Statins are widely used cholesterol-lowering drugs that selectively inhibit the enzyme 3-hydroxy-3-methylglutaryl CoA reductase, leading to decreased cholesterol biosynthesis. Emerging data indicate that statins stimulate apoptotic cell death in several types of proliferating tumor cells, including breast cancer cells, which is independent of its cholesterol-lowering property. The objective here was to elucidate the molecular mechanism(s) by which statins induce breast cancer cell death. Fluvastatin and simvastatin (5–10 μmol/L) treatment enhanced the caspase-3–like activity and DNA fragmentation in MCF-7 cells, and significantly inhibited the proliferation of MCF-7 cells but not MCF-10 cells (noncancerous epithelial cells). Statin-induced cytotoxic effects were reversed by mevalonate, an immediate metabolic product of the acetyl CoA/3-hydroxy-3-methylglutaryl CoA reductase reaction. Both simvastatin and fluvastatin enhanced nitric oxide (·NO) levels which were inhibited by mevalonate. Statin-induced ·NO and tumor cell cytotoxicity were inhibited by 1400W, a more specific inhibitor of inducible nitric oxide synthase (iNOS or NOS II). Both fluvastatin and simvastatin increased iNOS mRNA and protein expression. Stimulation of iNOS by statins via inhibition of geranylgeranylation by GGTI-298, but not via inhibition of farnesylation by FTI-277, enhanced the proapoptotic effects of statins in MCF-7 cells. Statin-mediated antiproliferative and proapoptotic effects were exacerbated by sepiapterin, a precursor of tetrahydrobiopterin, an essential cofactor of ·NO biosynthesis by NOS. We conclude that iNOS-mediated ·NO is responsible in part for the proapoptotic, tumoricidal, and antiproliferative effects of statins in MCF-7 cells. [Cancer Res 2007;67(15):7386–94]

Statins, a class of hyperlipidemic blockbuster drugs, are routinely used for lowering serum cholesterol in the treatment and prevention of hypercholesterolemia and other cardiovascular conditions (15). Statins are competitive inhibitors of 3-hydroxy-3-methylglutaryl CoA (HMG-CoA) reductase, the rate-limiting enzyme in cholesterol biosynthesis. Statins improve endothelial and cardiovascular function by enhancing nitric oxide (·NO) biosynthesis in endothelial cells (68). There are two groups of statins: (a) natural or fermentation-derived: lovastatin (Mevacor), simvastatin (Zocor), and pravastatin (Pravachol); (b) synthetic statins: fluvastatin (Lescol), atorvastatin (Lipitor), cerivastatin (Lipobay), and rosuvastatin (Crestor). Some statins such as simvastatin and lovastatin are more hydrophobic (i.e., lipid-soluble) than others (e.g., cerivastatin). A major biochemical function of statins is the ability to lower lipid levels, in particular, plasma cholesterol, low-density lipoprotein, and triglycerides. However, reports suggest that statins elicit vasoprotective and cardioprotective effects that are independent of their lipid-lowering effects (911). The pleiotropic effects (i.e., actions independent of the cholesterol-lowering property) of statins are dependent on their structural property (i.e., hydrophobicity; ref. 12).

Emerging research suggests that statins may prevent various types of cancers, including breast carcinoma, although results have been mixed (1315). Statins inhibit cancer cell proliferation by arresting the cell cycle at the G1-S phase and inducing apoptosis (16, 17). Recent studies have shown that fluvastatin markedly attenuates epidermal growth factor–induced invasion of pancreatic cancer cells by inhibiting the translocation of RhoA from the cytosol to the membrane (18). Statins have been shown to inhibit in vivo tumor growth at clinically relevant doses and to diminish metastasis in animal models (19, 20). The objective of this study was to determine the molecular mechanisms by which statins exert cytotoxicity in human breast cancer cells. Results from this study revealed that statin-induced inducible NO synthase (iNOS)/NO activity and inhibition of geranylgeranylation play a key role in enhancing MCF-7 cell apoptosis, and that supplementation with sepiapterin exacerbates statin-mediated breast cancer cell death via increased generation of ·NO.

Reagents, cell lines, and culture conditions. Simvastatin, fluvastatin, N-4-[2(R)-amino-3-mercaptopropyl]amino-2-naphthylbenzoyl-(l)-leucine methyl ester (GGTI-298), methyl {N-[2-phenyl-4-N-[2(R)-amino-3-mecaptopropylamino] benzoyl]}-methionate (FTI-277), and 4,5-diaminofluorescein diacetate (DAF-2-DA) were purchased from Calbiochem. Mevalonate, N-(3-aminomethyl)benzylacetamidine (1400W), [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (MTT), squalene, and sepiapterin were purchased from Sigma, Inc. ·NO-fluvastatin (NCX 6553) was purchased from Cayman Chemicals. The culture medium (MEM) and fetal bovine serum (FBS) were purchased from Life Technologies, Inc. All other chemicals were of reagent grade. All cell lines were purchased from the American Type Culture Collection.

MCF-7 and MDA-MB-231 cells were grown in 10% MEM containing 10% FBS, l-glutamine (4 mmol/L), penicillin (100 units/mL), and streptomycin (100 μg/mL), and incubated at 37°C in a humidified atmosphere of 5% CO2 and 95% air.

MTT reduction cytotoxicity assay. MTT was taken up by cells and was reduced to a colored formazan product that could be detected by spectrophotometry (λmax = 562 nm). Reduction of MTT was dependent on the mitochondrial respiratory function, and thus, measures the relative number of viable cells in the culture. After the treatment was completed, MCF-7 cells were washed twice with DPBS, resuspended in 1 mL of MEM without FBS, and incubated with 5 mg/mL of MTT solution for 1 h at 37°C. The medium was removed and the cells were solubilized in DMSO. The absorption was measured at 562 nm with reference at 630 nm.

Thymidine uptake studies. DNA synthesis was measured by monitoring the uptake of tritiated thymidine, [3H]TdR (Perkin-Elmer). Cells (5 × 105/mL) were cultured with different concentrations of simvastatin or fluvastatin (0–10 μmol/L) in the presence or absence of mevalonate (20 μmol/L), 1400W, or sepiapterin. Cells were pulse-chased with [3H]TdR [0.5 μCi (0.185 MBq)/well] during the last 3 h of a 24-h culture, harvested onto glass filters with an automatic cell harvester (Cambridge Technology), and radioactivity on the filters was measured by β-scintillation counting. All experiments were done in triplicate and repeated thrice.

Measurement of intracellular ·NO. Intracellular ·NO levels were monitored using a DAF-2-DA fluorescence probe (21). After the treatments, cells were washed with DPBS and incubated in 2 mL of fresh culture medium without FBS. DAF-2-DA was added at a final concentration of 710 μmol/L, and cells were incubated for 20 min. Cells were washed twice with DPBS and maintained in 1 mL of the culture medium for monitoring the fluorescence using a Nikon fluorescence microscope (excitation, 488 nm; emission, 610 nm) equipped with an FITC filter. Fluorescence intensity was calculated using the Metamorph software.

Nitrite and nitrate measurements. Nitrite and nitrate, the oxidative metabolites of ·NO, were measured by chemiluminescence, using a Sievers apparatus, following reduction with vanadium (III) chloride (22). Briefly, following treatments, the cells were washed thrice with DPBS and then incubated for 30 min at 37°C in 1 mL of Hanks' balanced salt mixture containing 25 μmol/L of l-arginine. The medium was collected and centrifuged for 5 min at 5,000 rpm, and 50 μL of the clear supernatant was used for nitrate and nitrite analysis. Each sample was analyzed in triplicate.

