1,25-Dihydroxyvitamin D3 and cisplatin synergistically induce apoptosis and cell cycle arrest in gastric cancer cells

  • Authors:
    • Anyu Bao
    • Yan Li
    • Yongqing Tong
    • Hongyun Zheng
    • Wei Wu
    • Chuandong Wei
  • View Affiliations

  • Published online on: February 24, 2014     https://doi.org/10.3892/ijmm.2014.1664
  • Pages: 1177-1184
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

1,25-Dihydroxyvitamin D3 [1,25(OH)2D3] plays an anticancer role in multiple types of cancer and potentiates the cytotoxic effects of several common chemotherapeutic agents. The hypercalcemia caused by 1,25(OH)2D3 alone or resistance to cisplatin weaken the anticancer effects of vitamin D. Thus, in this study, we aimed to investigate the synergistic effects of 1,25(OH)2D3 and cisplatin on the apoptosis and cell cycle progression of gastric cancer cells. BGC-823 human gastric cancer cells were treated with 1,25(OH)2D3 or cisplatin alone, or a combination of both agents. Cell apoptosis was assessed by TUNEL assay and flow cytometry. The expression of the apoptosis-related proteins, poly(ADP-ribose) polymerase (PARP), Bax, Bcl-2, caspase-3 and caspase-8, was examined using immunoblot analysis. ERK and AKT phosphorylation were examined by immunoblot analysis. The cell cycle distribution was determined by propidium iodide staining and flow cytometric analysis. p21 and p27 protein expression was also examined using immunoblot analysis. Our results revealed that co-treatment with 1,25(OH)2D3 enhanced cisplatin-induced apoptosis and upregulated the expression of Bax, and promoted the cleavage of PARP and caspase-3. The phosphorylation levels of ERK and AKT were reduced following combined treatment with 1,25(OH)2D3 and cisplatin. The percentage of cells in the G0/G1 phase was greater in the cells treated with the combined treatment than in those treated with either 1,25(OH)2D3 or cisplatin alone. p21 and p27 expression was upregulated following co-treatment with both agents. The results of this study suggest that 1,25(OH)2D3 potentiates cisplatin-mediated cell growth inhibition and cell apoptosis, which involves the upregulation of Bax, a decrease in ERK and AKT phosphorylation levels, and increased p21 and p27 levels.

Introduction

It has been previously demonstrated that vitamin D3 affects cell proliferation, differentiation and apoptosis (1). The antitumor activity of 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] is observed only when it is applied in supraphysiological doses, which may cause the side-effect of hypercalcemia (2). For this reason, the synthesis of analogs has been initiated in order to dissociate the calcemic effect from the anticancer activity of calcitriol. One strategy of improving the anticancer effects is to combine vitamin D with other agents in order to develop therapeutic interventions that allow dose reduction, the alleviation of toxicity and the maintenance of the growth inhibitory potential. According to the World Health Organization International Agency for Research on Cancer (IARC), gastric cancer is the second leading cause of cancer-related mortality worldwide (3). Despite significant progress in the treatment of patients with gastric cancer in recent years, there is a constant need for new therapies (4). Due to the low responsiveness of some patients suffering from gastric cancer to cisplatin therapy, there is a need to explore new combined therapeutic methods.

The role of vitamin D in inhibiting cancer cell growth, inducing cell differentiation and promoting cell apoptosis has been a research hotspot for the prevention and therapy of certain types of cancer, including gastric cancer (5). Although vitamin D exerts a less progressive direct killing effect on cancer cells, it can enhance the cytotoxicity of certain anticancer drugs, such as paclitaxel and may synergistically suppress the proliferation of cancer cells. The synergistic effects of vitamin D have been found in combination chemotherapy in various malignant somatic cells in vitro and in vivo (68).

Cisplatin is a major chemotherapeutic agent used in the treatment of gastric cancer. The National Comprehensive Cancer Network (NCCN) guideline suggested that cisplatin should be used as a first-line anticancer drug in the treatment of gastric cancer. Cisplatin exerts anticancer effects through various mechanisms; however, its most prominent mode of action is through the generation of DNA lesions followed by the activation of DNA damage response and the induction of cell apoptosis (9). Resistance to cisplatin arises through multiple mechanisms involving reduced drug uptake, increased drug inactivation, increased DNA damage repair, and the inhibition of transmission of DNA damage recognition signals to the apoptotic pathway (10). However, some patients with gastric cancer are not sensitive to cisplatin treatment. In addition, high-dose chemotherapy often results in several side-effects; a high concentration of vitamin D also causes hypocalcemia; thus, this limits the single use of both drugs.

