Molecular mechanism underlying the anticancer effect of simvastatin on MDA‑MB‑231 human breast cancer cells

  • Authors:
    • Yuan‑Yuan Shen
    • Yuan Yuan
    • Ying‑Ying Du
    • Yue‑Yin Pan
  • View Affiliations

  • Published online on: March 4, 2015     https://doi.org/10.3892/mmr.2015.3411
  • Pages: 623-630
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Breast carcinoma is the leading cause of cancer‑associated mortality in female individuals worldwide. Previous studies have investigated the pro‑apoptotic and antimetastatic effects of statins, and have demonstrated that simvastatin exhibits antitumor activity and potent chemopreventive effects. However, the mechanism underlying the effects of simvastatin in breast cancer remains to be elucidated. The present study demonstrated that simvastatin inhibited the proliferation of MDA‑MB‑231 human breast cancer cells in a dose‑dependent manner, decreased the protein expression of B cell lymphoma 2 (Bcl‑2) and increased the protein expression of Bcl‑2‑associated X protein in time‑ and dose‑dependent manners. In addition, simvastatin arrested cells in the G0/G1 phase of the cell cycle, downregulated the protein expression levels of cyclin D1 and cyclin‑dependent kinase (CDK)2, mediated the mitochondria‑dependent caspase cascade by increasing the protein expression levels of caspase‑3, ‑8 and ‑9, and downregulated the protein expression of X‑linked inhibitor of apoptosis, which induced cell apoptosis. In addition, simvastatin decreased the protein expression of matrix metalloproteinase (MMP)‑2 and suppressed the activation of nuclear factor (NF)‑κB in the MDA‑MB‑231 cells. Taken together, these results demonstrated that the antitumor effect of simvastatin in the human MDA‑MB‑231 breast cancer cell line was via the inhibition of cell proliferation, affecting the cell cycle, downregulating the expression levels of cyclin D1 and CDKs, inducing apoptosis and decreasing the expression of MMP‑2, possibly by inhibiting the activation of NF‑κB. Statin treatment may provide a novel therapeutic approach for the treatment of breast cancer.

Introduction

Breast cancer is the most common type of cancer in females and is the predominant cause of female cancer-associated mortality worldwide, accounting for 23% of the cases of newly diagnosed cancer and 14% of the total cancer-associated mortality (1). Current therapies using endocrine agents, particularly selective estrogen receptor (ER) modulators and, more recently, aromatase inhibitors, have successfully prevented or treated ER-positive breast cancer by interfering with estrogen signaling or production (2). However, these drugs have been observed to reduce the incidence of breast cancer by only 50% and had no effect in preventing ER-negative breast cancer, which accounts for 30% of all cases of breast cancer in North America (3,4). ER negativity is frequently combined with high grade tumors and the proliferation and overexpression of human epidermal growth factor receptor (HER)2/neu, resulting in a poor prognosis (5,6). Therefore, effective novel drugs with different molecular structures from conventional chemotherapeutic agents, which may aid in the prevention or treatment of ER-negative breast cancer require development.

Statins lower serum cholesterol levels by inhibiting 3-hydroxy-3-methyl glutaryl coenzyme A (HMG-CoA) reductase, the rate-determining enzyme in the mevalonate pathway (7). This pathway produces various end products, including cholesterol, dolichol, ubiquinone, isopentenyladenine, geranylgeranyl pyrophosphate and farnesyl pyrophosphate, which are critical for normal cellular functions, including cell proliferation, differentiation and survival, in normal and cancerous cells (8,9). Statins are currently used as cholesterol-lowering medications and exhibit effectiveness in the primary and secondary prevention of heart disease and stroke (10). In addition, statins interest for their use in cancer prevention has increased. The anticancer function of statins is based on preclinical evidence of their antiproliferative, pro-apoptotic and antiangiogenic properties. A growing body of evidence suggests that various statins possess anti-proliferative, anti-invasive, antimetastatic and pro-apoptotic properties in various types of cancer cell (1114). Simvastatin, one of the HMG-CoA reductase inhibitors, is currently used as a safe and well-tolerated therapeutic agent for the treatment of hypercholesterolemia, atherosclerosis and stroke (15). Simvastatin demonstrates in vitro and in vivo antitumor actions in several human malignancies including those of the breast, colon and prostate, which has been attributed to cell cycle arrest, thereby inhibiting cell proliferation and inducing apoptotic and necrotic cell death (11,12,16). A previous study revealed that the use of simvastatin, a highly lipophilic statin, reduced the risk of recurrence in Danish females with Stage I–III breast cancer, with 10 fewer cases per 100 females over 10 years (17). In addition, patients with breast cancer on long-term statin treatment have proportionately fewer ER/progesterone (PR)-negative tumors, which are of a lower grade and stage compared with patients who have never received statin treatment (18). By contrast, Bonovas et al concluded from a meta-analysis of seven randomized and nine observational breast cancer trials, that treatment with statins failed to significantly affect the risk of breast cancer (19). However, the efficacy and the molecular mechanism underlying the effects of simvastatin on breast cancer progression remain to be elucidated.

