0013-7227/03/$15.00/0 Printed in U.S.A.
Endocrinology 144(9):3852–3859 Copyright © 2003 by The Endocrine Society doi: 10.1210/en.2003-0098
Lovastatin Induces Apoptosis of Anaplastic Thyroid Cancer Cells via Inhibition of Protein Geranylgeranylation and de Novo Protein Synthesis WEN-BIN ZHONG, CHIH-YUAN WANG, TIEN-CHUN CHANG,
AND
WEN-SEN LEE
Graduate Institute of Physiology (W.-B.Z., W.-S.L.), College of Medicine, National Taiwan University, Taipei 110, Taiwan; Department of Internal Medicine (C.-Y.W.), Far-Eastern Memorial Hospital, Taipei 220, Taiwan; Department of Internal Medicine (C.-Y.W., T.-C.C.), National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei 110, Taiwan; and Graduate Institute of Medical Sciences (W.-S.L.), Taipei Medical University, Taipei 110, Taiwan Lovastatin has been used to treat hypercholesterolemia through blocking the mevalonate biosynthesis pathway. Inhibition of mevalonate synthesis may result in antiproliferation and cell apoptosis. The aim of the present study was to examine the apoptotic effect of lovastatin in human ARO cells and delineate its underlying molecular mechanism. Our results showed that lovastatin dose- and time-dependently induced apoptosis in ARO cells. Pretreatment with cycloheximide dose-dependently suppressed lovastatin-induced apoptosis, suggesting that de novo protein synthesis is required for lovastatin effect on the induction of apoptosis in ARO cells. Treatment of the cells with 50 M lovastatin induced cytochrome c translocation from mitochondria to cytosol; increases in caspase-2, -3, and -9 activity; and poly (ADPribose) polymerase degradation in a time-dependent manner. However, administration of mevalonate or geranylgeraniol,
A
NAPLASTIC THYROID CANCER (ATC) is one of the most aggressive malignant tumors, and patients with ATC have a poor prognosis with a mean survival time of 2– 6 months. Surgery, radiotherapy, or chemotherapy does not improve the survival time of such patients (1–3). Failure of chemotherapy is related to loss of sensitivity for most chemotherapeutic agents, such as doxorubicin and cisplatin (4, 5). Thus, it is important to search for new therapeutic agents more effective for ATC. Apoptosis occurs in various physiological and pathological processes, and it could be induced by activation of Fas signal, UV irradiation, certain chemical reagents, and depletion of cytokines or growth factors. The cellular alternations of apoptosis include loss of membrane asymmetry, chromatin condensation, and DNA fragmentation by DNase activated by caspases (6 – 8). Caspases are cysteine aspartate proteases synthesized as inactive precursor molecules, which could be converted to active heterodimers by proteolytic cleavage. Once cells undergo apoptosis, activation of initiator caspase can sequentially activate effector caspases to perform the proteolysis of specific cellular protein target such as poly
Abbreviations: AMC, 7-Amino-4-methylcoumarin; ATC, anaplastic thyroid cancer; CHX, cycloheximide; DTT, dithiothreitol; FOH, farnesol; FPP, farnesyl pyrophosphate; FTase, farnesyl transferase; GGOH, geranylgeraniol; GGPP, geranylgeranyl pyrophosphate; GGTase, geranylgeranyl transferase; HMG-CoA, 3-OH-3-methyl-glutaryl CoA; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; PARP, poly (ADP-ribose) polymerase; PMSF, phenylmethylsulfonylfluoride.
but not farnesol, dose-dependently prevented lovastatininduced poly (ADP-ribose) polymerase degradation and the occurrence of apoptosis, but treatment with geranylgeranyl transferase inhibitor, GGTI-298, which blocks the geranylgeranylation, induced an increase in the percentage of the apoptotic cells. These data suggest that geranylgeranylation is required for survival of the lovastatin-treated ARO cells. To support this notion, we demonstrate that lovastatin dosedependently decreased the translocation of RhoA and Rac1, but not Ras, from cytosol to membrane fraction. Moreover, the lovastatin-induced translocation inhibitions in RhoA and Rac1 were prevented by mevalonate and geranylgeraniol but not farnesol. In conclusion, our data suggest that lovastatin induced apoptosis in ARO cells by inhibiting protein geranylgeranylation of the Rho family but not farnesylation of the Ras family. (Endocrinology 144: 3852–3859, 2003)
(ADP-ribose) polymerase (PARP) and then lead to elimination of DNA repair function (9). It has been previously reported that apoptosis in thyroid cancer cells can be induced by manumycin A and paclitaxel, 7-hydroxystaurosporine, sodium butyrate and trichostatin A, and amiodarone (10 – 13). In addition, lovastatin was also reported to induce apoptosis in papillary thyroid cancer cells (14). Whether lovastatin can trigger the occurrence of apoptosis of anaplastic thyroid cancer cells remains to be determined. Lovastatin, a 3-OH-3-methyl-glutaryl CoA (HMG-CoA) reductase inhibitor, has been used to treat hypercholesterolemia through blocking the mevalonate biosynthesis pathway. Inhibition of mevalonate synthesis may result in reductions of isoprenoid prenylation, including geranylgeranyl pyrophosphate (GGPP) and farnesyl pyrophosphate (FPP) (15–17). Prenylation of small GTP-binding proteins, such as Ras superfamily proteins, with farnesyl or geranylgeranyl group, is required for their translocation to the plasma membrane and their functions (18, 19). Translocation of the small GTP-binding proteins is related to cell survival, proliferation, adhesion, differentiation, and invasion (20 – 24). These two posttranslational modifications of Ras superfamily are catalyzed by farnesyl transferase (FTase) and type I geranylgeranyl transferase (GGTase I). Protein farnesylation and geranylgeranylation can be selectively inhibited by peptidomimetic inhibitors, FTI-277 and GGTI-298, respectively (25, 26).