Western blot analysis. After treatment with statins, cells were washed with ice-cold DPBS and resuspended in 150 μL of radioimmune precipitation assay buffer [20 mmol/L Tris-HCl (pH 7.4), 2.5 mmol/L EDTA, 1% Triton X-100, 1% sodium deoxycholate, 1% SDS, 100 mmol/L NaCl, and 100 mmol/L sodium fluoride] containing 1 mmol/L of sodium vanadate, 10 μg/mL of aprotinin, 10 μg/mL of leupeptin, and 10 μg/mL of pepstatin inhibitors. Cells were homogenized by passing the suspension through a 25-gauge needle (20 strokes). The lysate was centrifuged at 750 × g for 10 min at 4°C to pellet out the nuclei. The remaining supernatant was centrifuged for 30 min at 12,000 × g. Protein was determined using the Lowry method and 50 μg of the lysate was used for the Western blot analysis. Proteins were resolved using SDS-polyacrylamide gels and blotted onto nitrocellulose membranes. Membranes were washed with TBS (140 mmol/L NaCl, 50 mmol/L Tris-HCl; pH 7.2) containing 0.1% Tween 20 (TBST) and 5% skimmed milk to block nonspecific protein binding. Membranes were incubated with 1 μg/mL of rabbit anti-iNOS polyclonal antibody (Abcam), mouse anti-cyclin D1 antibody, mouse anti-cyclin E antibody (BD Biosciences) or rabbit anti-p27 antibody (Chemicon International) in TBST for overnight at 4°C, washed five times with TBST, and then incubated with goat anti-rabbit or rabbit anti-mouse IgG horseradish peroxidase–conjugated secondary antibody (1:5,000) for 1.5 h at room temperature. Immunoreactive proteins were detected using the enhanced chemiluminescence method (Amersham Biosciences).

RT-PCR analysis. Following the treatments, the medium was aspirated and 1 mL of TRIzol reagent (Invitrogen) was added and total RNA was extracted using the manufacturer's protocol. Five micrograms of RNA was used for the first-strand cDNA synthesis using a first-strand cDNA synthesis kit (Amersham Biosciences). Four microliters of the cDNA mixture was used to amplify mRNAs from iNOS (5′-CATGGCTTGCCCCTGGAAGTTTCT-3′) and (5′-CCTCTATGGTGCCATCGGGCATC-3′), arginase I (5′-CTCTAAGGGACAGCCTCGAGGA-3′) and (5′-TGGGTTCACTTCCATGATATCTA-3′), arginase II (5′-ATGTCCCTAAGGGGCAGCCTCTCGCGT-3′) and (5′-CACAGCTGTAGCCATCTGACACAGCTC-3′), and eNOS (5′-CCAGCTAGCCAAAGTCACCAT-3′) and (5′-GTCTCGGAGCCATACAGGATT-3′).

Cell cycle analysis. For DNA content analysis, harvested cells were centrifuged at 1,000 × g for 5 min, fixed by the gradual addition of ice-cold 70% ethanol, and washed with PBS. Cells were then treated with RNase (10 μg/mL) for 30 min at 37°C, washed once with PBS, and resuspended and stained in 1 mL of 69 μmol/L of propidium iodide in 38 mmol/L of sodium citrate for 30 min at room temperature. The cell cycle phase distribution was determined by analytic DNA flow cytometry as described previously (23). The percentage of cells in each phase of the cell cycle was analyzed using Modfit software (Verity Software House).

Soft agar assay for colony formation. After cells were treated under various conditions, they were seeded in six-well plates. The plates were first covered with phenol red–free MEM containing 0.6% agar and 10% FBS. The middle layer contained cells (5 × 103) in phenol red–free MEM with 0.35% agar and 10% FBS. The top layer, consisting of the medium, was added to prevent drying of the agar in the plates. The plates were incubated for 21 days, after which the plates were stained in 0.5 mL of 0.005% crystal violet for 1 h and the cultures were inspected and photographed. The colony efficiency was determined by a count of the number of colonies >15 mm in diameter, which was calculated as the average number of colonies counted at 50× magnification in five individual fields manually (24).

Caspase-3–like proteolytic activity. Cells were washed twice in cold DPBS and lysed in buffer containing 10 mmol/L of Tris-HCl, 10 mmol/L of NaH2PO4/Na2HPO4 (pH 7.5), 130 mmol/L of NaCl, 1% Triton, and 10 mmol/L of sodium PPi. Cell lysates were incubated with a caspase-3 fluorogenic substrate N-acetyl-DEVD-7-amido-4-trifluoromethylcoumarin at 37°C for 1 h. 7-Amido-4-trifluoromethylcoumarin liberated from the substrate was measured using a fluorescence plate reader (Perkin-Elmer Life Sciences) with λex = 400 nm and λem = 505 nm (25). The fluorescence intensity was normalized to the protein levels measured with the Bradford protein assay kit (Sigma).

Measurement of apoptosis by terminal deoxynucleotidyl transferase–mediated nick-end labeling assay. The terminal deoxynucleotidyl transferase–mediated nick-end labeling (TUNEL) assay was used for microscopic detection of apoptosis (26). This assay is based on labeling of 3′-free hydroxyl ends of the fragmented DNA with fluorescein-dUTP catalyzed by terminal deoxynucleotidyl transferase. Procedures were followed according to a commercially available kit (ApoAlert) from Clontech. Apoptotic cells exhibit a strong nuclear green fluorescence that could be detected using a standard fluorescein filter (520 nm). All cells stained with propidium iodide exhibit a strong red cytoplasmic fluorescence at 620 nm. The apoptotic cells were detected by fluorescence microscopy equipped with rhodamine and FITC filters. The quantification of apoptosis was done using the Metamorph Image Analysis package.

Statistical analysis. Results were analyzed by a one-way ANOVA, and differences estimated by a Student's t test were considered to be statistically significant at P < 0.05.

Statin-induced MCF-7 cell cytotoxicity. We assessed the effectiveness of simvastatin and fluvastatin to induce cytotoxicity in MCF-7 cells. MCF-7 breast cancer cells were treated with fluvastatin or simvastatin at different concentrations (0.5–20 μmol/L) for 24 to 48 h. The changes in the number of viable cells were determined using the MTT assay, which monitors the intracellular conversion of MTT to formazan spectrophotometrically (λmax = 562 nm). As shown in Fig. 1A, statins potently diminished the number of viable MCF-7 cells. Statins induced cytotoxicity in both MCF-7 breast cancer (malignant) cells (Fig. 1A and B) and MDA-MB-231 (metastatic breast cancer cell lines; data not shown). Fluvastatin and simvastatin did not affect noncancerous mammary epithelial cells, MCF-10A (Fig. 1B). To determine whether statin-induced MCF-7 cell cytotoxicity was due to the inhibition of HMG-CoA reductase activity, cells were pretreated with mevalonate prior to adding simvastatin and fluvastatin. Results show that mevalonate significantly reversed the cytotoxic effects of statins (Fig. 1C), suggesting that the HMG-CoA reductase activity (leading to cholesterol biosynthesis or protein isoprenylation) plays a pivotal role in statin-induced tumor cell cytotoxicity. However, pretreatment with squalene, an immediate precursor of cholesterol biosynthesis, did not prevent statin-induced cytotoxicity (data not shown). This suggests that modulation of isoprenylation of proteins may play a key role in statin-mediated effects in MCF-7 cells.