The requirement for alternative therapeutic strategies is increasing and these findings, in parallel with limited knowledge of the affect of simvastatin on breast cancer, led to the present study evaluating its potential for therapeutic effects in breast cancer, as the antitumor and cancer chemopreventive effects of statins on breast cancer require further investigation. The present study hypothesized that statin therapy may reduce the progress of breast carcinoma by inhibiting cell proliferation, altering the cell cycle, inducing apoptosis, down-regulating the protein expression levels of cyclin D1 and cyclin dependent kinases (CDKs) and decreasing the expression of matrix metalloproteinase (MMP)-9. MDA-MB-231 cells were used in vitro to confirm this.

Materials and methods

Cell culture

The human MDA-MB-231 breast cancer cell line was kindly provided by the Laboratory of Molecular Biology of Anhui Medical University (Anhui, China). The cells (1×105/ml) were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Hyclone, Logan, UT, USA), supplemented with 10% heat inactivated fetal bovine serum, 100 U/ml penicillin, 100 μg/ml streptomycin and 2 mM L-glutamine, which were all purchased from Sijiqing Biological Engineering Materials (Hangzhou, China), and maintained in a humidified atmosphere of 5% CO2 at 37°C.

Drugs

Simvastatin (Sigma-Aldrich, St. Louis, MO, USA) was dissolved in dimethylsulfoxide (DMSO; Sigma-Aldrich) at a stock concentration of 1 mM and stored at −20°C. The final concentrations of simvastatin were 3.125, 6.25, 12.5, 25, 50, 100 μM. The final concentration of DMSO in the DMEM was maintained at <0.1%. An equal volume of solvent was added to cells as a control.

Cell proliferation assay

The proliferation rate of the MDA-MB-231 cells was evaluated using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT; Sigma-Aldrich) assay. The exponentially growing cells were plated at a density of 1×104 cells/well into 96-well plates, cultured overnight at 37°C and subsequently treated with various concentrations of simvastatin for 72 h. Following incubation with simvastatin for 72 h, 20 μl MTT solution (5 mg/ml) was added to each well and the plates were incubated for a further 4 h at 37°C. The colored formazan product was dissolved using 150 μl DMSO. The 96-well plates were then placed on a shaker for 10 min at room temperature to thoroughly dissolve the MTT product. The half maximal inhibitory concentration (IC50) value was determined as the concentration resulting in 50% cell growth inhibition following 72 h exposure to simvastatin compared with the untreated control cells. Six replicate wells were used for each drug concentration and the assessment was performed independently in triplicate.

Cell cycle analysis by flow cytometry

The cells (2×105/well) were plated into 6-well dishes and treated with simvastatin at the IC50 concentration (7.979±0.201 μM) for 72 h. The adherent cells were harvested by trypsinization (Sijiqing Biological Engineering Materials), washed twice with phosphate-buffered saline (PBS; Sijiqing Biological Engineering Materials) and fixed overnight in 70% ethanol (Sijiqing Biological Engineering Materials) at 4°C. The ethanol was removed and the cells were washed twice in PBS, prior to being resuspended in 1 ml propidium iodide (PI; Sigma-Aldrich)/Triton X-100 (Sigma-Aldrich) staining solution, containing PBS, 0.1% Triton X-100, 200 μg/ml RNAse A (Sigma-Aldrich) and 50 μg/ml PI in the dark for 30 min at 37°C. The cell cycle was measured by flow cytometry (BD Biosciences, San Jose, CA, USA) and analyzed using Cell Quest WinMDI 2.9 software (BD Biosciences). The cell cycle profiles, including the G0/G1, S and G2/M phases, were calculated using ModFit LT™ 4.0 software.