3852
Zhong et al. • Lovastatin and Apoptosis in ATC
It has been reported that lovastatin is able to induce either apoptosis or cell cycle arrest depending on the cell types. For instance, lovastatin-induced cell cycle arrest was observed in colon cancer and glioblastoma cells (27, 28), and the occurrence of apoptosis was seen in medulloblastoma (29) and myeloid leukemia cells (30, 31). Recently we also found that lovastatin could induce differentiation of ARO cells (Wang, C. Y., W. B. Zhong, T. C. Chang, S. M. Lai, and Y. F. Tsai, manuscript submitted). Because both farnesylation of Ras family and geranylgeranylation of Rho family have been reported to be involved in proliferation inhibition and the occurrence of apoptosis in different groups (15–17), it is important to determine which pathway is responsible for the lovastatin-induced apoptosis. Accordingly, we attempted to delineate the molecular mechanism underlying of the lovastatin-induced apoptosis in human ARO cells. Materials and Methods Reagents Anticytochrome c, anti-Fas (clone CH-11), and anti-PARP antibodies were obtained from Upstate Biotechnology (Lake Placid, NY). Caspase substrates, caspase inhibitors, GGTI-298, FTI-277, manumycin A, and Clostridium botulinum C3 transferase were obtained from Calbiochem (La Jolla, CA). Fetal bovine serum, culture medium RPMI 1640, penicillin, and streptomycin were obtained from Life Technologies, Inc. (Grand Island, NY). TNF␣ was purchased from Roche Molecular Biochemicals (Vienna, Austria). Y-27632 was purchased from Tocris Cookson Ltd. (Avonmouth, UK). Cycloheximide, actinomycin D, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), mevalonic acid, geranylgeraniol (GGOH), and farnesol (FOH) were purchased from Sigma Chemical Co. (St. Louis, MO). Lovastatin is a gift from Standard Chemical and Pharmacy Co., Ltd. Taiwan.
Cell culture The human ATC cell line, ARO cells (33), was a gift from Dr. Chen, S.D. (Chang Gung Memorial Hospital, Touyuan, Taiwan). ARO cells were cultured in RPMI 1640 medium supplemented with 5% fetal bovine serum, 100 U/ml penicillin, and 100 g/ml streptomycin at 37 C with 5% CO2. They were subcultured twice a week and only those in exponential growth phase were used in these experiments.
DNA fragmentation analysis with agarose gel electrophoresis After treatment with lovastatin, the cells at the indicated time point were harvested and then suspended with lysis buffer containing 20 mm NaCl, 10 mm Tris-HCl (pH 8.0), 25 mm EDTA, 1% sodium dodecyl sulfate, and 1 mg/ml proteinase K for 24 h in a 55 C water bath. Standard phenol/ chloroform/isoamyl alcohol method (25:24:1) was used to remove protein and extract nucleic acid. RNA and digested with RNase A (100 g/ml) for 12 h at 37 C, and DNA concentrations were determined. DNA extracts were electrophoresed on a 2% agarose gel at 50 V for 45 min and visualized with ethidium bromide staining under UV illumination.
Determination of apoptosis by hypodiploid DNA in flow cytometry Apoptosis was assessed according to the percentage of cells with hypodiploid DNA, using the propidium iodide-staining technique. Briefly, the treated cells were washed in PBS and centrifuged, dispersed in 70% ethanol, and stored overnight at ⫺20 C. On the following day, they were washed with PBS and incubated at 37 C for 30 min with PBS containing 1 mg/ml RNase A (Sigma) and 50 g/ml propidium iodide (Sigma) at room temperature in the dark for 1 h. The stained cells were detected using a FACSCalibur flow cytometer (Becton Dickinson, San Jose, CA) and analyzed using the CELLQuest software. Apoptotic nuclei
Endocrinology, September 2003, 144(9):3852–3859 3853
were distinguished by their hypodiploid DNA content from the diploid DNA content of normal nuclei.
Western blot analysis Thirty micrograms of cytoplasmic proteins were electrophoresed on 10% polyacrylamide gels and transferred to nitrocellulose membranes. Immunoblotting was carried out with specific antibodies in PBS with 0.2% Tween 20 (Sigma) and 5% BSA (Sigma). Specific proteins were visualized using the enhanced chemiluminescence method (Amersham Pharmacia Biotech).
Analysis of caspase activity To study the caspase activation profiles in lovastatin-induced apoptosis, we assayed caspase activity using an assay kit (Promega, Madison, WI). This assay is based on the spectrophotometric detection of the 7-amino-4methylcoumarin (AMC) after cleavage from the labeled substrates. Cells were harvested in lysis buffer [25 mm HEPES, 1 mm EGTA, 5 mm EDTA, 5 mm MgCl2, 5 mm dithiothreitol (DTT), 0.01% 3-[(3-cholamidopropyl) dimethyl-ammonio]-1-propane-sulfonate, 10 g/ml pepstatin, 10 g/ml leupeptin, 1 mm phenylmethylsulfonylfluoride (PMSF), pH 7.4]. Cell lysates were clarified by centrifugation at 10,000 ⫻ g for 5 min, and clear lysates containing 50 g protein were incubated with 25 m Ac-YVADAMC (caspase-1 substrate), Ac-VDVAD-AMC (caspase-2 substrate), AcDEVD-AMC (caspase-3 substrate), Ac-IETD-AMC (caspase-8 substrate), or Ac-LEHD-AMC (caspase-9 substrate) at 37 C for 1 h. The released AMC levels were measured using a spectrofluorometer (F-4500, Hitachi, Tokyo, Japan) with excitation at 360 nm and emission at 460 nm.