Figure 1.

Effects of statin and mevalonate on cell death and cell proliferation in MCF-7 and MCF-10A cells. A, MCF-7 cells were treated with simvastatin or fluvastatin (5–10 μmol/L) for 24 to 48 h and cell survival was measured by the MTT assay. B, MCF-7 and MCF-10A cells were treated with simvastatin or fluvastatin (5–10 μmol/L) for a period of 48 h and cell survival was measured by the MTT assay. C, the effect of mevalonate (20 μmol/L) on cell death induced by simvastatin or fluvastatin was determined using the MTT assay. D, the effects of varying concentrations of simvastatin and fluvastatin in the presence or absence of mevalonate (20 μmol/L) on cell proliferation were determined by measuring 3H-thymidine uptake into cells after a 48-h treatment. Columns, means from three different experiments; bars, SD. *, significantly different compared with untreated conditions (P < 0.05); #, significantly different compared with simvastatin or fluvastatin alone (P < 0.05).

Figure 1.

Effects of statin and mevalonate on cell death and cell proliferation in MCF-7 and MCF-10A cells. A, MCF-7 cells were treated with simvastatin or fluvastatin (5–10 μmol/L) for 24 to 48 h and cell survival was measured by the MTT assay. B, MCF-7 and MCF-10A cells were treated with simvastatin or fluvastatin (5–10 μmol/L) for a period of 48 h and cell survival was measured by the MTT assay. C, the effect of mevalonate (20 μmol/L) on cell death induced by simvastatin or fluvastatin was determined using the MTT assay. D, the effects of varying concentrations of simvastatin and fluvastatin in the presence or absence of mevalonate (20 μmol/L) on cell proliferation were determined by measuring 3H-thymidine uptake into cells after a 48-h treatment. Columns, means from three different experiments; bars, SD. *, significantly different compared with untreated conditions (P < 0.05); #, significantly different compared with simvastatin or fluvastatin alone (P < 0.05).

Close modal

To further confirm the loss of cell proliferation (as detected by the MTT assay), we measured DNA synthesis in MCF-7 cells treated with simvastatin or fluvastatin for a period of 24 h and monitored the uptake of 3H-thymidine during the last 3 h of the incubation. As seen in Fig. 1D, both simvastatin and fluvastatin inhibited the uptake of 3H-thymidine, and this inhibition was partially reversed by mevalonate (Fig. 1D).

Role of l-arginine metabolizing enzymes in statin-induced cytotoxicity. As statins are known to protect against endothelial dysfunction by modulating the NOS and ·NO levels in endothelial cells (68), we surmised that statins might also regulate NOS and ·NO levels in MCF-7 cells. To this end, we initially measured the DAF-2–derived green fluorescence. Both simvastatin and fluvastatin significantly increased ·NO-mediated DAF fluorescence (Fig. 2A). To identify the source of ·NO, we initially monitored the eNOS protein levels by Western blotting in MCF-7 cells treated with and without statins, and found no detectable eNOS protein levels in control and treated MCF-7 cells (data not shown). However, quite unexpectedly, iNOS protein and iNOS mRNA levels were up-regulated in cells treated with statins (Fig. 2B). To further confirm that increased expression of iNOS protein corresponds to increased activity, we measured NO2/NO3 levels in the medium. Results indicate that statins increased NO2/NO3 levels (Fig. 2B). Mevalonate suppressed this increase in NO2/NO3 levels (Fig. 2B), indicating that HMG-CoA reductase–dependent pathways regulate iNOS expression and regulation.

Figure 2.

Effects of statins and mevalonate on nitric oxide generation, arginase levels, and cell death in MCF-7 cells. A, cells were treated with simvastatin or fluvastatin (10 μmol/L) in the presence or absence of mevalonate (20 μmol/L) for 48 h and intracellular ·NO was measured by DAF fluorescence as described in Materials and Methods. Columns, DAF fluorescence intensity (as a percentage of control) calculated using Metamorph Image Analysis software. B, inducible NOS mRNA was measured by RT-PCR, protein levels measured by Western analysis, and NO2/NO3 levels were measured as described in Materials and Methods. Cells were treated with simvastatin and fluvastatin (5–20 μmol/L) for 40 h in the presence and absence of mevalonate (20 μmol/L). C, MCF-7 cells were treated with simvastatin or fluvastatin (10 μmol/L) for 40 h in the presence or absence of mevalonate (20 μmol/L) and RT-PCR was done using the gene-specific primers for measuring arginase II transcript levels. D, MCF-7 cells were treated with varying concentrations of fluvastatin or ·NO-fluvastatin (0–1 μmol/L) for a period of 48 h and cell death was analyzed by the MTT assay. Points, means of three independent experiments; bars, SD. *, significantly different compared with untreated conditions (P < 0.05); #, significantly different compared with simvastatin or fluvastatin alone (P < 0.05).

Figure 2.

Effects of statins and mevalonate on nitric oxide generation, arginase levels, and cell death in MCF-7 cells. A, cells were treated with simvastatin or fluvastatin (10 μmol/L) in the presence or absence of mevalonate (20 μmol/L) for 48 h and intracellular ·NO was measured by DAF fluorescence as described in Materials and Methods. Columns, DAF fluorescence intensity (as a percentage of control) calculated using Metamorph Image Analysis software. B, inducible NOS mRNA was measured by RT-PCR, protein levels measured by Western analysis, and NO2/NO3 levels were measured as described in Materials and Methods. Cells were treated with simvastatin and fluvastatin (5–20 μmol/L) for 40 h in the presence and absence of mevalonate (20 μmol/L). C, MCF-7 cells were treated with simvastatin or fluvastatin (10 μmol/L) for 40 h in the presence or absence of mevalonate (20 μmol/L) and RT-PCR was done using the gene-specific primers for measuring arginase II transcript levels. D, MCF-7 cells were treated with varying concentrations of fluvastatin or ·NO-fluvastatin (0–1 μmol/L) for a period of 48 h and cell death was analyzed by the MTT assay. Points, means of three independent experiments; bars, SD. *, significantly different compared with untreated conditions (P < 0.05); #, significantly different compared with simvastatin or fluvastatin alone (P < 0.05).

Close modal

In addition to NOS-mediated oxidation of l-arginine to ·NO and citrulline, l-arginine can also be metabolized by arginases to l-ornithine and urea within the urea cycle and is subsequently converted to polyamines (27). Polyamines are known to increase cell proliferation (28). Because iNOS is significantly induced by statin treatment, it was of interest to measure the levels of arginases (Arg I and Arg II) in statin-treated MCF-7 cells. Arg I transcript levels could not be detected in MCF-7 cells, but Arg II levels were significantly down-regulated in statin-treated cells which was reversed by mevalonate (Fig. 2C). This result suggests a “cross-talk” between arginase and iNOS that plays a role in statin toxicity in MCF-7 cells. As statins increased ·NO levels, we wondered whether N-nitroso-fluvastatin (·NO-fluvastatin) supplementation in MCF-7 would be more effective in causing MCF-7 cell death as compared with fluvastatin alone. ·NO-fluvastatin is a hybrid molecule comprised of both statin and ·NO activities (29). Results show that ·NO-fluvastatin was more potent than fluvastatin alone in causing MCF-7 cell death (Fig. 2D). This clearly implicates a major role for ·NO in statin-induced MCF-7 cell death.