Flow cytometric analysis of apoptosis

The cells were plated in the exponential growth phase into six-well plates, allowed to attach overnight at 37°C and treated with simvastatin at the IC50 concentration for 72 h. Following treatment, the adherent and floating cells were collected, washed twice with precooled (4°C) PBS and resuspended in 400 μl binding buffer (10 mM HEPES/NaOH pH 7.4; 140 mM NaCl; KCl; MgCl2; and 2.5 mM CaCl2). The cells were incubated with 5 μl annexin V-fluorescein isothiocyanate (BestBio, Shanghai, China) at room temperature in the dark for 15 min and then with 10μl PI (40 μg/ml) at room temperature in the dark for 5 min. The cell suspensions were transferred to flow cytometric analysis tubes and detected using flow cytometry. Cells without drug treatment were used as a control.

Western blotting

The MDA-MB-231 cells, growth with or without simvastatin, were washed with ice-cold PBS solution and scraped in lysis buffer (50 mM Tris-HCl pH7.4; 250 mM NaCl; 0.5% Triton X100; 10% glycerol; 1 mM dichlorodiphenyltrichloroethane; and 1 mM phenylmethylsulfonyl fluoride). The lysates were centrifuged at 16,853 × g for 30 min at 4°C and the supernatant was collected. Briefly, the protein concentration of each sample was determined using a Bicinchoninic Acid Protein Assay kit (Beyotime Institute of Biotechnology, Inc., Shanghai, China). Equal quantities of protein from each sample were loaded onto 10% SDS-polyacrylamide minigels (HyClone Laboratories, Inc.) and electrophoresed. The proteins were transblotted onto polyvinylidene difluoride (PVDF) membranes (Millipore, Billerica, MA, USA) and then blocked with a solution of PBS, containing 5% non-fat milk and 0.1% Tween 20 (HyClone Laboratories, Inc.) for 2 h. The PVDF membranes were probed with specific primary antibodies against anti-B cell lymphoma-2 (Bcl-2; rabbit monoclonal; 1:2,000; Cell Signaling Technologies, Inc., Danvers, MA, USA), anti-Bcl-2 associated X protein (Bax; rabbit monoclonal; 1:2,000; Cell Signaling Technologies, Inc.), rabbit X-linked inhibitor of the apoptosis protein antibody (Xiap; rabbit monoclonal; 1:500; Santa Cruz Biotechnology, Inc., Carlsbad, CA, USA), anti-cyclin D1 (1:1,000; rabbit monoclonal; Abcam, Cambridge, MA, USA), rabbit CDK2 (1:1,000; rabbit monoclonal; Abcam), anti-caspase-3 (1:1,000; mouse monoclonal; Abcam), caspase-8 (1:1,000; mouse monoclonal; Abcam) and caspase-9 (1:1,000; mouse monoclonal; Abcam), rabbit MMP-2 antibody (1:1,500; rabbit monoclonal; Cell Signaling Technologies, Inc.), rabbit nuclear factor-κB antibody (NF-κB p65; mouse monoclonal; 1:1,500; Cell Signaling Technologies, Inc.) and anti-β-actin (1:1,500; rabbit monoclonal; Cell Signaling Technologies, Inc.). Following washing with Tris-buffered saline (Cell Signaling Technologies, Inc.), containing 0.1% Tween-20 three times, the PVDF membranes were incubated with horseradish peroxidase-conjugated secondary antibody (1:5,000) at room temperature for 1 h. Positive bands were detected using enhanced chemilluminescence reagents (Millipore) and β-actin was used as a loading control.

Statistical analysis

The data were analyzed from three independent experiments and are expressed as the mean ± standard deviation. One-way analysis of variance and Student’s t-test were performed to determine the statistical significance of any differences between the control and treatment groups. All statistical analyses were performed using GraphPad Prism 5.0 software (GraphPad, Inc., La Jolla, CA, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Dose-dependent antiproliferative effects of simvastatin in the human breast cancer cell line

The effects of simvastatin on the proliferation of MDA-MB-231 cells were determined using an MTT assay. The MDA-MB-231 cells were treated with different doses of simvastatin (3.125–100 μM) for 72 h. A dose-dependent decrease in the cell viability was observed following treatment with simvastatin, exhibiting an IC50 value of 7.979±0.201 μM following exposure for 72 h (Fig. 1).