Cytochrome c release The release of cytochrome c from the mitochondria to the cytosol during lovastatin-induced apoptosis was examined. Mitochondrial and cytosolic fractions were prepared by resuspending cells in ice-cold buffer A (250 mm sucrose, 20 mm HEPES, 10 mm KCl, 1.5 mm MgCl2, 1 mm EDTA, 1 mm EGTA, 1 mm DTT, 17 mg/ml PMSF, 8 mg/ml aprotinin, and 2 mg/ml leupeptin). After chilled for 30 min on ice, the cells were disrupted by 15 strokes with a glass homogenizer. The homogenate was centrifuged twice to remove unbroken cells and nuclei (750 ⫻ g, 10 min, 4 C). The pellet was resuspended in buffer A, which represents the mitochondrial fraction. The supernatant was again centrifuged at 100,000 ⫻ g for 60 min. The supernatant from this final centrifugation step represents the cytosolic fraction, which was electrophoresed on 12% SDS-polyacrylamide gels after determination of protein concentrations with Bio-Rad reagents, transferred onto polyvinyl difluoride membrane and then probed with anti-cytochrome c or antiglycerol-3-phosphate dehydrogenase antibody. The membrane was reacted with peroxidase-conjugated secondary antibody and then developed in the enhanced chemiluminescence (Amersham Biosciences, Buckinghamshire, UK).
PARP cleavage We examined whether lovastatin-treated ARO cells can induce PARP cleavage, which in turn causes apoptosis. After treatment with lovastatin, the cells were lysed in 10 mm HEPES (pH 8.0), 150 mm NaCl, 0.1 mm EDTA, 1% Nonidet P-40, 1 mm DTT, 0.5 mm PMSF, 2.0 g/ml leupeptin, and 2.0 g/ml aprotinin and then incubated on ice for 30 min followed by centrifugation for 20 min at 10,000 ⫻ g. Protein concentrations were determined using Bio-Rad reagents with standard BSA. The protein extracts from lovastatin-treated ARO cells were electrophoresed, transferred, and then probed with anti-PARP monoclonal antibody.
Separation of particulate and cytosolic fractions The cells were washed with cold PBS and lysed by freeze-thawing in ice-cold lysis buffer containing 50 mm HEPES (pH 7.5), 50 mm NaCl, 2 mm EDTA, 1 mm MgCl2, 10 mm sodium fluoride, 1 mm DTT, 10 mg/ml leupeptin, 10 mg/ml aprotinin, and centrifuged at 100,000 ⫻ g for 30 min at 4 C, and the supernatant was collected as the cytosolic fraction. Pellets were homogenized in the above-mentioned lysis buffer containing 2% Triton X-114 and centrifuged at 1000 ⫻ g for 10 min at 4 C. The supernatant was collected as the membrane fraction. The protein concentra-
3854
Endocrinology, September 2003, 144(9):3852–3859
tions in the cytosolic fraction and the membrane fraction were measured and adjusted to the same concentration and then subjected to immunoblotted with antibodies against pan-Ras (clone F132; Santa Cruz Biotechnology, Inc., Santa Cruz, CA), RhoA (clone 119, Santa Cruz Biotechnology), or Rac1 (clone C-14, Santa Cruz Biotechnology).
Zhong et al. • Lovastatin and Apoptosis in ATC
tions of lovastatin for 48 h or 50 m lovastatin for various times, as indicated. The percentage of the hypodiploid cell (G0/G1 subpopulation, an indicator of apoptosis) was increased in lovastatin-treated ARO cells in a dose- (upper panel) and time-dependent manner (lower panel).
MTT assay for cell growth and viability The viability of ARO cells was evaluated using MTT assay. This assay is based on the cleavage of the tetrazolium salt MTT to a dark blue formazan product by mitochondrial dehydrogenase in viable cells. The absorbance of viable cells was measured in a Spectra Microplate Reader (SLT-Labinstruments, Gro¨ dig, Austria) with a test wavelength of 570 nm and a reference wavelength of 630 nm.
Statistical analysis A t test was used to determine the significance between untreated and treated cells, and P ⬍ 0.05 was considered significant.
Results Lovastatin induced apoptosis in ARO anaplastic thyroid cancer cells
To study the lovastatin effect on the occurrence of apoptosis, ARO cells were treated with various concentrations of lovastatin for 48 h. At concentrations of 0 –25 m lovastatin, apoptosis was not observed. However, when lovastatin concentration was increased to 50 m, DNA fragmentation, an indicator of apoptosis, was observed in ARO cells (Fig. 1A). We then examined the DNA content frequency histograms. Figure 1B shows representative DNA content frequency histograms from the ARO cells treated with various concentra-
FIG. 1. Induction of DNA fragmentation in ARO cells by lovastatin. Treatment of ARO cells for 48 h with lovastatin at a concentration of 50 M or higher, the occurrence of DNA fragmentation was observed (A). The lovastatin-induced increase in G0/G1 subpopulation (hypodiploid cells) in ARO cells is in a dose- (B, upper panel) and time- (B, lower panel) dependent manner. The percentage of hypodiploid cell is indicated. Lv, Lovastatin.