Inhibition of geranylgeranylation by statins induces iNOS expression and cell death in MCF-7 cells. The present data showed that cholesterol-independent pathways were responsible for statin-induced effects. Statins have been reported to deplete the availability of prenylated substrates (30). Posttranslational prenylation of small GTPases by the addition of a geranylgeranyl or farnesyl moiety is critical for cellular localization and signaling activity (31). To further confirm the involvement of isoprenoids on statin-induced, iNOS-dependent cell death, we investigated the effects of isoprenylation inhibitors. Pretreatment of MCF-7 cells with geranylgeranyltransferase inhibitor (GGTI-298), but not farnesyltransferase inhibitor (FTI-277) decreased the number of viable cells, as indicated by the MTT assay (Fig. 3A), and decreased cell proliferation, as indicated by diminished 3H-thymidine uptake (Fig. 3B). To investigate whether inhibition of geranylgeranylation or farnesylation was responsible for enhanced iNOS expression, iNOS protein levels were measured in the presence of either GGTI-298 or FTI-277. As shown, GGTI and not FTI-277 dose-dependently increased iNOS protein levels (Fig. 3C). Concomitantly, inhibition of geranylgeranylation, but not farnesylation, increased NO2/NO3 levels (Fig. 3D). However, treatment of noncancerous mammary epithelial cells with fluvastatin and simvastatin did not induce NO2/NO3 levels (Supplemental Fig. S1). Based on these results, we conclude that GGTI mimics the effects of statins, and therefore, it is likely that statin-mediated iNOS/·NO induction and cytostatic/cytotoxic effects in MCF-7 cells occur through reduced geranylgeranylation of downstream signaling targets, such as Rho or Rac GTPases.

Figure 3.

Effects of geranylgeranyltransferase inhibitor (GGTI-298) and farnesyl inhibitor (FTI-277) on cell death, cell proliferation, and ·NO levels in MCF-7 cells. A, cells were treated with GGTI or FTI for a period of 48 h and cell death was measured by the MTT assay. B, the cells were treated as described in (A), and cell proliferation was determined by measuring 3H-thymidine uptake as described in Materials and Methods. C, MCF-7 cells were treated with GGTI or FTI (10–20 μmol/L) for 40 h and iNOS protein levels were measured by Western immunoblotting analysis. D, the cells were treated as described in (A), and NO2/NO3 levels were measured at the end of the experiment using the ·NO analyzer. Columns, means of three independent experiments; bars, SD. *, significantly different compared with untreated conditions (P < 0.05); #, significantly different compared with FTI treatment alone (P < 0.05).

Figure 3.

Effects of geranylgeranyltransferase inhibitor (GGTI-298) and farnesyl inhibitor (FTI-277) on cell death, cell proliferation, and ·NO levels in MCF-7 cells. A, cells were treated with GGTI or FTI for a period of 48 h and cell death was measured by the MTT assay. B, the cells were treated as described in (A), and cell proliferation was determined by measuring 3H-thymidine uptake as described in Materials and Methods. C, MCF-7 cells were treated with GGTI or FTI (10–20 μmol/L) for 40 h and iNOS protein levels were measured by Western immunoblotting analysis. D, the cells were treated as described in (A), and NO2/NO3 levels were measured at the end of the experiment using the ·NO analyzer. Columns, means of three independent experiments; bars, SD. *, significantly different compared with untreated conditions (P < 0.05); #, significantly different compared with FTI treatment alone (P < 0.05).

Close modal

Contrasting effects of iNOS inhibitor and iNOS activator on statin-induced MCF-7 cell apoptosis. Pretreatment with 1400W, a specific inhibitor of iNOS (32), partially reversed the statin-dependent antiproliferative effects (Fig. 4A) and diminished statin-dependent NO2/NO3 generation (Fig. 4B). Conversely, treatment with sepiapterin (which is a precursor of the NOS cofactor BH4) significantly enhanced the statin-dependent increase in NO2/NO3 levels (Fig. 4B). Sepiapterin also enhanced the GGTI-298–dependent increase in NO2/NO3 levels (compare Figs. 3D with 4C). Sepiapterin treatment alone or in the presence of FTI-277 had no effect on NO2/NO3 levels (Fig. 4B and C). These findings support the model that statin-mediated inhibition of geranylgeranylation causes an iNOS-dependent increase in NO2/NO3 levels, which promotes cell death or loss of cell proliferation. Consistent with this model, we found that 1400W partially suppresses the antiproliferative effects of GGTI-298 (Fig. 4D), just as 1400W partially suppresses the antiproliferative effects of statins (Fig. 4A). Further support for this model is provided by the contrasting effects of 1400W and sepiapterin on statin-induced apoptosis (Fig. 5). Treatment with simvastatin or fluvastatin promotes apoptosis, as indicated by increased caspase-3–like activity in the cells (Fig. 5A) and increased TUNEL staining (Fig. 5B). These statin-induced indicators of apoptosis were diminished by treatment with the iNOS inhibitor 1400W and enhanced by treatment with sepiapterin (Fig. 5A and B). Taken together, these results indicate that ·NO modulation plays a key role in the antiproliferative and proapoptotic effects of statins in tumor cells.

Figure 4.

Effects of 1400W, sepiapterin, and mevalonate on statin-induced cell death and ·NO levels in MCF-7 cells. A, cells were treated with simvastatin or fluvastatin (10 μmol/L) in the presence or absence of a specific iNOS inhibitor, 1400W (10 μmol/L) for 48 h and cell death was measured by the MTT assay. B, the cells were treated as described in (A) or treated with statins in the presence or absence of sepiapterin (50 μmol/L) for 40 h, and NO2/NO3 levels were measured using the ·NO analyzer. C, the cells were treated with GGTI or FTI (20 μmol/L) for 48 h and NO2/NO3 levels were measured at the end of the experiment using the ·NO analyzer. D, the cells were treated with GGTI (5–10 μmol/L) in the presence or absence of 1400W (10 μmol/L) for a period of 48 h and cell proliferation was determined by measuring 3H-thymidine uptake as described in Materials and Methods. Columns, means of at least three independent experiments; bars, SD. *, significantly different compared with untreated conditions (P < 0.05); #, significantly different compared with simvastatin or fluvastatin alone (P < 0.05; A and B) or GGTI alone (D).

Figure 4.

Effects of 1400W, sepiapterin, and mevalonate on statin-induced cell death and ·NO levels in MCF-7 cells. A, cells were treated with simvastatin or fluvastatin (10 μmol/L) in the presence or absence of a specific iNOS inhibitor, 1400W (10 μmol/L) for 48 h and cell death was measured by the MTT assay. B, the cells were treated as described in (A) or treated with statins in the presence or absence of sepiapterin (50 μmol/L) for 40 h, and NO2/NO3 levels were measured using the ·NO analyzer. C, the cells were treated with GGTI or FTI (20 μmol/L) for 48 h and NO2/NO3 levels were measured at the end of the experiment using the ·NO analyzer. D, the cells were treated with GGTI (5–10 μmol/L) in the presence or absence of 1400W (10 μmol/L) for a period of 48 h and cell proliferation was determined by measuring 3H-thymidine uptake as described in Materials and Methods. Columns, means of at least three independent experiments; bars, SD. *, significantly different compared with untreated conditions (P < 0.05); #, significantly different compared with simvastatin or fluvastatin alone (P < 0.05; A and B) or GGTI alone (D).

Close modal
Figure 5.

