fragmented Hoechst-positive nuclei versus total Hoechst-positive nuclei of three experiments in triplicate. D, apoptosis of P5 and P1(0.5) cells cultured for 24 h in doxorubicin-free, serum-free medium or treated for 24 h with 10 μmol/L celecoxib. Apoptotic cells were stained with Annexin V (i) or by using the TUNEL technique (ii); apoptosis was expressed as percentage of Annexin V– or TUNEL-positive cells versus total cells of three experiments in triplicate. *, P < 0.05 versus control. Comparison was made by one-way ANOVA, followed by Bonferroni post hoc test.

Figure 5.

Effect of celecoxib on apoptosis in P5 and P1(0.5) cells. A, Western blot analysis of procaspase-3 levels in P5 and P1(0.5) cells at basal conditions and after treatment with 10 or 50 μmol/L celecoxib for 3, 6, and 24 h; results of densitometric analysis are reported in the graph as percentage of control band density. β-Actin was used as a protein loading control; three separate experiments were done and one representative of all is shown. B, P-glycoprotein and procaspase-3 expression at basal conditions, after 72 h transfection with MDR1 siRNA and after treatment with 10 μmol/L celecoxib for 3 and 6 h following 72 h transfection (MDR1 siRNA + celecoxib 10 μmol/L) in P1(0.5) cells. β-Actin was used as a protein loading control; three separate experiments were done and one representative is shown. C, apoptosis of P5 and P1(0.5) cells cultured for 24 h in doxorubicin-free, serum-free medium or treated for 24 h with 10 or 50 μmol/L celecoxib. Apoptotic nuclei were visualized by Hoechst staining and identified by using a fluorescence microscope (original magnification, ×40). Apoptosis was expressed as a percentage of fragmented Hoechst-positive nuclei versus total Hoechst-positive nuclei of three experiments in triplicate. D, apoptosis of P5 and P1(0.5) cells cultured for 24 h in doxorubicin-free, serum-free medium or treated for 24 h with 10 μmol/L celecoxib. Apoptotic cells were stained with Annexin V (i) or by using the TUNEL technique (ii); apoptosis was expressed as percentage of Annexin V– or TUNEL-positive cells versus total cells of three experiments in triplicate. *, P < 0.05 versus control. Comparison was made by one-way ANOVA, followed by Bonferroni post hoc test.

Close modal

The inhibition of P-glycoprotein expression by MDR1 siRNA transfection in P1(0.5) cells prevented the activating effect of celecoxib on caspase-3 (Fig. 5B).

Treatment of P1(0.5) cells with 10 μmol/L celecoxib for 24 h significantly increased the percentage of cells with fragmented Hoechst-positive nuclei and the percentage of Annexin V– and TUNEL-positive cells, whereas the same concentration of celecoxib had no effect on P5 cells (Fig. 5C ; and D, i and ii).

Furthermore, treatment with 10 μmol/L celecoxib increased the number of cells in apoptosis in all P-glycoprotein overexpressing cell lines (Supplementary Fig. S1B and S1D, i and ii). The effect of 10 μmol/L celecoxib for 6, 12, 18, and 24 h on the death of P5 and P1(0.5) cells by trypan blue dye exclusion assay was also evaluated. As shown in Fig. 6B, celecoxib significantly increased cell death in P1(0.5) cells compared with doxorubicin-free, serum-free controls, whereas the same concentration of celecoxib had no effect on viability of P5 cells (Fig. 6A). Moreover, preincubation with 20 μmol/L Z-VAD, a universal inhibitor of caspases, abrogated the enhanced cell death induced by celecoxib treatment in MDR-positive cells (Fig. 6B).

Figure 6.

Effect of celecoxib on cell death of (A) P5 and (B) P1(0.5) cells, evaluated by trypan blue exclusion assay. A and B, P5 and P1(0.5) cells were cultured for 24 h in doxorubicin-free, serum-free medium or treated for 6, 12, 18, and 24 h with 10 μmol/L celecoxib before cell count. Cells were also preincubated for 1 h with 20 μmol/L z-VAD before the addition of celecoxib (z-VAD +celecoxib 10 μmol/L). Columns, means of five separate experiments in triplicate, expressed as percentage of dead cells out of the total amount of cells; bars, SE. *, P < 0.05 versus control; **, P < 0.05 versus z-VAD + celecoxib. Comparison was made by two-way ANOVA, followed by Bonferroni post hoc test.