De novo protein synthesis is required for the apoptosis induced by lovastatin
We further examined whether RNA transcription or protein synthesis was involved in the lovastatin-induced apoptosis. As shown in Fig. 2A, both cycloheximide (CHX) and actinomycin D can enhance TNF␣- or anti-Fas antibody-induced apoptosis in ARO cells, suggesting that the synthesis of cell survival signaling molecules is required for the ARO cells preventing the TNF␣- or anti-Fas antibody-induced cell death. In contrast, the lovastatin-induced apoptosis in ARO cells was dosedependently prevented by CHX, but not actinomycin D, treatment (Fig. 2B), suggesting that de novo protein synthesis is required for the lovastatin-induced apoptosis in the ARO cells. Lovastatin-induced cytochrome c release and caspase activation
To determine whether the specific caspases were involved in lovastatin-induced apoptosis of ARO cells, we analyzed the enzymatic activity of each caspase in vitro using individual tetrapeptide as the substrate, including Ac-YVADAMC (caspase-1 substrate), Ac-VDVAD-AMC (caspase-2 substrate), Ac-DEVD-AMC (caspase-3 substrate), Ac-IETD-
Zhong et al. • Lovastatin and Apoptosis in ATC
Endocrinology, September 2003, 144(9):3852–3859 3855
a dose-dependent manner (Fig. 4A), suggesting that inhibition of mevalonate synthesis plays an important role in lovastatin-induced apoptosis. To confirm this hypothesis, we applied GGOH and FOH (downstream metabolites of mevalonate) and examined their effect on the lovastatininduced apoptosis. As illustrated in Fig. 4B, the GGOH, which is metabolized to GGPP in the cells, at a concentration of 10 m completely prevented the lovastatin-induced apoptosis in ARO cells. In contrast, the FOH, which is metabolized to FPP in the cells, enhanced the lovastatin-induced apoptosis in ARO cells (Fig. 4C). PARP degradation, an indicator for caspase activation, was also analyzed in cell lysates by Western immunoblotting. Administration of mevalonate or GGOH, but not FOH, prevented PARP degradation (Fig. 4D). Taken together, these findings indicate that inhibition of GGPP synthesis, which is essential for protein geranylgeranylation, occurred in lovastatin-induced apoptosis in ARO cells. Inhibition of geranylgeranylation induces apoptosis in ARO cells
FIG. 2. Requirement of de novo protein synthesis for lovastatininduced apoptosis in ARO cells. Both CHX and ActD enhanced TNF␣ and anti-Fas antibody-induced apoptosis in ARO cells (A). The lovastatin-induced apoptosis in ARO cells was dose-dependently prevented by CHX (cycloheximide), but not ActD (actinomycin D), treatment (B).
To investigate whether lovastatin-induced cell death was due to inhibition of protein geranylgeranylation, we applied GGTI-298 to block the GGTase activity. Treatment of ARO cells with GGTI-298 (10 –30 m) dose-dependently induced an increased population of apoptotic cells (data not shown). These results further supported the hypothesis that inhibition of protein geranylgeranylation can induce apoptosis in ARO cells. Interestingly, administration of FTase inhibitors, FTI-277 and manumycin A, also dose-dependently induced the apoptosis in ARO cells. Taken together, our results indicate that both inhibition of protein geranylgeranylation and farnesylation could trigger the occurrence of apoptosis in ARO cells. However, only geranylgeranylation is involved in the lovastatin-induced apoptosis in ARO cells.
AMC (caspase-8 substrate), or Ac-LEHD-AMC (caspase-9 substrate). Incubation of ARO cells with lovastatin resulted in a time-dependent increase in the Ac-DEVD-AMC and Ac-VDVAD-AMC cleavage activities. The lovastatininduced increases in Ac-DEVD-AMC and Ac-VDVAD-AMC cleavage activities began at 12 h after treatment with 50 m lovastatin and stayed at the high levels even after 48-h treatment (Fig. 3A). The increased Ac-LEHD-AMC cleavage activity also began at 12 h after treatment of lovastatin, stayed at plateau, and then declined at 36 h after treatment. As shown in Fig. 3, B and C, treatment of the ARO cells with 50 m lovastatin induced cytochrome c translocation from the mitochondria into cytoplasm fraction at 12 h after lovastatin treatment and degradation of PARP, the substrate for caspase-3, at 24 h after treatment. That was paralleled with the time course of the induction of caspase activities and apoptosis. These results suggested that lovastatin induced activations of caspases-2, -3, and -9 but not caspases-1 and -8.
Inhibitory effect of lovastatin on translocation of Rho family from cytosol to membrane
Lovastatin-induced apoptosis was prevented by GGOH treatment
It has been demonstrated that C. botulinum C3 exoenzyme catalyzes the specific ADP-ribosylation and inactivates Rho proteins (RhoA, RhoB, and RhoC). To study the role of RhoA in ARO cell survival, we treated ARO cells with C3 exoen-
In the present study, we found that mevalonate could prevent the lovastatin-induced the occurrence of apoptosis in
To investigate whether lovastatin-induced apoptosis was due to exhaustion of intracellular GGPP pool and then blockade of protein geranylgeranylation in ARO, we examined the effect of lovastatin on translocation of small GTP-binding proteins (Ras, RhoA, and Rac1) from cytosol to membrane fraction. Treatment of ARO cells with lovastatin dosedependently decreased the levels of RhoA and Rac1 protein, but not Ras protein, in the membrane fraction (Fig. 5A). The lovastatin-induced translocation inhibition of the Rho family (RhoA and Rac1) was prevented by treatment with mevalonate and GGOH but not by FOH (Fig. 5B). These results suggest that activation of Rho family might be involved in the lovastatin-induced apoptosis in ARO cells. Requirement of RhoA and RhoA kinase for ARO cell survival
3856
Endocrinology, September 2003, 144(9):3852–3859
Zhong et al. • Lovastatin and Apoptosis in ATC
FIG. 3. Lovastatin-induced cytochrome c release and caspase activation. Treatment of ARO cells with lovastatin induced increases in caspase-2, -3, and -9 activities (A). Lovastatin (50 M)induced accumulation of cytochrome c in the cytoplasm was observed beginning at 12 h after treatment (B). The degradation of PARP was noted at 24 h after treatment (C).