Effects of simvastatin treatment on Bcl-2 and Bax

The Bcl-2 and Bax family are important in the regulation of apoptosis, proliferation and invasion of tumor cells (20). The present study examined the protein expression levels of Bcl-2 and Bax using western blot analysis. To assess the effects of time and dose on response, the MDA-MB-231 cells were cultured with different concentrations of simvastatin for different durations. The most marked effects were observed following treatment with simvastatin at the IC50 for 72 h (P<0.05). In addition, treatment with 20 μm simvastatin significantly downregulated the protein expression of Bcl-2 and upregulated the protein expression of Bax in the MDA-MB-231 cells compared with the other concentrations (P<0.05). These results demonstrated that the effect of simvastatin on MDA-MB-231 cells occurred in a time- and dose-dependent manner (Fig. 2).

Cell cycle effects of simvastatin

The cell cycle distribution of the cells exposed to simvastatin at the IC50 concentration for 72 h was assessed by flow cytometry. The percentage of G0/G1 phase cells was markedly increased following simvastatin treatment compared with the control group (P<0.05; Fig. 3A). This finding suggested that simvastatin arrested the cells at the G0/G1 phase of the cell cycle, which may be a mechanism underlying its antitumor effect. In addition, the cell cycle checkpoint proteins, cyclin D1 and CDK2, which are associated with distributional change, were also assessed. The protein expression levels of cyclin D1 and CDK were markedly decreased following pretreatment with simvastatin for 72 h, and occurred in a dose-dependent manner (P<0.05; Fig. 3B).

Effects of simvastatin on cell apoptosis

To examine whether the observed growth inhibition was caused by increased apoptosis, the present study investigated the apoptotic response of the MDA-MD-231 cell line treated with the IC50 of simvastatin using an annexin V/PI assay. As shown in Fig. 4A, the apoptotic rates induced by simvastatin in the MDA-MB-231 cells after 72 h were 9.54%. Furthermore, protein expression of caspase-3, -8 and -9 was detected following treatment with the IC50 concentration of simvastatin for 72 h. Notably, the protein expression levels of caspase-3, -8 and -9 were significantly increased in the simvastatin-treated MDA-MB-231 cells compared with the control group (P<0.05; Fig. 4B). These results demonstrated that simvastatin activated the caspase cascade reaction and was, therefore, important in the apoptotic response of MDA-MB-231 cells. In addition, Xiap is important in the regulation of tumor cell apoptosis. The present study measured the protein expression of Xiap in the MDA-MB-231 cells and found that simvastatin significantly downregulated the protein expression of Xiap (P<0.05; Fig. 4C).

Simvastatin suppresses the expression of MMP-2 and the activation of NF-κB in MDA-MB-231 cells

The protein expression of MMP-2 was examined by western blot analysis, which revealed that treatment with simvastatin decreased the protein expression of MMP-2 in a dose-dependent manner (P<0.05). p65 is a major component of NF-κB, and the levels of NF-κB p65 were also examined. Following co-culture with simvastatin at the IC50 for 72 h, the expression of NF-κB p65 was significantly suppressed in the simvastatin-treated group (P<0.05). The results demonstrated that simvastatin suppressed the expression of MMP-2 and the activation of NF-κB in the MDA-MB-231 cells (Fig. 5).

Discussion

Breast cancer is one of the most life-threatening types of cancer among female individuals worldwide (1). Statins are widely used cholesterol-lowering drugs, and the use of statins has been observed to significantly lower the risk of cancer (7,17). Although an increasing quantity of evidence suggests that statins may have useful activity in breast cancer prevention and/or therapy (21), the molecular mechanisms underlying the neoplastic development and progression of statins in the breast remain to be elucidated. In the present study, the effect of simvastatin on MDA-MB-231 breast cancer cells was observed and the underlying mechanisms were investigated.