Figure 6.

Effect of celecoxib on cell death of (A) P5 and (B) P1(0.5) cells, evaluated by trypan blue exclusion assay. A and B, P5 and P1(0.5) cells were cultured for 24 h in doxorubicin-free, serum-free medium or treated for 6, 12, 18, and 24 h with 10 μmol/L celecoxib before cell count. Cells were also preincubated for 1 h with 20 μmol/L z-VAD before the addition of celecoxib (z-VAD +celecoxib 10 μmol/L). Columns, means of five separate experiments in triplicate, expressed as percentage of dead cells out of the total amount of cells; bars, SE. *, P < 0.05 versus control; **, P < 0.05 versus z-VAD + celecoxib. Comparison was made by two-way ANOVA, followed by Bonferroni post hoc test.

Close modal

The presence of intrinsic or acquired MDR is the major factor responsible for chemotherapy failure in cancer patients who are undergoing chemotherapy. The MDR phenotype is characterized by the overexpression of P-glycoprotein in plasma membrane that works as a pump to extrude anticancer drugs from cells. However, in addition to the overexpression of P-glycoprotein, the MDR phenotype is associated with other modifications of cell biology that make cancer cells more resistant to many other mechanisms of cell damage (2, 41, 42). We have previously shown that the development of the MDR phenotype is associated with the constitutive expression of COX-2 in hepatocellular carcinoma cell lines (16). P-glycoprotein can regulate the mitochondrion-dependent apoptotic pathway, conferring resistance to a caspase-dependent mechanism of apoptosis induced by antineoplastic agents (23). The antitumor effect of COX-2 inhibitors has been described in recent years although the molecular mechanisms involved remain elusive (43, 44). The relationship between COX-2 and P-glycoprotein suggests that celecoxib, a specific COX-2 activity inhibitor, may improve results of chemotherapy by increasing the sensitivity of tumor cells to anticancer drugs.

In the present study, we investigated the effect of celecoxib on COX-2 and P-glycoprotein expression and on factors involved in the mitochondrial apoptosis pathway, in MDR-positive cell lines, and in their drug-sensitive parental cell clones. To determine whether the possible antitumor effects of celecoxib could be dose dependent, two concentrations of celecoxib were used, the lowest (10 μmol/L) that can be reached in the clinic and another that is 5-fold higher (50 μmol/L; ref. 45). To our surprise, the lower concentration of celecoxib, in addition to the inhibitory effect on COX-2 activity (16), markedly reduced the expression of P-glycoprotein without affecting COX-2 expression, whereas 50 μmol/L celecoxib, although inhibited COX-2 activity, weakly enhanced the expression of P-glycoprotein and COX-2. It is known that the higher concentration of celecoxib inhibits both COX-1 and COX-2 activities (46); thus, it is likely that total inhibition of arachidonic acid metabolism due to celecoxib could be sensed by cells as being damaging and could trigger a cell response with overexpression of COX-2 and P-glycoprotein proteins. These results are in agreement with other studies that have shown a COX-2–mediated regulation of P-glycoprotein expression (18). The observation that only 10 μmol/L celecoxib reduce P-glycoprotein expression suggests a direct inhibitory effect of celecoxib on P-glycoprotein, which has no effect on COX-2 expression in MDR-positive cells transfected with MDR1 siRNA. Moreover, although 10 and 50 μmol/L celecoxib inhibit COX-2 activity, only the lower concentration sharply reduces P-glycoprotein expression without altering COX-2 expression.

The double-face activity of celecoxib, depending on its concentration, on the expression of COX-2 and P-glycoprotein proteins, was also shown for nitric oxide on apoptosis. In fact, NO can either be proapoptotic or antiapoptotic according to its concentration (47). However, this is the first report to document that celecoxib can inhibit the expression of one protein (i.e., P-glycoprotein) at low concentration and, on the contrary, can increase its expression when a higher concentration is involved. It is unknown whether this aspect of the biology of celecoxib can explain the same controversial results observed in clinical trials. Clearly, it is a point that should be kept in mind when planning studies using this drug.