zyme (0 –5 g/ml) for 72 h and showed a dose-dependent decrease in cell viability and increase in hypodiploid cell population (Fig. 6A), suggesting that RhoA is important for ARO cell survival. To confirm further, we inhibited the activity of RhoA kinase, a downstream effector of RhoA signaling, and examined the occurrence of apoptosis in ARO cells. As illustrated in Fig. 6B, treatment of ARO cells with a RhoA kinase inhibitor, Y-27632, at a range of concentrations (20 –180 m) for 48 h caused a dose-dependent increase in the percentage of hypodiploid cell, and decrease in cell viability (Fig. 6B). Taken together, these results suggested that RhoA/ RhoA kinase is required for ARO cell survival. Discussion
Formation of the HMG-CoA reductase is a rate-limiting step of the mevalonate pathway. Inhibition of HMG-CoA reductase results in the depletion of mevalonate, a metabolite of HMG-CoA, and its downstream derivatives. Several studies have shown that lovastatin can inhibit mevalonate synthesis, and administration of mevalonate prevents the lovastatin-induced cell death in various cell types, including myeloid leukemia cells, colon cancer cells, medulloblastoma cells, and C6 glial cells (27–31). In the present study, we demonstrated that mevalonate could completely prevent lovastatin-induced cell death in ARO cells, suggesting that the lovastatin-induced cytotoxicity is involved in an inhibition of mevalonate production. Mevalonate is a pivotal component involved in isoprenoid synthetic pathway. Activation of this pathway leads to the formation of sterols, ubiquinone, farnesyl, and geranylgeranyl pyrophosphate (15–17). Both farnesyl and geranylgeranyl pyrophosphate are involved in
protein prenylation. Therefore, lovastatin-induced inhibition of the biosynthesis of other lipid derivatives might also contribute to the antiapoptotic effect of lovastatin in ARO cells. In the present study, we demonstrated that GGOH, a metabolite of mevalonate, prevented the lovastatin-induced the occurrence of apoptosis in ARO cells, suggesting that protein geranylgeranylation is required for the survival of lovastatintreated ARO cells (Fig. 4B). However the lovastatin-induced proliferation inhibition in ARO cells can be prevented by treatment with mevalonate but not GGOH (data no shown), suggesting that there are two different signaling pathways involved in mevalonate effect on the lovastatin-induced cell proliferation regulation and cell apoptosis. It has previously been reported that FPP, a metabolite of mevalonate, can have some degree of protection on lovastatin-induced apoptosis, that might be due, at least in part, to the conversion of GGPP from FPP (30, 34, 35). Our explanation for the discrepancy between the previous reports and our data is that FOH can be converted to FPP to exert its effect on Ras farnesylation and regulation of the cell proliferation. FOH itself, on the other hand, can also enhance the lovastatin-induced proapoptotic effect. Our data showed that administration of FOH did not cause cell death in ARO cells because treatment with FOH alone without lovastatin did not change significantly the cell death. It has been reported that FOH and GGOH can trigger apoptosis in various cancer cell lines. GGOH induced apoptosis in human HL-60 cells through activation of caspase-3 (36), whereas FOH induced apoptosis by suppression of phosphatidylcholine synthesis in human lung cancer cells and HL-60 cells (36, 37). To further examine whether protein geranylgeranylation
Zhong et al. • Lovastatin and Apoptosis in ATC
Endocrinology, September 2003, 144(9):3852–3859 3857
FIG. 4. Prevention of lovastatin-induced apoptosis in ARO cells by geranylgeraniol treatment. Both mevalonate (A) and GGOH (B) prevented lovastatin-induced apoptosis in a dose-dependent manner. FOH (30 M) enhanced the lovastatin-induced apoptosis in ARO cells (C). Both mevalonate and GGOH, but not FOH, prevented PARP from caspase-mediated proteolytic degradation in lovastatin-treated ARO cells (D). Asterisk, Nonspecific bands.
FIG. 5. Lovastatin inhibits translocation of the Rho family from cytosolic fraction to membrane fraction. Treatment of ARO cells with lovastatin dose-dependently decreased the levels of RhoA and Rac1 protein in cytoplasmic membrane fraction (A). The lovastatin-induced inhibition of the translocation of the Rho family (RhoA and Rac1) was prevented by 100 M mevalonate or 30 M GGOH but not 30 M FOH (B).
is required for ARO cell survival, we applied GGTI-298 and FTI-277 (two selective CAAX mimetic inhibitors) to inhibit GGTase and FTase, respectively. Our results showed that in the absence of lovastatin, both GGTI-298 and FTI-277 dose-
dependently induced apoptosis in ARO cells, although the FTI-277 showed less effect, compared with GGTI-298. To confirm that the protein farnesylation is also essential in preventing ARO cells from apoptosis, we conducted the same experiment using manumycin A, a FPP competitor, and showed an apoptotic effect in a degree similar to GGTI-298 effect in ARO cells. Previous studies in lovastatin have been emphasized on the inhibition of Ras farnesylation because overexpression or mutations of Ras were frequently observed in tumors (38, 39). However, recent studies have indicated that inhibition of geranylgeranylation, but not farnesylation, is the main mechanism regulating the lovastatininduced apoptosis. Both geranylgeranylation and farnesylation have been suggested to play an important role in regulating cancer cell survival (30, 40). Consistent with these reports, our results also showed that both geranylgeranylation and farnesylation are required in ARO cell survival, and the prevention of lovastatin-induced apoptosis by GGOH further indicated that protein geranylgeranylation, but not farnesylation, played a crucial role in cellular survival when the lovastatin is present. Exoenzyme C3 transferase, which is specific for Rho proteins (RhoA, RhoB, and RhoC), but not the other Rho family members (such as Rac1 and Cdc42), has been used to evaluate the functions of Rho proteins through ADP-ribosylation to inactivate the Rho proteins. Our study also showed that treatment of ARO cells with C3 exoenzyme resulted in apoptosis within 48 h. The Rho pathway has been suggested to
3858
Endocrinology, September 2003, 144(9):3852–3859
FIG. 6. Inhibition of RhoA/RhoA kinase induces apoptosis in ARO cells. Treatment of ARO cells with a RhoA inhibitor, C3 exoenzyme (A), or a RhoA Kinase inhibitor, Y-27632 (B) caused a dose-dependent increase in the percentage of sub-G0/G1 population (right axis for bar plot) and decreased cell viability (left axis for line plot).