Effects of 1400W, sepiapterin, and mevalonate on statin-induced caspase-3–like activity and DNA fragmentation. A, the caspase-3–like proteolytic activity was measured in MCF-7 cells treated with simvastatin or fluvastatin (10 μmol/L) for 48 h in the presence or absence of 1400W (10 μmol/L), mevalonate (20 μmol/L), or sepiapterin (50 μmol/L). Cell lysates were incubated with the fluorogenic caspase-3 substrate (DEVD-AFC) for 1 h at 37°C and the released fluorescent active product was measured in a fluorescence spectrophotometer using an excitation/emission of 400/505 nm, respectively. B, cells were treated with simvastatin or fluvastatin (5–10 μmol/L) for 48 h in the presence or absence of 1400W (10 μmol/L), sepiapterin (50 μmol/L), or mevalonate (20 μmol/L) and stained for TUNEL-positive cells as an index of DNA fragmentation monitored by fluorescence microscopy (original magnification, ×100). Photographs are overlaid images of propidium iodide– and FITC-stained cells (arrow, TUNEL-positive cells). Yellow, apoptotic cells; red, nonapoptotic cells.

Figure 5.

Effects of 1400W, sepiapterin, and mevalonate on statin-induced caspase-3–like activity and DNA fragmentation. A, the caspase-3–like proteolytic activity was measured in MCF-7 cells treated with simvastatin or fluvastatin (10 μmol/L) for 48 h in the presence or absence of 1400W (10 μmol/L), mevalonate (20 μmol/L), or sepiapterin (50 μmol/L). Cell lysates were incubated with the fluorogenic caspase-3 substrate (DEVD-AFC) for 1 h at 37°C and the released fluorescent active product was measured in a fluorescence spectrophotometer using an excitation/emission of 400/505 nm, respectively. B, cells were treated with simvastatin or fluvastatin (5–10 μmol/L) for 48 h in the presence or absence of 1400W (10 μmol/L), sepiapterin (50 μmol/L), or mevalonate (20 μmol/L) and stained for TUNEL-positive cells as an index of DNA fragmentation monitored by fluorescence microscopy (original magnification, ×100). Photographs are overlaid images of propidium iodide– and FITC-stained cells (arrow, TUNEL-positive cells). Yellow, apoptotic cells; red, nonapoptotic cells.

Close modal

Effect of statins on cell cycle distribution: role of ·NO. As ·NO has previously been reported to exert tumor cell cycle alterations (33), we investigated the cytostatic effect of statins in MCF-7 cells. MCF-7 cells were treated with simvastatin and fluvastatin for 48 h in the presence and absence of 1400W (10 μmol/L) and mevalonate (20 μmol/L). Cell cycle progression was examined using FACScan flow cytometry analysis. As shown in Table 1, both simvastatin and fluvastatin (5–10 μmol/L) arrested MCF-7 cells in G0/G1 phase, and as a result, the number of cells in the S phase was decreased. Similar effects were observed with ·NO-fluvastatin at a much lower concentration (1 μmol/L) as compared with native fluvastatin (Table 1). Statin-induced cell cycle alterations were partially reversed by the iNOS inhibitor (1400W) and almost completely reversed by mevalonate (Table 1). As cell cycle progression from G0 through G1 phase involves activation of the cell regulatory proteins, cyclins D and E, we investigated the effects of statins and iNOS inhibitor on the cell cycle proteins. As expected, the cell cycle regulatory proteins, cyclin D1 and cyclin E (which are responsible for driving the cell cycle progression from G0/G1 to S phase transition) were significantly decreased with statin treatments and restored in part by 1400W or mevalonate (Fig. 6A). The levels of the cyclin-dependent kinase inhibitor, p27, were also down-regulated by statin treatments (Fig. 6A). Therefore, under our experimental conditions, it seems that the decrease in cell cycle regulatory proteins was independent of the levels of cyclin-dependent kinase (cdk) inhibitor(s) and possibly other cdk-independent regulatory mechanisms are involved.

Table 1.

Cell cycle distribution

TreatmentG0/G1 (%)S (%)G2-M (%)
Control 76.8 9.9 13.3 
Simvastatin (5 μmol/L) 84.6 1.3 14.1 
Simvastatin (10 μmol/L) 85.2 0.0 14.8 
Fluvastatin (5 μmol/L) 83.8 3.1 13.2 
Fluvastatin (10 μmol/L) 86.1 2.5 11.4 
NO-Fluvastatin (1 μmol/L) 85.2 0.9 13.9 
Simvastatin (5 μmol/L) + 1400W 78.2 6.4 15.4 
Fluvastatin (5 μmol/L) + 1400W 77.4 6.0 16.6 
NO-Fluvastatin (1 μmol/L) + 1400W 75.9 6.0 18.2 
Simvastatin (5 μmol/L) + mevalonate 76.0 8.3 15.7 
Fluvastatin (5 μmol/L) + mevalonate 76.1 8.0 15.9 
NO-Fluvastatin (1 μmol/L) + mevalonate 76.5 8.5 15.1 
1400W (10 μmol/L) 75.4 12.2 12.4 
Mevalonate (20 μmol/L) 75.8 9.9 14.2 
TreatmentG0/G1 (%)S (%)G2-M (%)
Control 76.8 9.9 13.3 
Simvastatin (5 μmol/L) 84.6 1.3 14.1 
Simvastatin (10 μmol/L) 85.2 0.0 14.8 
Fluvastatin (5 μmol/L) 83.8 3.1 13.2 
Fluvastatin (10 μmol/L) 86.1 2.5 11.4 
NO-Fluvastatin (1 μmol/L) 85.2 0.9 13.9 
Simvastatin (5 μmol/L) + 1400W 78.2 6.4 15.4 
Fluvastatin (5 μmol/L) + 1400W 77.4 6.0 16.6 
NO-Fluvastatin (1 μmol/L) + 1400W 75.9 6.0 18.2 
Simvastatin (5 μmol/L) + mevalonate 76.0 8.3 15.7 
Fluvastatin (5 μmol/L) + mevalonate 76.1 8.0 15.9 
NO-Fluvastatin (1 μmol/L) + mevalonate 76.5 8.5 15.1 
1400W (10 μmol/L) 75.4 12.2 12.4 
Mevalonate (20 μmol/L) 75.8 9.9 14.2 

NOTE: The cell cycle distribution of MCF-7 cells treated with either simvastatin or fluvastatin (5–10 μmol/L) for 40 h in the presence or absence of 1400W (10 μmol/L) or mevalonate (20 μmol/L) was measured by flow cytometry of propidium iodide–stained cells, as described in Materials and Methods.

Figure 6.

Effects of 1400W and mevalonate on statin-induced cell cycle protein alterations in MCF-7 cells and clonogenic growth in soft agar. A, cells were treated with simvastatin or fluvastatin (10 μmol/L) in the presence or absence of 1400W (10 μmol/L) or mevalonate (20 μmol/L) for 40 h and cyclins D1 and E protein levels were measured by Western immunoblotting analysis using the corresponding polyclonal or monoclonal antibodies. Data are representative of three separate experiments. B, anchorage-independent colony formation of MCF-7 cells treated with simvastatin or fluvastatin (10 μmol/L) in the presence or absence of mevalonate (20 μmol/L), 1400W (10 μmol/L), or sepiapterin (50 μmol/L). Cells were also treated with either GGTI-298 or FTI-277 (10 μmol/L) alone. Treatments were carried out with the abovementioned conditions for 40 h, and the treated cells were seeded onto soft agar plates. After 21 days, colonies were stained with 0.005% crystal violet and viewed under 10× magnification and colonies were counted manually. C, the effect of statins and nitric oxide on cancer cell cycle progression. The check points at which statins (solid lines) and ·NO (dotted lines) induce growth arrest.