Based on these preliminary data, we decided to investigate the effect of celecoxib at a low concentration, which can be achieved in the serum of patients after standard anti-inflammatory and antineoplastic therapy, rather than using a higher concentration, which is not clinically achievable (45). First, we found that the MDR-positive clone P1(0.5) has a higher expression of antiapoptotic proteins Bcl-xL and Bcl-2 and lower expression of proapoptotic Bax compared with P5 cells. Expressions of Bcl-xL and Bcl-2 seemed to be correlated with that of P-glycoprotein, as evaluated by MDR1 siRNA transfection experiments. These data confirmed what is reported in the literature on the association between P-glycoprotein expression and the abrogation of cytochrome c release in mitochondrial apoptotic pathway (23).

Although it is known that celecoxib induces apoptosis in cultured hepatocellular carcinoma cells through COX-dependent and COX-independent mechanisms (28, 48), the precise function of celecoxib on the apoptotic signaling pathways remains uncertain. Zhang et al. (49) have shown that celecoxib at 50 μmol/L induced apoptosis through a mechanism involving Akt inactivation without altering Bcl-2 and Bcl-xL protein levels in rat cholangiocarcinoma cells. In agreement with them, we found that 50 μmol/L celecoxib did not alter Bcl-xL or Bcl-2 expression (data not shown). Only the 10 μmol/L celecoxib reduced both Bcl-xL or Bcl-2 protein expression in P-glycoprotein–overexpressing cells and remained without effect in the same cells after transfection with MDR1 siRNA. Thus, the inhibitory effect of 10 μmol/L celecoxib on Bcl-xL or Bcl-2 seems to be dependent on the expression of P-glycoprotein because its suppression by the MDR1 siRNA made 10 μmol/L celecoxib completely ineffective.

To assess the role of the activity of P-glycoprotein in this mechanism, experiments were done to test the effect of known P-glycoprotein activity inhibitors, such as verapamil and taurolithocholic acid, on the behavior of Bcl-xL, Bcl-2, and Bax proteins. These two compounds did not modify the expression of P-glycoprotein, procaspase-3, Bcl-xL, Bcl-2, and Bax proteins, ruling out the possibility that P-glycoprotein activity can modulate the expression of these proteins. The proapoptotic effect of celecoxib was further confirmed by the observation that 10 μmol/L celecoxib, although not affecting the expression of the proapoptotic protein Bax, caused its translocation from cytosol to mitochondria in P1(0.5) cells.

These data show that celecoxib restores the release of cytochrome c from the mitochondria when MDR-positive cells are exposed to 10 μmol/L celecoxib. This effect was shown only in the P-glycoprotein–overexpressing cell clones, whereas no effects were observed in no P-glycoprotein–overexpressing cells. Release of cytochrome c into cytosol initiates the activation of caspase-3 and increases the number of apoptotic cells. Moreover, the celecoxib-induced activation of caspase-3 is mediated by P-glycoprotein expression because the procaspase-3 cleavage was abolished when cells were transfected with the MDR1 siRNA. Recently, Mantovani et al. (50) have shown that P-glycoprotein is cleaved in a caspase-3–dependent manner. Considering the findings of the present work together with those by Mantovani et al., it can be hypothesized that the expression of P-glycoprotein blocks cytochrome c release from mitochondria to cytosol and, consequently, the activation of caspase-3. Therefore, the celecoxib-induced decrease in P-glycoprotein expression causes the release of cytochrome c and the activation of caspase-3. This enzyme cleaves P-glycoprotein, priming a caspase-3–mediated amplification of the apoptotic signaling pathways. If this is true, for the first time a self-enhancing mechanism (celecoxib-mediated reduction of P-glycoprotein expression leads to cytochrome c release and to the activation of caspase-3, which, in turn, can reduce P-glycoprotein levels) has been identified and could be considered a step to a no-return pathway for apoptosis in tumor cells. Celecoxib significantly increased the percentage of apoptotic cells, estimated by Hoechst 33258 staining, Annexin V assay, and TUNEL assay; and cell death, estimated by trypan blue exclusion assay, in P1(0.5) cells, without affecting apoptosis and viability of P5 cells. Accordingly, the preincubation of cells with z-VAD, a universal inhibitor of caspases, abrogated the enhanced cell death induced by celecoxib treatment. The recent evidence that P-glycoprotein is expressed and active in mitochondria membrane further contributes to the understanding of molecular mechanisms of these phenomena (37).