be involved in the regulation of ARO cell survival. The activities of RhoA and Rac1, but not Ras, were suppressed in lovastatin-induced apoptosis of ARO cells (Fig. 5), suggesting that activation of the Rho family prevents the lovastatininduced apoptosis in ARO cells. RhoA kinase, a direct intermediate effector of RhoA, has been implicated in the regulation of RhoA signaling. In the present study, RhoA kinase inhibitor, Y-27632, was used to evaluate the role of RhoA/RhoA kinase signal pathway in the prevention of apoptosis. Treatment of ARO cells with Y-27632 induced apoptosis and decreased cell viability. These findings suggested that RhoA/RhoA kinase pathway is involved in the regulation of survival signaling of ARO cells. However, we cannot rule out that other geranylgeranylated proteins (such as Rac1) may also play important roles in preventing lovastatin-induced apoptosis, since the lovastatin-induced suppression of Rac1 activation was also restored by cotreatment of ARO cells with GGOH or mevalonate. In addition, CHX enhanced the apoptotic effect of TNF␣ or the agonistic Fas antibody in ARO cells. In contrast, CHX prevented the lovastatin-mediated apoptosis in ARO cells, suggesting that there are labile proapoptotic factors involved in lovastatin-mediated death pathway in ARO cells. Pan et al. (41) combined manumycin A and paclitaxel to treat the anaplastic thyroid cancer cells and showed that the enhanced apoptotic effect caused by combined treatment
Zhong et al. • Lovastatin and Apoptosis in ATC
with paclitaxel and manumycin A was due to the release of cytochrome c from mitochondria to cytoplasm and the activations of caspases-3, -8, and -9. In addition, Di Matola et al. (42) and Vitale et al. (43) also reported that lovastatin could induce apoptosis in papillary thyroid cancer cells through inhibition of the protein prenylation. This apoptotic process is CHX sensitive, p53 independent, and involved in activations of caspase-3 and caspase-6 via cytochrome c releasing from mitochondria. In consistent with their findings, we demonstrated that lovastatin-induced apoptosis involved cytochrome c release, caspase-9 activation, and proteolytic cleavage of PARP following the activation of caspase-2 and -3 but not caspase-8 activation. Although our present study demonstrated that lovastatin induced the occurrence of apoptosis in the ARO cells in vitro, the effective apoptosis-inducing concentrations of lovastatin related to tissue concentrations achieved when this drug is used in humans still needs further investigation. Statin (HMG-CoA reductase inhibitors) treatment induced a suppression of tumor growth and the occurrence of apoptosis in vitro and in vivo. The concentrations of statins used in cell culture models are relatively high and are not easily reached in vivo. The dose of lovastatin used for lowing serum cholesterol in animal models is about 1.5–2.5 mg/kg weight, but 0.25–1.0 mg/kg body weight is used in human clinical treatment (44). In a phase I study, on the other hand, the plasma concentrations of lovastatin measured in cancer patients are also relatively low (0.1–3.9 m) (45). Because the myopathy and hepatotoxicity caused by the statin treatment is seldom observed in clinical long-term therapy, the use of lovastatin (even at a dose as high as 80 mg/d) for clinical therapeutic purpose in hypercholesterolemia is quite safe (46, 47). Previous reports, however, indicated that treatment of the patients with statins at the doses used for coronary artery disease prevention could cause a significant reduction of colon cancer incidence (48). Moreover, lovastatin treatment induced cellular differentiation in ARO cells (at a dose of 10 m or less for cell culture experiment) and in anaplastic thyroid cancer cells (at a dose of 80 mg/d for patients in a clinical trial) (32). Accordingly, statins at lower concentrations than those used in the present study may possibly be effective. We believe that the effective doses of statins could be reduced by combined treatment of cancer cells with statins and the wellestablished chemotherapeutic drugs. However, the dose of statins used for cancer therapy needs to be further studied, and the derivatives of statins need to be intensively developed. Moreover, lovastatin could induce the differentiation in the thyroid cancer cells and thereafter made them more sensitive to radioactive iodine therapy, suggesting the clinical potential applications of lovastatin in thyroid cancer treatment. In conclusion, both protein geranylgeranylation and farnesylation are required for ARO cell survival, and lovastatininduced occurrence of apoptosis is through inhibiting protein geranylgeranylation of the Rho family but not farnesylation of the Ras family. In contrast to TNF␣ and Fas signaling mechanisms, lovastatin-induced apoptosis required de novo protein synthesis.
Zhong et al. • Lovastatin and Apoptosis in ATC
Acknowledgments Received January 23, 2003. Accepted May 16, 2003. Address all correspondence and requests for reprints to: Tien-Chun Chang, M.D., Ph.D., Department of Internal Medicine, National Taiwan University Hospital, College of Medicine, 7, Chung-Shan South Road, Taipei 110, Taiwan. E-mail:
[email protected]; or Wen-Sen Lee, Ph.D., Graduate Institute of Medical Sciences, Taipei Medical University, 250 Wu-Hsing Street, Taipei 110, Taiwan. E-mail: wslee@tmu. edu.tw. This work was supported by research grants from the National Science Council of the Republic of China (NSC 90-2314-B0002-253 and NSC 91-2745-P-038-005).