The present study demonstrated that simvastatin significantly inhibited the proliferation of the breast cancer cells. The acceleration of the cell cycle is an initial factor in tumor growth, and control of cell cycle progression in cancer cells is a potentially effective strategy for the control of tumor growth (22,23). The results revealed that simvastatin arrested the cells at the G1/S cell cycle transition and directly induced G1/S phase arrest in the MDA-MB-231 cells. Cell cycle progression is regulated by CDKs and cyclin-dependent kinase inhibitors, whose activity is highly controlled and coordinated by their association with cyclins (24). CDK inhibitors interact with active CDK-cyclin complexes and exert tumor-suppressive functions that downregulate cell cycle progression (25,26). A previous study demonstrated that simvastatin induced cell cycle arrest at G0/G1 by downregulating the expression of CDKs and cyclins, which was accompanied by apoptosis and reduced cell proliferation (16). The present study demonstrated that simvastatin significantly decreased the protein expression levels of cyclin D1 and CDK2 in breast tumor cells, which revealed that simvastatin-induced cell cycle arrest at G0/G1 was associated with downregulation in the protein expression levels of cyclin D1 and CDK2. This may be a direct mechanism of simvastatin against the growth of breast cancer cells.

Apoptosis is a fundamental cellular activity and is crucial for eliminating genetically damaged cells, which is key in the pathogenesis of cancer, and the proteins associated with this process have been a focus of interest in investigations of cancer onset and progression (27). The upregulation of pro-apoptotic gene expression and downregulation of anti-apoptotic gene expression induce the initiation of apoptosis, and the progression of cancer depends predominantly on the balance between pro-apoptotic proteins, including Bax, and anti-apoptotic proteins, including Bcl-2 (20). Several studies have suggested that the survival or death of human breast cancer cells is determined by an altered balance between pro-apoptotic and anti-apoptotic proteins, including the ratio of Bcl-2 to Bax (28,29). Increased expression levels of mitochondrial anti-apoptotic proteins contribute to augmented survival of several types of cancer cells, including breast cancer (30). The present study demonstrated that simvastatin increased the expression of Bax and downregulated the expression of Bcl-2, suggesting that the simvastatin-induced apoptosis of MDA-MB-231 cells was associated with modulation of the expression levels of Bax and Bcl-2.

Caspases are also involved in the execution of apoptosis associated with these two signaling pathways. Bcl-2 and Bax activate the caspase cascade reaction and are important in the regulation of the intrinsic pathway of apoptosis (31). As demonstrated in previous studies, Bcl-2 prevents the activation of caspase-3 in response to a variety of apoptotic signals (32,33). In the caspase family, either caspase-8 or -9 and the subsequent effector, caspase-3, are crucial in the apoptotic process and initiation of a caspase cascade triggers the proteolytic activation of executioner caspases, including caspase-3, to perform the final steps in the apoptotic process (34). Inhibition of the expression of Xiap promotes the development of apoptosis, therefore, Xiap suppresses apoptosis through the inhibition of caspases (35,36). In the present study, simvastatin increased the expression levels of caspase-3, -8 and -9, and downregulated the expression of Xiap. This suggested that the simvastatin-induced MDA-MB-231 apoptosis was also associated with activation of the caspase signaling pathway and modulation of the expression of Xiap.

The MMP family consists of 23 zinc-dependent endopeptidases, which are all involved in the degradation of the extracellular matrix. MMPs are upregulated in almost every type of cancer and their expression is often associated with a poor prognosis for patients (37). Based on their unique ability to degrade gelatinases, a major constituent of the basement membrane, MMP-2 and MMP-9, are the most important MMPs involved in tumor invasion and metastasis (38). It has been reported that the expression levels and activities of MMPs are associated with an advanced stage of breast cancer, increased invasion of tumor cells and building of metastatic formations (39). Atorvastatin, a member of the statin drug family, suppresses the expression levels of MMP-2 and MMP-9 in human endothelial cells (40). In addition, it also inhibits the RhoA-JNK-c-Jun-MMP2 cascade, resulting in a decrease in osteosarcoma cell invasion (41). The present study demonstrated that simvastatin inhibited the expression levels of MMPs, potentially inhibiting the invasion and metastasis of breast cancer.

The NF-κB complex, an essential cell mediator, is composed of a family of inducible transcription factors, expressed in almost all cell types (42). The overexpression of NF-κB implies an aggressive tumor in breast cancer and can predict tumors, which are likely to have a poor prognosis (43). A previous study revealed that the expression of NF-κB is necessary for the maintenance of the malignant phenotype, and provides a therapeutic approach for the treatment of cancer (44). The activation of NF-κB upregulates the expression of the anti-apoptotic protein, Bcl-2, and regulates the expression levels of cyclin D1 and MMPs (45,46). In the present study, simvastatin inhibited the expression of NF-κB, and this may be an important mechanism underlying the anticancer effects of simvastatin in breast cancer.