In conclusion, this work confirms that celecoxib exerts more than one pharmacologic action on cells. In the model of human hepatocellular carcinoma cells overexpressing the MDR phenotype as well as in other MDR-positive cells, celecoxib exerts a direct effect on the expression of P-glycoprotein and therefore on apoptotic pathways. MDR phenotype–expressing cells are intrinsically resistant to apoptosis; however, for the first time, we show that apoptosis can be restored by a P-glycoprotein–dependent mechanism in MDR-positive cell lines. The use of low doses of COX-2 inhibitors seems to be a promising approach to improving cancer chemotherapy outcome because celecoxib, which directly reduces P-glycoprotein expression, might enhance the susceptibility of tumor cells to apoptosis induced by anticancer drugs.

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

Grant support: Italian Ministry of University, Scientific and Technological Research (Ministero dell'Istruzione, dell'Università e della Ricerca, Progetto Nazionale cofinanziato COFIN N. 2002067115), the University of Florence (R. Mazzanti), and Faculty of Pharmacy of Novara (L. Tessitore).

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 Prof. I.M. Arias (Cell Biology and Metabolism Branch, National Institute of Child Health and Human Development, NIH, Bethesda, MD) for his critical and stimulating discussion on the pathophysiology of liver disease, and Prof. A.M. Vannucchi and Dr. C. Bogani (Department of Hematology, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy) for their technical expertise in flow cytometry.