References 1. Tan RK, Finley III RK, Driscoll D, Bakamjian V, Hicks Jr WL, Shedd DP 1995 Anaplastic carcinoma of the thyroid: a 24-year experience. Head Neck 17:41– 48 2. Hadar T, Mor C, Shvero J, Levy R, Segal K 1993 Anaplastic carcinoma of the thyroid. Eur J Surg Oncol 19:511–516 3. Venkatesh YS, Ordonez NG, Schultz PN, Hickey RC, Goepfert H, Samaan NA 1990 Anaplastic carcinoma of the thyroid: a clinicopathologic study of 121 cases. Cancer 66:321–330 4. Tennvall J, Lundell G, Hallquist A, Wahlberg P, Wallin G, Tibblin S 1994 Combined doxorubicin, hyperfractionated radiotherapy, and surgery in anaplastic thyroid carcinoma: report on two protocols. The Swedish Anaplastic Thyroid Cancer Group. Cancer 74:1348 –1354 5. Schlumberger M, Parmentier C, Delisle MJ, Couette JE, Droz JP, Sarrazin D 1991 Combination therapy for anaplastic giant cell thyroid carcinoma. Cancer 67:564 –566 6. Kerr JF, Wyllie AH, Currie AR 1972 Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26:239 –257 7. Raff MC 1992 Social controls on cell survival and cell death. Nature 356: 397– 400 8. Nunez G, Benedict MA, Hu Y, Inohara N 1998 Caspases: the proteases of the apoptotic pathway. Oncogene 17:3237–3245 9. Nagata S 1997 Apoptosis by death factor. Cell 88:355–365 10. Pan J, Xu G, Yeung SC 2001 Cytochrome c release is upstream to activation of caspase-9, caspase-8, and caspase-3 in the enhanced apoptosis of anaplastic thyroid cancer cells induced by manumycin and paclitaxel. J Clin Endocrinol Metab 86:4731– 4740 11. Wang SH, Phelps E, Utsugi S, Baker Jr JR 2001 Susceptibility of thyroid cancer cells to 7-hydroxystaurosporine-induced apoptosis correlates with Bcl-2 protein level. Thyroid 11:725–731 12. Greenberg VL, Williams JM, Cogswell JP, Mendenhall M, Zimmer SG 2001 Histone deacetylase inhibitors promote apoptosis and differential cell cycle arrest in anaplastic thyroid cancer cells. Thyroid 11:315–325 13. Di Matola T, D’Ascoli F, Fenzi G, Rossi G, Martino E, Bogazzi F, Vitale M 2000 Amiodarone induces cytochrome c release and apoptosis through an iodine-independent mechanism. J Clin Endocrinol Metab 85:4323– 4330 14. Di Matola T, D’Ascoli F, Luongo C, Bifulco M, Rossi G, Fenzi G, Vitale M 2001 Lovastatin-induced apoptosis in thyroid cells: involvement of cytochrome c and lamin B. Eur J Endocrinol 145:645– 650 15. Sacks FM, Pfeffer MA, Moye LA, Rouleau JL, Rutherford JD, Cole TG, Brown L, Warnica JW, Arnold JM, Wun CC, Davis BR, Braunwald E 1996 The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. N Engl J Med 335:1001–1009 16. Goldstein JL, Brown MS 1990 Regulation of the mevalonate pathway. Nature 343:425– 430 17. Alberts AW, Chen J, Kuron G, Hunt V, Huff J, Hoffman C, Rothrock J, Lopez M, Joshua H, Harris E, Patchett A, Monaghan R, Currie S, Stapley E, AlbersSchonberg G, Hensens O, Hirshfield J, Hoogsteen K, Liesch J, Springer J 1980 Mevinolin: a highly potent competitive inhibitor of hydroxymethylglutaryl-coenzyme A reductase and a cholesterol-lowering agent. Proc Natl Acad Sci USA 77:3957–3961 18. Gagel RF 1997 An review of molecular abnormalities leading to thyroid carcinogenesis: a 1993 perspective. Stem Cells 15(Suppl 2):7–13 19. Khosravi-Far R, Clark GJ, Abe K, Cox AD, McLain T, Lutz RJ, Sinensky M, Der CJ 1992 J Biol Chem 267:24363–24368 20. Nishida K, Kaziro Y, Satoh T 1999 Anti-apoptotic function of Rac in hematopoietic cells. Oncogene 18:407– 415 21. Spencer ML, Shao H, Andres DA 2002 Induction of neurite extension and survival in pheochromocytoma cells by the Rit GTPase. J Biol Chem 277: 20160 –20168 22. Sears RC, Nevins JR 2002 Signaling networks that link cell proliferation and cell fate. J Biol Chem 277:11617–11620 23. Bouzahzah B, Albanese C, Ahmed F, Pixley F, Lisanti MP, Segall JD, Condeelis J, Joyce D, Minden A, Der CJ, Chan A, Symons M, Pestell RG 2001 Rho family GTPases regulate mammary epithelium cell growth and metastasis through distinguishable pathways. Mol Med 7:816 – 830
Endocrinology, September 2003, 144(9):3852–3859 3859