Figure 6.

Effects of 1400W and mevalonate on statin-induced cell cycle protein alterations in MCF-7 cells and clonogenic growth in soft agar. A, cells were treated with simvastatin or fluvastatin (10 μmol/L) in the presence or absence of 1400W (10 μmol/L) or mevalonate (20 μmol/L) for 40 h and cyclins D1 and E protein levels were measured by Western immunoblotting analysis using the corresponding polyclonal or monoclonal antibodies. Data are representative of three separate experiments. B, anchorage-independent colony formation of MCF-7 cells treated with simvastatin or fluvastatin (10 μmol/L) in the presence or absence of mevalonate (20 μmol/L), 1400W (10 μmol/L), or sepiapterin (50 μmol/L). Cells were also treated with either GGTI-298 or FTI-277 (10 μmol/L) alone. Treatments were carried out with the abovementioned conditions for 40 h, and the treated cells were seeded onto soft agar plates. After 21 days, colonies were stained with 0.005% crystal violet and viewed under 10× magnification and colonies were counted manually. C, the effect of statins and nitric oxide on cancer cell cycle progression. The check points at which statins (solid lines) and ·NO (dotted lines) induce growth arrest.

Close modal

Effects of statins on anchorage-independent growth of MCF-7 cells. The extent of malignancy of cells corresponds to the attainment of anchorage-independent growth (34). To test the effects of statins on anchorage-independent growth, the long-term effects of statins, GGTI, and FTI on the clonogenic growth of MCF-7 cells were determined. MCF-7 cells were treated with simvastatin or fluvastatin in the presence or absence of either 1400W or mevalonate or sepiapterin. In separate experiments, cells were treated with either GGTI-298 or FTI-277. At the end of the treatments, ∼5 × 103 cells were seeded onto soft agar to determine their clonogenic efficiency after 21 days. Simvastatin and fluvastatin (10 μmol/L) and GGTI-298 (10 μmol/L) but not FTI-277 (10 μmol/L) drastically lowered the visible colony formation in soft agar (Fig. 6B). In the presence of either 1400W or mevalonate, the colony formation was restored in statin-treated cells (Fig. 6B). Sepiapterin supplementation completely inhibited the colony growth at a lower concentration of simvastatin or fluvastatin (5 μmol/L; Fig. 6B). For quantitative estimation of the number of colonies, please refer to Supplemental Fig. S2. Taken together, these results indicate that statins are able to inhibit cell proliferation and anchorage-independent growth of MCF-7 cells by modulating ·NO levels through a pathway involving the inhibition of geranylgeranylation.

In this study, we report the following: (a) statins diminish proliferation and promote apoptosis in MCF-7 breast cancer cells through elevation of inducible NOS expression and ·NO formation from oxidation of l-arginine to l-citrulline using BH4 as a cofactor; (b) supplementation with sepiapterin, a precursor to BH4 biosynthesis, enhanced statin-mediated proapoptotic and antiproliferative effects in MCF-7 cells; (c) statin-mediated tumoricidal effects occur through inhibition of geranylgeranylation and not through inhibition of farnesylation.

Statins as cancer-therapeutic/cancer-preventive agents. Recently, statins have been studied as potential cancer therapeutics. As early as 1992, it was reported that lovastatin treatment diminished tumor cell proliferation in xenografts of pancreatic cancer in nude mice (35). Epidemiologic studies support the effectiveness of statins as anticancer therapeutics (3639). Cancer cells usually exhibit constitutively elevated levels of HMG-CoA reductase and low-density lipid receptor levels to satisfy their increased need for isoprenoids and lipids, and potentially making them more sensitive than normal cells to the isoprenoid-depleting effects of statins (40). The clinical reports on the antitumor effects of statins in breast cancer have yielded mixed results, ranging from no association to both positive and negative associations (41, 42). However, results from a recent study conducted in a large population of postmenopausal women with breast cancer concluded that there exists a significant beneficial association between breast cancer and hydrophobic or lipophilic statin therapy (43). However, when statins were considered together as a class including both hydrophobic and hydrophilic statins, no statistically significant association with breast cancer regression was detected in the same group (43). For instance, the use of pravastatin (hydrophilic statin) did not show any positive effect in association with breast cancer (43). This lack of an in vivo effect of pravastatin was consistent with the results of in vitro cell culture studies in which only hydrophobic statins (e.g., lovastatin) but not hydrophilic statins (e.g., pravastatin) induced significant antiproliferative and antitumor effects in breast cancer cells. These findings have been confirmed by other investigators (17). By and large, the in vitro cell culture experiments are a reasonable predictor of the in vivo antitumor and/or chemopreventive effects of statins in mammary carcinogenesis models (17). Therefore, defining the biological mechanisms by which statins induce apoptosis and exert antiproliferative effects in breast tumor cells is important. The present study shows a novel mechanism by which statins (lipophilic) exert proapoptotic effects in MCF-7 cells.

·NO as a tumoricidal agent. The cytostatic and cytotoxic effects of ·NO in tumor cells are well-established (33, 44, 45). Potential strategies for treating cancer through modulation of ·NO biosynthesis and its downstream signaling pathway have previously been described (45). Inducible NOS activity has been implicated in macrophage-induced antitumor activity. Tumor cells transfected with iNOS gene (NOS II gene) exhibited a less aggressive phenotype in both in vitro and in vivo assays due to increased apoptosis (46). Inducible NOS gene transfection in human lung carcinoma cells was shown to down-regulate the expression of the antiapoptotic protein, survivin (47), leading to enhanced tumor cell death.

The geranylgeranylated members of the Rho family of small GTPases may participate in the statin-mediated increase of ·NO levels in MCF-7 cells. This probability is supported by our finding that GGTI mimics the effects of statins by increasing iNOS expression, increasing ·NO levels, and decreasing the proliferation of the cells. Previous studies indicate that inhibiting isoprenylation of RhoA with statins or GGTI, or inhibiting the RhoA activity with bacterial toxins, increases ·NO levels in a variety of cell types. RhoA can modulate ·NO levels by regulating iNOS expression (48, 49). Thus, the loss of RhoA activity in statin-treated cells may contribute to increased iNOS expression and elevated ·NO levels.

·NO production is compromised by the interplay between NOS, arginases, and arginosuccinate synthetase in tumor cells (27). l-Arginine is a common substrate for both NOS and arginase enzymes. l-Arginine is oxidized by NOS enzymes to l-citrulline and ·NO through the formation of an intermediate, N-hydroxy-l-arginine, which is also a potent competitive inhibitor of arginase. Arginase, both cytosolic and mitochondrial forms, metabolizes l-arginine to l-ornithine, the precursor of polyamines that are essential for cell proliferation (27). Although the Km for arginine is 1,000-fold higher for arginase as compared with the NOS enzymes, the Vmax of arginase is >1,000 times that of the NOS enzymes (27). This suggests similar rates of l-arginine utilization by arginase and NOS enzymes. Thus, ·NO production can be regulated by controlling the availability of l-arginine, by inhibiting the arginase enzyme, or by stimulating the NOS activity. In MCF-7 cells, we were able to detect only Arg II transcript levels, but not Arg I levels, and its levels were significantly decreased by statin treatments.