There is increasing interest in cancer prevention and in the drugs that, used in low doses, either alone or in combination, which have different modes of action and low toxicity, act as chemopreventive agents (47). Therefore, the present study investigated other molecules, which are used for the treatment of well-known pathological diseases and have effects on cancer cell proliferation. Statins sensitize cancer cells to chemotherapeutic drugs, and evidence indicates that treatment with simvastatin increases the antitumor activity of cisplatin and docetaxel, common chemotherapeutic agents used against a wide range of types of cancer (48). Therefore, the present study demonstrated the antiproliferative and anticarcinogenic effects of simvastatin in a breast cancer cell line, the results of which suggested that simvastatin may be promising as a therapeutic approach for the treatment of cancer.

Acknowledgments

This study was supported by the Anhui Provincial Science and Technology Agency Foundation of China (nos. 1301042214, 12070403072 and KJ2012A157). The authors would like to thank Dr Yuan Yuan, The Central Laboratory of Binhu Hospital and The Third Affiliated Hospital of Anhui Medical University, for their assistance.

References

1 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Williams C and Lin CY: Oestrogen receptors in breast cancer: basic mechanisms and clinical implications. Ecancermedicalscience. 7:3702013.PubMed/NCBI

3 

Fisher B, Costantino JP, Wickerham DL, et al: Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst. 90:1371–1388. 1998. View Article : Google Scholar : PubMed/NCBI

4 

Vogel VG, Costantino JP, Wickerham DL, et al: Effects of tamoxifen vs raloxifene on the risk of developing invasive breast cancer and other disease outcomes: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 295:2727–2741. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Baqai T and Shousha S: Oestrogen receptor negativity as a marker for high-grade ductal carcinoma in situ of the breast. Histopathology. 42:440–447. 2003. View Article : Google Scholar : PubMed/NCBI

6 

Provenzano E, Hopper JL, Giles GG, Marr G, Venter DJ and Armes JE: Biological markers that predict clinical recurrence in ductal carcinoma in situ of the breast. Eur J Cancer. 39:622–630. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Istvan ES and Deisenhofer J: Structural mechanism for statin inhibition of HMG-CoA reductase. Science. 292:1160–1164. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Zhang FL and Casey PJ: Protein prenylation: molecular mechanisms and functional consequences. Annu Rev Biochem. 65:241–269. 1996. View Article : Google Scholar : PubMed/NCBI

9 

Jackson SM, Ericsson J and Edwards PA: Signaling molecules derived from the cholesterol biosynthetic pathway. Subcell Biochem. 28:1–21. 1997.PubMed/NCBI

10 

Hebert PR, Gaziano JM, Chan KS and Hennekens CH: Cholesterol lowering with statin drugs, risk of stroke, and total mortality. An overview of randomized trials. JAMA. 278:313–321. 1997. View Article : Google Scholar : PubMed/NCBI

11 

Kochuparambil ST, Al-Husein B, Goc A, Soliman S and Somanath PR: Anticancer efficacy of simvastatin on prostate cancer cells and tumor xenografts is associated with inhibition of Akt and reduced prostate-specific antigen expression. J Pharmacol Exp Ther. 336:496–505. 2011. View Article : Google Scholar

12 

Ghosh-Choudhury N, Mandal CC, Ghosh Choudhury N and Ghosh Choudhury G: Simvastatin induces derepression of PTEN expression via NFkappaB to inhibit breast cancer cell growth. Cell Signal. 22:749–758. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Sassano A and Platanias LC: Statins in tumor suppression. Cancer Lett. 260:11–19. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Pelaia G, Gallelli L, Renda T, et al: Effects of statins and farnesyl transferase inhibitors on ERK phosphorylation, apoptosis and cell viability in non-small lung cancer cells. Cell Prolif. 45:557–565. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Goldstein JL and Brown MS: Regulation of the mevalonate pathway. Nature. 343:425–430. 1990. View Article : Google Scholar : PubMed/NCBI