1
Endicott JA, Ling V. The biochemistry of P-glycoprotein-mediated multidrug resistance.
Annu Rev Biochem
1989
;
58
:
137
–71.
2
Gottesman MM, Ling V. The molecular basis of multidrug resistance in cancer: the early years of P-glycoprotein research.
FEBS Lett
2006
;
580
:
998
–1009.
3
Mazzanti R, Croop JM, Gatmaitan Z, et al. Benzquinamide inhibits P-glycoprotein mediated drug efflux and potentiates anticancer agent cytotoxicity in multidrug resistant cells.
Oncol Res
1992
;
4
:
359
–65.
4
Koga H, Sakisaka S, Ohishi M, et al. Expression of cyclooxygenase-2 in human hepatocellular carcinoma: relevance to tumor dedifferentiation.
Hepatology
1999
;
29
:
688
–96.
5
Smith WL, Garavito RM, DeWitt DL. Prostaglandin endoperoxide H synthases (cyclooxygenases)-1 and -2.
J Biol Chem
1996
;
27
:
33157
–60.
6
Eberhart CE, Dubois RN. Eicosanoids and the gastrointestinal tract.
Gastroenterology
1995
;
109
:
285
–301.
7
O'Neill GP, Ford-Hutchinson AW. Expression of mRNA for cyclooxygenase-1 and cyclooxygenase-2 in human tissues.
FEBS Lett
1993
;
330
:
156
–60.
8
Fosslien E. Biochemistry of cyclooxygenase (COX)-2 inhibitors and molecular pathology of COX-2 in neoplasia.
Crit Rev Clin Lab Sci
2000
;
37
:
431
–502.
9
Dubois RN, Abramson SB, Crofford L, et al. Cyclooxygenase in biology and disease.
FASEB J
1998
;
12
:
1063
–73.
10
McAdam BF, Mardini IA, Habib A, et al. Effect of regulated expression of human cyclooxygenase isoforms on eicosanoid and isoeicosanoid production in inflammation.
J Clin Invest
2000
;
105
:
1473
–82.
11
Murata H, Tsuji S, Tsujii M, et al. Promoter hypermethylation silences cyclooxygenase-2 (Cox-2) and regulates growth of human hepatocellular carcinoma cells.
Lab Invest
2004
;
84
:
1050
–9.
12
McGinty A, Chang YW, Sorokin A, Bokemeyer D, Dunn MJ. Cyclooxygenase-2 expression inhibits trophic withdrawal apoptosis in nerve growth factor-differentiated PC12 cells.
J Biol Chem
2000
;
275
:
12095
–101.
13
Trifan OC, Hla T. Cyclooxygenase-2 modulates cellular growth and promotes tumorigenesis.
J Cell Mol Med
2003
;
7
:
207
–22.
14
Cianchi F, Cortesini C, Fantappiè O, et al. Cyclooxygenase-2 activation mediates the proangiogenic effect of nitric oxide in colorectal cancer.
Clin Cancer Res
2004
;
10
:
2694
–704.
15
Nakano A, Watanabe N, Nishizaki Y, Takashimizu S, Matsuzaki S. Immunohistochemical studies on the expression of P-glycoprotein and p53 in relation to histological differentiation and cell proliferation in hepatocellular carcinoma.
Hepatol Res
2003
;
25
:
158
–65.
16
Fantappiè O, Masini E, Sardi I, et al. The MDR phenotype is associated with the expression of COX-2 and iNOS in a human hepatocellular carcinoma cell line.
Hepatology
2002
;
35
:
843
–52.
17
Lasagna N, Fantappiè O, Solazzo M, et al. Hepatocyte growth factor and inducible nitric oxide synthase are involved in multidrug resistance-induced angiogenesis in hepatocellular carcinoma cell lines.
Cancer Res
2006
;
66
:
2673
–82.
18
Patel VA, Dunn MJ, Sorokin A. Regulation of MDR-1 (P-glycoprotein) by cyclooxygenase-2.
J Biol Chem
2002
;
277
:
38915
–20.
19
Ziemann C, Schafer D, Rudell G, Kahl GF, Hirsch-Ernst KI. The cyclooxygenase system participates in functional mdr1b overexpression in primary rat hepatocyte cultures.
Hepatology
2002
;
35
:
579
–88.
20
Sorokin A. Cyclooxygenase-2: potential role in regulation of drug efflux and multidrug resistance phenotype.
Curr Pharm Des
2004
;
10
:
647
–57.
21
Surowiak P, Materna V, Denkert C, et al. Significance of cyclooxygenase 2 and MDR1/P-glycoprotein coexpression in ovarian cancers.
Cancer Lett
2006
;
235
:
272
–80.
22
Johnstone RW, Cretney E, Smyth MJ. P-glycoprotein protects leukemia cells against caspase-dependent, but not caspase-independent, cell death.
Blood
1999
;
93
:
1075
–85.
23
Kojima H, Endo K, Moriyama H, et al. Abrogation of mitochondrial cytochrome c release and caspase-3 activation in acquired multidrug resistance.
J Biol Chem
1998
;
273
:
16647
–50.
24
Shimizu S, Konishi A, Kodama T, Tsujimoto Y. BH4 domain of antiapoptotic Bcl-2 family members closes voltage-dependent anion channel and inhibits apoptotic mitochondrial changes and cell death.
Proc Natl Acad Sci U S A
2000
;
97
:
3100
–5.
25
Jang MH, Jun do Y, Rue SW, Han K, Park W, Kim YH. Arginine antimetabolite l-canavanine induces apoptotic cell death in human Jurkat T cells via caspase-3 activation regulated by Bcl-2 or Bcl-xL.
Biochem Biophys Res Commun
2002
;
295
:
283
–8.
26
Takahashi M, Saito H, Atsukawa K, Ebinuma H, Okuyama T, Ishii H. Bcl-2 prevents doxorubicin-induced apoptosis of human liver cancer cells.
Hepatol Res
2003
;
25
:
192
–201.
27
Meric JB, Rottey S, Olaussen K, et al. Cyclooxygenase-2 as a target for anticancer drug development.