Sepiapterin enhances statin-mediated proapoptotic effects. BH4 is an indispensable cofactor for ·NO generation by all isoforms of NOS (50). In the absence of BH4, NOS switches its activity from ·NO synthesis to superoxide generation (Supplemental Fig. S2). Inducible NOS transfection of fibroblast cells deficient in BH4 synthesizing enzymatic machinery failed to produce ·NO in the absence of exogenous supplementation with BH4 (50). This clearly shows the critical need for intracellular BH4 for iNOS-dependent generation of ·NO. De novo biosynthesis of BH4 is catalyzed by guanosine-5′-triphosphate (GTP) cyclohydrolase 1. Mammalian cells can also generate BH4 by a “salvage pathway” which converts sepiapterin to BH4 by sepiapterin reductase and dihydrofolate reductase (50). Intracellular BH4 is also manipulated by inhibiting either GTP cyclohydrolase 1 with 2,4-diamino-6-hydroxypyrimidine, or dihydrofolate reductase using methotrexate. Alternatively, treatment of cells with sepiapterin or 7,8-dihydrobiopterin increases BH4. Thus, pharmacologic manipulation of BH4 levels is a good strategy to modulate ·NO in cancer cells. Determining how statins modulate l-arginine metabolism through the arginase and iNOS pathways would likely have a significant effect on our understanding of the stimulation and inhibition of breast cancer growth. As shown in the present study, treatment of MCF-7 cells with sepiapterin enhances the potency of statins via enhanced synthesis of ·NO. As shown in Supplemental Fig. S3, supplementation with drugs activating iNOS activity may synergistically enhance statin-induced antiproliferative and proapoptotic effects in breast cancer cells.

Role of ·NO as a cytostatic agent. Recent research suggests that one of the potential areas of intervention to arrest breast cancer progression is to block the activity of HMG-CoA reductase and lower the levels of mevalonate and other products (39). This pathway controls critical cellular functions including cell signaling and cell cycle progression associated with tumor initiation and tumor metastasis. Cell cycle progression is mainly governed by the activity of a family of protein kinases (cdk). Cyclins were shown to promote cell cycle transitions by binding and activating specific cdks. Cyclin D is involved in the early to mid-G1 phase, whereas cyclin E activation occurs in the late G1 to early S phase (Fig. 6C). The molecular mechanism(s) by which statins induce G1 arrest in breast cancer cells are not known. The present results show that statins inhibit cyclin D and cyclin E expression in MCF-7 cells, and that this inhibition was abrogated by pretreatment with mevalonate and 1400W, a selective inhibitor of iNOS (Fig. 5A). This finding implicates a role for statin-derived ·NO in the modulation of the cell cycle regulatory proteins (Fig. 5A). Previous reports have shown that DETA-NONOate, a nitric oxide donor, induced cytostasis in human breast cancer cells MDA-MB-231, and that the arrest of cells in the G1 phase of the cell cycle was accompanied by cyclin D1 down-regulation (33). However, our results show that although cyclin D1 and cyclin E were significantly down-regulated by statins, it seems that these events were not controlled by cdk inhibitor p27, the levels of which were also decreased with statin treatments. These results were consistent with a recent report showing that the decrease in cell cycle regulatory proteins by statin treatments was not dependent on p21 and p27 protein levels in prostate cancer cells (16).

In conclusion, the present study shows, for the first time, that (a) ·NO generated via the induction of iNOS by selective inhibition of geranylgeranylation plays an important role in statin-induced cell death in MCF-7 breast cancer cells and (b) supplementation with sepiapterin exacerbates the proapoptotic effects of statins in these cells through augmentation of ·NO synthesis.

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

Current address for S. Kotamraju: Discovery Research Division, Institute of Life Sciences, University of Hyderabad Campus Gachibowli, Hyderabad, 500 046 India.

Grant support: George Kress Foundation, Inc. (Green Bay, WI).