16 

Relja B, Meder F, Wilhelm K, Henrich D, Marzi I and Lehnert M: Simvastatin inhibits cell growth and induces apoptosis and G0/G1 cell cycle arrest in hepatic cancer cells. Int J Mol Med. 26:735–741. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Ahern TP, Pedersen L, Tarp M, et al: Statin prescriptions and breast cancer recurrence risk: a Danish nationwide prospective cohort study. J Natl Cancer Inst. 103:1461–1468. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Kumar AS, Benz CC, Shim V, Minami CA, Moore DH and Esserman LJ: Estrogen receptor-negative breast cancer is less likely to arise among lipophilic statin users. Cancer Epidemiol Biomarkers Prev. 17:1028–1033. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Bonovas S, Filioussi K, Tsavaris N and Sitaras NM: Use of statins and breast cancer: a meta-analysis of seven randomized clinical trials and nine observational studies. J Clin Oncol. 23:8606–8612. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Adams JM and Cory S: The Bcl-2 apoptotic switch in cancer development and therapy. Oncogene. 26:1324–1337. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Riemsma R, Forbes CA, Kessels A, et al: Systematic review of aromatase inhibitors in the first-line treatment for hormone sensitive advanced or metastatic breast cancer. Breast Cancer Res Treat. 123:9–24. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Mork CN, Faller DV and Spanjaard RA: A mechanistic approach to anticancer therapy: targeting the cell cycle with histone deacetylase inhibitors. Curr Pharm Des. 11:1091–1104. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Meeran SM and Katiyar SK: Cell cycle control as a basis for cancer chemoprevention through dietary agents. Front Biosci. 13:2191–2202. 2008. View Article : Google Scholar :

24 

Murray AW: Recycling the cell cycle: cyclins revisited. Cell. 116:221–234. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Graña X and Reddy EP: Cell cycle control in mammalian cells: role of cyclins, cyclin dependent kinases (CDKs), growth suppressor genes and cyclin-dependent kinase inhibitors (CKIs). Oncogene. 11:211–219. 1995.PubMed/NCBI

26 

Pavletich NP: Mechanisms of cyclin-dependent kinase regulation: structures of Cdks, their cyclin activators, and Cip and INK4 inhibitors. J Mol Biol. 287:821–828. 1999. View Article : Google Scholar : PubMed/NCBI

27 

Lefranc F, Facchini V and Kiss R: Proautophagic drugs: a novel means to combat apoptosis-resistant cancers, with a special emphasis on glioblastomas. Oncologist. 12:1395–1403. 2007. View Article : Google Scholar

28 

Korsmeyer SJ, Shutter JR, Veis DJ, Merry DE and Oltvai ZN: Bcl-2/Bax: a rheostat that regulates an anti-oxidant pathway and cell death. Semin Cancer Biol. 4:327–332. 1993.PubMed/NCBI

29 

Oltvai ZN, Milliman CL and Korsmeyer SJ: Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell. 74:609–619. 1993. View Article : Google Scholar : PubMed/NCBI

30 

Renault TT and Chipuk JE: Death upon a kiss: mitochondrial outer membrane composition and organelle communication govern sensitivity to BAK/BAX-dependent apoptosis. Chem Biol. 21:114–123. 2014. View Article : Google Scholar :

31 

Fang Y, Elahi A, Denley RC, Rao PH, Brennan MF and Jhanwar SC: Molecular characterization of permanent cell lines from primary, metastatic and recurrent malignant peripheral nerve sheath tumors (MPNST) with underlying neurofibro-matosis-1. Anticancer Res. 29:1255–1262. 2009.PubMed/NCBI

32 

Boulakia CA, Chen G, Ng FW, et al: Bcl-2 and adenovirus E1B 19 kDA protein prevent E1A-induced processing of CPP32 and cleavage of poly(ADP-ribose) polymerase. Oncogene. 12:529–535. 1996.PubMed/NCBI

33 

Chinnaiyan AM, Orth K, O’Rourke K, Duan H, Poirier GG and Dixit VM: Molecular ordering of the cell death pathway. Bcl-2 and Bcl-xL function upstream of the CED-3-like apoptotic proteases. J Biol Chem. 271:4573–4576. 1996.PubMed/NCBI

34 

Zou H, Henzel WJ, Liu X, Lutschg A and Wang X: Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell. 90:405–413. 1997. View Article : Google Scholar : PubMed/NCBI