Crit Rev Oncol Hematol
2006
;
59
:
51
–64.
28
Wu T. Cyclooxygenase-2 in hepatocellular carcinoma.
Cancer Treat Rev
2006
;
32
:
28
–44.
29
Leng J, Han C, Demetris AJ, Michalopoulos GK, Wu T. Cyclooxygenase-2 promotes hepatocellular carcinoma cell growth through Akt activation: evidence for Akt inhibition in celecoxib-induced apoptosis.
Hepatology
2003
;
38
:
756
–68.
30
Fodera D, D'Alessandro N, Cusimano A, et al. Induction of apoptosis and inhibition of cell growth in human hepatocellular carcinoma cells by COX-2 inhibitors.
Ann N Y Acad Sci
2004
;
1028
:
440
–9.
31
Grosch S, Tegeder I, Niederberger E, Brautigam L, Geisslinger G. COX-2 independent induction of cell cycle arrest and apoptosis in colon cancer cells by the selective COX-2 inhibitor celecoxib.
FASEB J
2001
;
15
:
2742
–4.
32
Rahman MA, Dhar DK, Yamaguchi E, et al. Coexpression of inducible nitric oxide synthase and COX-2 in hepatocellular carcinoma and surrounding liver: possible involvement of COX-2 in the angiogenesis of hepatitis C virus-positive cases.
Clin Cancer Res
2001
;
7
:
1325
–32.
33
Cui W, Yu CH, Hu KQ. In vitro and in vivo effects and mechanisms of celecoxib-induced growth inhibition of human hepatocellular carcinoma cells.
Clin Cancer Res
2005
;
11
:
8213
–21.
34
MacNab GM, Alexander JJ, Lecatsas G, Bey EM, Urbanowicz JM. Hepatitis B surface antigen produced by a human hepatoma cell line.
Br J Cancer
1976
;
34
:
509
–15.
35
Riganti C, Miraglia E, Viarisio D, et al. Nitric oxide reverts the resistance to doxorubicin in human colon cancer cells by inhibiting the drug efflux.
Cancer Res
2005
;
65
:
516
–25.
36
Brandon EF, Bosch TM, Deenen MJ, et al. Validation of in vitro cell models used in drug metabolism and transport studies; genotyping of cytochrome P450, phase II enzymes and drug transporter polymorphisms in the human hepatoma (HepG2), ovarian carcinoma (IGROV-1) and colon carcinoma (CaCo-2, LS180) cell lines.
Toxicol Appl Pharmacol
2006
;
211
:
1
–10.
37
Solazzo M, Fantappiè O, Lasagna N, Sassoli C, Nosi D, Mazzanti R. P-gp localization in mitochondria and its functional characterization in multiple drug-resistant cell lines.
Exp Cell Res
2006
;
312
:
4070
–8.
38
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.
39
Wu H, Hait WN, Yang JM. Small interfering RNA-induced suppression of MDR1 (P-glycoprotein) restores sensitivity to multidrug-resistant cancer cells.
Cancer Res
2003
;
63
:
1515
–9.
40
Mazzanti R, Fantappiè O, Kamimoto Y, Gatmaitan Z, Gentilini P, Arias IM. Bile acid inhibition of P-glycoprotein-mediated transport in multidrug-resistant cells and rat liver canalicular membrane vesicles.
Hepatology
1994
;
20
:
170
–6.
41
Mazzanti R, Fantappiè O, Fabrizio P, Relli P. Susceptibility to lipid peroxidation of human hepatocellular carcinoma cell lines with different levels of multiple drug-resistant phenotype.
Lab Invest
1995
;
73
:
419
–23.
42
Breier A, Barancik M, Sulova Z, Uhrik B. P-glycoprotein-implications of metabolism of neoplastic cells and cancer therapy.
Curr Cancer Drug Targets
2005
;
5
:
457
–68.
43
Hashitani S, Urade M, Nishimura N, et al. Apoptosis induction and enhancement of cytotoxicity of anticancer drugs by celecoxib, a selective cyclooxygenase-2 inhibitor, in human head and neck carcinoma cell lines.
Int J Oncol
2003
;
23
:
665
–72.
44
Awara WM, El-Sisi AE, El-Sayad ME, Goda AE. The potential role of cyclooxygenase-2 inhibitors in the treatment of experimentally-induced mammary tumour: does celecoxib enhance the anti-tumour activity of doxorubicin?
Pharmacol Res
2004
;
50
:
487
–98.
45
Dvory-Sobol H, Cohen-Noyman E, Kazanov D, et al. Celecoxib leads to G2/M arrest by induction of p21 and down-regulation of cyclin B1 expression in a p53-independent manner.
Eur J Cancer
2006
;
42
:
422
–6.
46
Gierse JK, Koboldt CM, Walker MC, Seibert K, Isakson PC. Kinetic basis for selective inhibition of cyclooxygenases.
Biochem J
1999
;
339
:
607
–14.
47
Li CQ, Wogan GN. Nitric oxide as a modulator of apoptosis.
Cancer Lett
2005
;
226
:
1
–15.
48
Kern MA, Haugg AM, Koch AF, et al. Cyclooxygenase-2 inhibition induces apoptosis signaling via death receptors and mitochondria in hepatocellular carcinoma.
Cancer Res
2006
;
66
:
7059
–66.
49
Zhang Z, Lai GH, Sirica AE. Celecoxib-induced apoptosis in rat cholangiocarcinoma cells mediated by Akt inactivation and Bax translocation.
Hepatology
2004
;
39
:
1028
–37.
50
Mantovani I, Cappellini A, Tazzari PL, Papa V, Cocco L, Martelli AM. Caspase-dependent cleavage of 170-kDa P-glycoprotein during apoptosis of human T-lymphoblastoid CEM cells.
J Cell Physiol
2006
;
207
:
836
–44.

Supplementary data