1,25(OH)2D3 has been shown to synergistically or additively enhance the antitumor activities of a number of chemotherapeutic agents, including carboplatin, cisplatin, docetaxel and paclitaxel in prostate cancer (11), breast cancer (12), bladder cancer (13) and murine models of squamous cell carcinoma (SCC) (14). In the present study, we investigated the synergistic effects of 1,25(OH)2D3 and cisplatin on apoptosis and cell cycle distribution, as well as their mechanisms of action in BGC-823 gastric cancer cells in vitro. Our aim was to examine the biological effects of combined treatment with 1,25(OH)2D3 and cisplatin against gastric cancer cells. We also wished to to evaluate the effects of co-treatment with 1,25(OH)2D3 and cisplatin on the apoptosis and cell cycle distribution of gastric cancer cells, and to explore the possible mechanisms responsible for the the synergistic anticancer effects of 1,25(OH)2D3 and cisplatin.

Materials and methods

Drugs

1,25(OH)2D3 was purchased from Sigma-Aldrich (St. Louis, MO, USA). 1,25(OH)2D3 was dissolved in absolute ethanol (ETOH) to the concentration of 10−3 M and stored in solution at −20°C. 1,25(OH)2D3 was freshly diluted in culture medium to reach the required concentrations prior to each experiment. The ethanol concentration in each test condition never exceeded 0.1%.

Cisplatin was purchased from Shanghai Haoran Bio-Technology Co., Ltd. (Shanghai, China). Cisplatin was dissolved in sterile 0.9% NaCl to the concentration of 50 μg/ml and stored in solution at 4°C. Cisplatin was freshly diluted in culture medium to reach the required concentrations prior to each experiment.

Cell culture

The BGC-823 gastric cancer cell line was purchased from the Central Laboratory of Xiangya Medical College of Central South University, Changsha, China. The cells were cultured according to standard conditions. In brief, the BGC-823 gastric cancer cells were grown in RPMI-1640, 10% heat-inactivated fetal bovine serum (FBS), 100 U/ml penicillin and 100 mg/ml streptomycin at 37°C in a humid environment with 5% CO2. Cell media and reagents were obtained from Gibco-Invitrogen (Carlsbad, CA, USA). The culture media were changed every 48 h, and the cells were passaged every 2–3 days to produce new generations. The cells were plated in 25-cm2 flasks (Costar Life Sciences, Tewksbury MA, USA), and split every 48 h by washing with D-Hank’s solution and detached using 0.05% trypsin-EDTA. Half of the cells were plated in new flasks with fresh culture medium and the remaining cells were used for the experiments. The cells which had undergone 3 passages were selected for the experiments.

Drug treatment

The cells in the 1,25(OH)2D3 treatment group were cultured in RPMI-1640 culture medium with 10 nM 1,25(OH)2D3 for 72 h. The cells in the cisplatin treatment group were treated with 0.2 μg/ml cisplatin solution for 2 h following normal culture for 24 h; the cells were then cultured with fresh medium, followed by washing twice with D-Hank’s solution. The cells in the group co-treated with 1,25(OH)2D3 and cisplatin were treated with 0.2 μg/ml cisplatin for 2 h following culture for 24 h with 1,25(OH)2D3 alone, and were then cultured with fresh culture medium with 10 nM 1,25(OH)2D3. The control group was treated with ETOH in RPMI-1640 culture medium. The total culture time was 72 h for each group.

Preparation of cell extracts

The BGC-823 cells were seeded in 6-well plates (5×104 cells/well) and left overnight to attach. The cells were then treated with 1,25(OH)2D3 or cisplatin alone, or a combination of both. The cells treated with an equivalent amount of ETOH were used as the vehicle control. The cells were washed with D-Hank’s solution and replenished with fresh medium every 24 h. Following treatment for 72 h, the cells were harvested for immunoblot analysis using RIPA buffer (Thermo Scientific, Waltham, MA, USA) supplemented with protease inhibitors.

Immunoblot analysis

The cell protein concentration was measured using a BCA protein assay kit (Thermo Scientific) according to the manufacturer’s instructions. An immunoblot analysis was performed using a Bio-Rad wet electroblotting system (Bio-Rad, Hercules, CA, USA) according to the manufacturer’s instructions. The results from immunoblot analysis were quantified by measuring the optical density of the immunoreactive bands using ImageJ software.

Mouse and rabbit antibodies against Bax, Bcl-2, caspase-3, caspase-8, poly(ADP-ribose) polymerase (PARP), cleaved PARP, phosphorylated (p-)ERK1/2, ERK1/2, p-AKT, AKT, p21, p27 and β-actin were purchased from Cell Signaling Technology (Danvers, MA, USA). β-actin was used as the loading control to ensure equal protein loading among all wells in immunoblot analysis.