24. Parsons JT, Martin KH, Slack JK, Taylor JM, Weed SA 2000 Focal adhesion kinase: a regulator of focal adhesion dynamics and cell movement. Oncogene 19:5606 –5613 25. Miquel K, Pradines A, Sun J, Qian Y, Hamilton AD, Sebti SM, Favre G 1997 GGTI-298 induces G0 –G1 block and apoptosis whereas FTI-277 causes G2-M enrichment in A549 cells. Cancer Res 57:1846 –1850 26. Lerner EC, Zhang TT, Knowles DB, Qian Y, Hamilton AD, Sebti SM 1997 Inhibition of the prenylation of K-Ras, but not H- or N-Ras, is highly resistant to CAAX peptidomimetics and requires both a farnesyl transferase and a geranylgeranyl transferase I inhibitor in human tumor cell lines. Oncogene 15:1283–1288 27. Wachtershauser A, Akoglu B, Stein J 2001 HMG-CoA reductase inhibitor mevastatin enhances the growth inhibitory effect of butyrate in the colorectal carcinoma cell line Caco-2. Carcinogenesis 1061–1067 28. Bouterfa HL, Sattelmeyer V, Czub S, Vordermark D, Roosen K, Tonn JC 2000 Inhibition of Ras farnesylation by lovastatin leads to downregulation of proliferation and migration in primary cultured human glioblastoma cells. Anticancer Res 20:2761–2771 29. Macaulay RJ, Wang W, Dimitroulakos J, Becker LE, Yeger H 1999 Lovastatininduced apoptosis of human medulloblastoma cell lines in vitro. J Neurooncol 42:1–11 30. Van De Donk NW, Kamphuis MM, Lokhorst HM, Bloem AC 2002 The cholesterol lowering drug lovastatin induces cell death in myeloma plasma cells. Leukemia 16:1362–1371 31. Xia Z, Tan MM, Wong WWL, Dimitroulakos J, Minden MD, Penn LZ 2001 Blocking protein geranylgeranylation is essential for lovastatin-induced apoptosis of human acute myeloid leukemia cells. Leukemia 15:1398 –1407 32. Deleted in proof 33. Fagin JA, Matsuo K, Karmakar A, Chen DL, Tang SH, Koeffler HP 1993 High prevalence of mutations of the p53 gene in poorly differentiated human thyroid carcinomas. J Clin Invest 91:179 –184 34. Guijarro C, Blanco-Colio LM, Ortego M, Alonso C, Ortiz A, Plaza JJ, Diaz C, Hernandez G, Egido J 1998 3-Hydroxy-3-methylglutaryl coenzyme a reductase and isoprenylation inhibitors induce apoptosis of vascular smooth muscle cells in culture. Circ Res 83:490 –500 35. Goldstein JL, Brown MS 1990 Regulation of the mevalonate pathway. Nature 343:425– 430 36. Anthony ML, Zhao M, Brindle KM 1999 Inhibition of phosphatidylcholine biosynthesis following induction of apoptosis in HL-60 cells. J Biol Chem 274:19686 –19692 37. Miquel K, Pradines A, Terce F, Selmi S, Favre G 1998 Competitive inhibition of choline phosphotransferase by geranylgeraniol and farnesol inhibits phosphatidylcholine synthesis and induces apoptosis in human lung adenocarcinoma A549 cells. J Biol Chem 273:26179 –26186 38. Bos JL 1989 Ras oncogenes in human cancer: a review. Cancer Res 49:4682– 4689 39. Danesi R, McLellan CA, Myers CE 1995 Specific labeling of isoprenylated proteins: application to study inhibitors of the post-translational farnesylation and geranylgeranylation. Biochem Biophys Res Commun 206:637– 643 40. Bouterfa HL, Sattelmeyer V, Czub S, Vordermark D, Roosen K, Tonn JC 2000 Inhibition of Ras farnesylation by lovastatin leads to downregulation of proliferation and migration in primary cultured human glioblastoma cells. Anticancer Res 20:2761–2771 41. Pan J, Xu G, Yeung SC 2000 Cytochrome c release is upstream to activation of caspase-9, caspase-8, and caspase-3 in the enhanced apoptosis of anaplastic thyroid cancer cells induced by manumycin and paclitaxel. J Clin Endocrinol Metab 86:4731– 4740 42. Di Matola T, D’Ascoli F, Luongo C, Bifulco M, Rossi G, Fenzi G, Vitale M 2001 Lovastatin-induced apoptosis in thyroid cells: involvement of cytochrome c and lamin B. Eur J Endocrinol 145:645– 650 43. Vitale M, Di Matola T, Rossi G, Laezza C, Fenzi G, Bifulco M 1999 Prenyltransferase inhibitors induce apoptosis in proliferating thyroid cells through a p53-independent CrmA-sensitive, and caspase-3-like protease-dependent mechanism. Endocrinology 140:698 –704 44. Agarwal B, Rao CV, Bhendwal S, Ramey WR, Shirin H, Reddy BS, Holt PR 1999 Lovastatin augments sulindac-induced apoptosis in colon cancer cells and potentiated chemopreventive effects of sulindac. Gastroenterology 117:838 – 847 45. Thibault A, Samid D, Tompkins AC, Figg WD, Cooper MR, Hohl RJ, Trepel J, Liang B, Patronas N, Venzon DJ, Reed E, Myers CE 1996 Phase I study of lovastatin, an inhibitor of the mevalonate pathway, in patients with cancer. Clin Cancer Res 2:483– 491 46. Plosker GL, Wagstaff AJ 1996 Fluvastatin: a review of its pharmacology and used in the management of hypercholesterolemia. Drug 51:433– 459 47. Davigono J, Hanefeld M, Nakaya N, Hunninghake DB, Insull W, Ose L 1998 Clinical efficacy and safety of cerivastatin: summary of pivotal phase IIb/III studies. Atherosclerosis 139(Suppl 1):S15–S22 48. Sacks FM, Pfeffer MA, Moye LA, Rouleau JL, Cole TG, Brown L, Warnica JW, Arnold JM, Wun CC, Davis BR, Braunwald E 1996 The effect of pravastatin on coronary events after myocardial infraction in patients with average cholesterol levels. N Engl J Med 335:1001–1009