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
Watanabe Y, Ito T, Shiomi M, et al. Preventive effect of pravastatin sodium, a potent inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase, on coronary atherosclerosis and xanthoma in WHHL rabbits.
Biochim Biophys Acta
1988
;
960
:
294
–302.
2
Hoeg JM, Brewer HB, Jr. 3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors in the treatment of hypercholesterolemia.
JAMA
1987
;
258
:
3532
–6.
3
Khush KK, Waters DD. Effects of statin therapy on the development and progression of heart failure: mechanisms and clinical trials.
J Card Fail
2006
;
12
:
664
–74.
4
Downs JR, Clearfield M, Weis S, et al. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levels: results of AFCAPS/TexCAPS. Air Force/Texas Coronary Atherosclerosis Prevention Study.
JAMA
1998
;
279
:
1615
–22.
5
Sacks FM, Pfeffer MA, Moye LA, et al. The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and recurrent events trial investigators.
N Engl J Med
1996
;
335
:
1001
–9.
6
Kano H, Hayashi T, Sumi D, et al. A HMG-CoA reductase inhibitor improved regression of atherosclerosis in the rabbit aorta without affecting serum lipid levels: possible relevance of up-regulation of endothelial NO synthase mRNA.
Biochem Biophys Res Commun
1999
;
259
:
414
–9.
7
Hernandez-Perera O, Perez-Sala D, Navarro-Antolin J, et al. Effects of the 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors, atorvastatin and simvastatin, on the expression of endothelin-1 and endothelial nitric oxide synthase in vascular endothelial cells.
J Clin Invest
1998
;
101
:
2711
–9.
8
Laufs U, La Fata V, Plutzky J, Liao JK. Upregulation of endothelial nitric oxide synthase by HMG CoA reductase inhibitors.
Circulation
1998
;
97
:
1129
–35.
9
Rosenson RS. Pluripotential mechanisms of cardioprotection with HMG-CoA reductase inhibitor therapy.
Am J Cardiovasc Drugs
2001
;
1
:
411
–20.
10
Mason RP, Walter MF, Jacob RF. Effects of HMG-CoA reductase inhibitors on endothelial function: role of microdomains and oxidative stress.
Circulation
2004
;
109
:
II34
–41.
11
Tsiara S, Elisaf M, Mikhailidis DP. Early vascular benefits of statin therapy.
Curr Med Res Opin
2003
;
19
:
540
–56.
12
Koga T, Shimada Y, Kuroda M, Tsujita Y, Hasegawa K, Yamazaki M. Tissue-selective inhibition of cholesterol synthesis in vivo by pravastatin sodium, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor.
Biochim Biophys Acta
1990
;
1045
:
115
–20.
13
Muck AO, Seeger H, Wallwiener D. Inhibitory effect of statins on the proliferation of human breast cancer cells.
Int J Clin Pharmacol Ther
2004
;
42
:
695
–700.
14
Maisonneuve P, Lowenfels AB. Statins and the risk of colorectal cancer.
N Engl J Med
2005
;
353
:
952
–4; author reply 952–4.
15
Koyuturk M, Ersoz M, Altiok N. Simvastatin induces apoptosis in human breast cancer cells: P53 and estrogen receptor independent pathway requiring signalling through JNK.
Cancer Lett
2006
;
250
:
220
–8. Cited in PubMed; PMID: 17125918.
16
Sivaprasad U, Abbas T, Dutta A. Differential efficacy of 3-hydroxy-3-methylglutaryl CoA reductase inhibitors on the cell cycle of prostate cancer cells.
Mol Cancer Ther
2006
;
5
:
2310
–6.
17
Campbell MJ, Esserman LJ, Zhou Y, et al. Breast cancer growth prevention by statins.
Cancer Res
2006
;
66
:
8707
–14.
18
Kusama T, Mukai M, Iwasaki T, et al. Inhibition of epidermal growth factor-induced RhoA translocation and invasion of human pancreatic cancer cells by 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors.
Cancer Res
2001
;
61
:
4885
–91.
19
Kusama T, Mukai M, Iwasaki T, et al. 3-Hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors reduce human pancreatic cancer cell invasion and metastasis.
Gastroenterology
2002
;
122
:
308
–17.
20
Paragh G, Kertai P, Kovacs P, Paragh G, Jr., Fulop P, Foris G. HMG CoA reductase inhibitor fluvastatin arrests the development of implanted hepatocarcinoma in rats.
Anticancer Res
2003
;
23
:
3949
–54.
21
Rodriguez J, Specian V, Maloney R, Jourd'heuil D, Feelisch M. Performance of diamino fluorophores for the localization of sources and targets of nitric oxide.
Free Radic Biol Med
2005
;
38
:
356
–68.
22
Pritchard KA, Jr., Ackerman AW, Gross ER, et al. Heat shock protein 90 mediates the balance of nitric oxide and superoxide anion from endothelial nitric-oxide synthase.
J Biol Chem
2001
;
276
:
17621
–4.
23
Vindelov L, Christensen IJ. An integrated set of methods for routine flow cytometric DNA analysis.
Methods Cell Biol
1990
;
33
:
127
–37.
24
Liu S, Sugimoto Y, Sorio C, Tecchio C, Lin YC. Function analysis of estrogenically regulated protein tyrosine phosphatase γ (PTPγ) in human breast cancer cell line MCF-7.
Oncogene
2004
;
23
:
1256
–62.
25
Wang S, Konorev EA, Kotamraju S, Joseph J, Kalivendi S, Kalyanaraman B. Doxorubicin induces apoptosis in normal and tumor cells via distinctly different mechanisms. Intermediacy of H(2)O(2)- and p53-dependent pathways.
J Biol Chem
2004
;
279
:
25535
–43.
26
Kotamraju S, Chitambar CR, Kalivendi SV, Joseph J, Kalyanaraman B. Transferrin receptor-dependent iron uptake is responsible for doxorubicin-mediated apoptosis in endothelial cells: role of oxidant-induced iron signaling in apoptosis.
J Biol Chem
2002
;
277
:
17179
–87.
27
Morris SM, Jr. Enzymes of arginine metabolism.
J Nutr
2004
;
134
:
2743
–7S.
28
Chang C-I, Liao JC, Kuo L. Macrophage arginase promotes tumor cell growth and suppresses nitric oxide-mediated tumor cytotoxicity.
Cancer Res
2001
;
61
:
1100
–6.
29
Ongini E, Impagnatiello F, Bonazzi A, et al. Nitric oxide (NO)-releasing statin derivatives, a class of drugs showing enhanced antiproliferative and anti-inflammatory properties.
Proc Natl Acad Sci U S A
2004
;
101
:
8497
–502.
30
Schafer WR, Kim R, Sterne R, Thorner J, Kim SH, Rine J. Genetic and pharmacological suppression of oncogenic mutations in ras genes of yeast and humans.
Science
1989
;
245
:
379
–85.
31
Kaibuchi K, Kuroda S, Amano M. Regulation of the cytoskeleton and cell adhesion by the rho family GTPases in mammalian cells.
Annu Rev Biochem
1999
;
68
:
459
–86.
32
Garvey EP, Oplinger JA, Furfine ES, et al. 1400W is a slow, tight-binding, and highly selective inhibitor of inducible nitric-oxide synthase in vitro and in vivo.
J Biol Chem
1997
;
272
:
4959
–63.
33
Pervin S, Singh R, Chaudhury G. Nitric oxide-induced cytostasis and cell cycle arrest of a human breast cancer cell line (MDA-MB-231): potential role of cyclin D1.
Proc Natl Acad Sci U S A
2001
;
98
:
3583
–8.
34
Reddig PJ, Juliano RL. Clinging to life: cell to matrix adhesion and cell survival.
Cancer Metastasis Rev
2005
;
24
:
425
–39.
35
Sumi S, Beauchamp RD, Townsend CM, Jr., et al. Inhibition of pancreatic adenocarcinoma cell growth by lovastatin.
Gastroenterology
1992
;
103
:
982
–9.
36
Thibault A, Samid D, Tompkins AC, et al. Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer.
Clin Cancer Res
1996
;
2
:
483
–91.
37
Larner J, Jane J, Laws E, Packer R, Myers C, Shaffrey M. A phase I-II trial of lovastatin for anaplastic astrocytoma and glioblastoma multiforme.
Am J Clin Oncol
1998
;
21
:
579
–83.
38
Kawata S, Yamasaki E, Nagase T, et al. Effect of pravastatin on survival in patients with advanced hepatocellular carcinoma. A randomized controlled trial.
Br J Cancer
2001
;
84
:
886
–91.
39
Demierre MF, Higgins PD, Gruber SB, Hawk E, Lippman SM. Statins and cancer prevention.
Nat Rev Cancer
2005
;
5
:
930
–42.
40
He L, Mo H, Hadisusilo S, Qureshi AA, Elson CE. Isoprenoids suppress the growth of murine B16 melanomas in vitro and in vivo.
J Nutr
1997
;
127
:
668
–74.
41
Bonovas S, Filioussi K, Tsavaris N, Sitaras NM. Statins and breast cancer prevention: time for randomized controlled trials.
J Clin Oncol
2006
;
24
:
2130
–1.
42
Stamm JA, Ornstein DL. The role of statins in cancer prevention and treatment.
Oncology
2005
;
19
:
739
–50.
43
Cauley JA, McTiernan A, Rodabough RJ, et al. Statin use and breast cancer: prospective results from the women's health initiative.
J Natl Cancer Inst
2006
;
98
:
700
–7.
44
Yim CY, McGregor JR, Kwon OD, et al. Nitric oxide synthesis contributes to IL-2-induced antitumor responses against intraperitoneal meth A tumor.
J Immunol
1995
;
155
:
4382
–90.
45
Fukumura D, Kashiwagi S, Jain RK. The role of nitric oxide in tumour progression.
Nat Rev Cancer
2006
;
6
:
521
–34.
46
Le X, Wei D, Huang S, Lancaster JR, Jr., Xie K. Nitric oxide synthase II suppresses the growth and metastasis of human cancer regardless of its up-regulation of protumor factors.
Proc Natl Acad Sci U S A
2005
;
102
:
8758
–63.
47
Chao JI, Kuo PC, Hsu TS. Down-regulation of survivin in nitric oxide-induced cell growth inhibition and apoptosis of the human lung carcinoma cells.
J Biol Chem
2004
;
279
:
20267
–76.
48
Zuckerbraun BS, Barbato JE, Hamilton A, Sebti S, Tzeng E. Inhibition of geranylgeranyltransferase I decreases generation of vascular reactive oxygen species and increases vascular nitric oxide production.
J Surg Res
2005
;
124
:
256
–63.
49
Muniyappa R, Xu R, Ram JL, Sowers JR. Inhibition of rho protein stimulates iNOS expression in rat vascular smooth muscle cells.
Am J Physiol Heart Circ Physiol
2000
;
278
:
H1762
–8.
50
Nichol CA, Smith GK, Duch DS. Biosynthesis and metabolism of tetrahydrobiopterin and molybdopterin.
Annu Rev Biochem
1985
;
54
:
729
–64.

Supplementary data