35 

Dai Y, Qiao L, Chan KW, et al: Loss of XIAP sensitizes rosiglitazone-induced growth inhibition of colon cancer in vivo. Int J Cancer. 122:2858–2863. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Sasaki H, Sheng Y, Kotsuji F and Tsang BK: Down-regulation of X-linked inhibitor of apoptosis protein induces apoptosis in chemoresistant human ovarian cancer cells. Cancer Res. 60:5659–5666. 2000.PubMed/NCBI

37 

Yadav L, Puri N, Rastogi V, Satpute P, Ahmad R and Kaur G: Matrix metalloproteinases and cancer - roles in threat and therapy. Asian Pac J Cancer Prev. 15:1085–1091. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Li HC, Cao DC, Liu Y, et al: Prognostic value of matrix metalloproteinases (MMP-2 and MMP-9) in patients with lymph node-negative breast carcinoma. Breast Cancer Res Treat. 88:75–85. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Lin ZM, Zhao JX, Duan XN, et al: Effects of tissue factor, PAR-2 and MMP-9 expression on human breast cancer cell line MCF-7 invasion. Asian Pac J Cancer Prev. 15:643–646. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Izidoro-Toledo TC, Guimaraes DA, Belo VA, Gerlach RF and Tanus-Santos JE: Effects of statins on matrix metalloproteinases and their endogenous inhibitors in human endothelial cells. Naunyn Schmiedebergs Arch Pharmacol. 383:547–554. 2011. View Article : Google Scholar : PubMed/NCBI

41 

Fromigué O, Hamidouche Z and Marie PJ: Blockade of the RhoA-JNK-c-Jun-MMP2 cascade by atorvastatin reduces osteosarcoma cell invasion. J Biol Chem. 283:30549–30556. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Karin M and Lin A: NF-kappaB at the crossroads of life and death. Nat Immunol. 3:221–227. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Jana D, Das S, Sarkar DK, Mandal S, Maji A and Mukhopadhyay M: Role of nuclear factor-κB in female breast cancer: a study in Indian patients. Asian Pac J Cancer Prev. 13:5511–5515. 2012. View Article : Google Scholar

44 

Prajoko YW and Aryandono T: Expression of nuclear factor kappa B (NF-κB) as a predictor of poor pathologic response to chemotherapy in patients with locally advanced breast cancer. Asian Pac J Cancer Prev. 15:595–598. 2014. View Article : Google Scholar

45 

Gallogly MM, Shelton MD, Qanungo S, et al: Glutaredoxin regulates apoptosis in cardiomyocytes via NFkappaB targets Bcl-2 and Bcl-xL: implications for cardiac aging. Antioxid Redox Signal. 12:1339–1353. 2010. View Article : Google Scholar :

46 

Hoesel B and Schmid JA: The complexity of NF-κB signaling in inflammation and cancer. Mol Cancer. 12:862013. View Article : Google Scholar

47 

Hu LX, Du YY, Zhang Y and Pan YY: Synergistic effects of exemestane and aspirin on MCF-7 human breast cancer cells. Asian Pac J Cancer Prev. 13:5903–5908. 2012. View Article : Google Scholar

48 

Stoehr M, Mozet C, Boehm A, Aigner A, Dietz A and Wichmann G: Simvastatin suppresses head and neck squamous cell carcinoma ex vivo and enhances the cytostatic effects of chemotherapeutics. Cancer Chemother Pharmacol. 73:827–837. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2015
Volume 12 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shen YY, Yuan Y, Du YY and Pan YY: Molecular mechanism underlying the anticancer effect of simvastatin on MDA‑MB‑231 human breast cancer cells. Mol Med Rep 12: 623-630, 2015
APA
Shen, Y., Yuan, Y., Du, Y., & Pan, Y. (2015). Molecular mechanism underlying the anticancer effect of simvastatin on MDA‑MB‑231 human breast cancer cells. Molecular Medicine Reports, 12, 623-630. https://doi.org/10.3892/mmr.2015.3411
MLA
Shen, Y., Yuan, Y., Du, Y., Pan, Y."Molecular mechanism underlying the anticancer effect of simvastatin on MDA‑MB‑231 human breast cancer cells". Molecular Medicine Reports 12.1 (2015): 623-630.
Chicago
Shen, Y., Yuan, Y., Du, Y., Pan, Y."Molecular mechanism underlying the anticancer effect of simvastatin on MDA‑MB‑231 human breast cancer cells". Molecular Medicine Reports 12, no. 1 (2015): 623-630. https://doi.org/10.3892/mmr.2015.3411