TUNEL assay

The In Situ Cell Death Detection Fluorescein kit (Roche Applied Science, Indianapolis, IN, USA) was used to detect cell apoptosis. Cell apoptosis was analyzed by TUNEL assay. In brief, the procedure was as follows: BGC-823 gastric cancer cell suspension was prepared following scheduled experiment treatment, the test sample was washed 3 times in phosphate-baffered saline (PBS) and adjusted to 2×107 cells/ml. Subsequently, 100 μl/tube suspension were transferred into a V-bottomed EP tube, and then 100 μl/tube of a freshly prepared fixation solution were added (4% paraformaldehyde in PBS, pH 7.4) to the cell suspension. The cells were then resuspended and incubated for 60 min at 22°C. The EP tubes were centrifuged at 300 × g for 10 min and the fixative was removed by flicking off or suction. The cells were then washed once with PBS, the EP tubes were centrifuged at 300 × g for 10 min again, and finally, the cells were resuspended in 100 μl/tube permeabilization solution (0.1% Triton X-100 in 0.1% sodium citrate) for 5 min on ice. The TUNEL reaction mixture was prepared immediately according to the kit’s instructions prior to use in the experiments and kept on ice until use. The cells were washed twice with PBS, then resuspended in 50 μl/tube TUNEL reaction mixture and incubated for 60 min at 37°C in a humidified atmosphere in the dark. The cells were then transferred to a tube to a final volume of 500 μl in PBS. The well-prepared samples were tested in a Beckman flow cytometry (Beckman Coulter, Miami, FL, USA) for the analysis of cell apoptosis.

Cell cycle analysis

The BGC-823 gastric cancer cells were treated with the vehicle control (ETOH), 1,25(OH)2D3 alone, cisplatin alone, or combined treatment with 1,25(OH)2D3 and cisplatin for 24 h. Following treatment, the BGC-823 cells (1×106/sample) were collected by trypsin digestion, then washed twice in cold PBS and fixed for 24 h in 70% ETOH at −20°C. The cells were then washed twice in PBS and incubated with RNAse (8 μg/ml; Fermentas, St. Leon-Rot, Germany) at 37°C for 1 h. The cells were stained with propidium iodide (0.5 mg/ml; Sigma-Aldrich) for 30 min at 37°C in the dark. The cellular DNA content was determined using a Beckman flow cytometry (Beckman Coulter) and ModFit LT 3.0 software (Verity Software House Inc., Topsham, ME, USA). The experiment was repeated 3 times.

Data analysis

Statistical analysis was performed by employing GraphPad Prism 5.0 software (GraphPad Software, CA, USA). All data are presented as the means ± standard error of the mean (SEM). Each experiment was repeated 3 times. The difference between the mean values of 2 groups was evaluated using the Student’s t-test. A value of P<0.05 was considered to indicate a statistically significant difference.

Results

1,25(OH)2D3 enhances the anticancer and apoptotic effects of cisplatin and in BGC-823 gastric cancer cells

The apoptosis of BGC-823 gastric cancer cells was evaluated by TUNEL assay. The apoptosis of the treated cancer cells was expressed using a fluorescent signal determined by flow cytometry. The density plots obtained by flow cytometry are shown as Fig. 1. The peaks shown in different colors represent the intensity of cell apoptosis following treatment with ETOH (vehicle control), 1,25(OH)2D3 or cisplatin alone, as well as co-treatment with 1,25(OH)2D3 and cisplatin.

Treatment with 10 nM 1,25(OH)2D3 or 0.2 μg/ml cisplatin alone significantly enhanced cell apoptosis compared with the control, as indicated by the increased fluorescence intensity of the DNA fragments in apoptotic cells (P<0.05). Co-treatment with 10 nM 1,25(OH)2D3 and 0.2 μg/ml cisplatin led to a significantly (P<0.05) greater number of apoptotic BGC-823 cells compared to treatment with cisplatin or 1,25(OH)2D3 alone (Table I).

Table I

Effects of treatment with 1,25(OH)2D3 alone or in combination with cisplatin on the apoptosis of BGC-823 cells.

Table I

Effects of treatment with 1,25(OH)2D3 alone or in combination with cisplatin on the apoptosis of BGC-823 cells.

GroupFluorescence intensity (means ± SEM)
Control (ETOH)4.73±0.55
1,25(OH)2D39.2±1.14a
Cisplatin14.17±4.01a
1,25(OH)2D3 + cisplatin23.07±3.00a,b

a P<0.05 compared to the control;

b P<0.05 compared to treatment with cisplatin or 1,25(OH)2D3 alone.

{ label (or @symbol) needed for fn[@id='tfn3-ijmm-33-05-1177'] } 1,25(OH)2D3, 1,25-dihydroxyvitamin D3; SEM, standard error of the mean.

Effects of co-treatment with 1,25(OH)2D3 and cisplatin on the expression of apoptosis-related proteins in BGC-823 gastric cancer cells

Following treatment with 1,25(OH)2D3 alone or in combination with cisplatin for 72 h, the cells were harvested for immunoblot analysis. The expression of a series of apoptosis-related proteins was then determined. The cleavage of PARP was significantly higher in the group co-treated with 1,25(OH)2D3 and cisplatin (P<0.01) compared with the group treated with cisplatin or 1,25(OH)2D3 alone (Fig. 2). In addition, the expression of caspase-3, a key member of the caspase family, was significantly reduced in the cells co-treated with 1,25(OH)2D3 and cisplatin (P<0.01) compared with the cells treated with cisplatin or 1,25(OH)2D3 alone. However, no significant change was observed in caspase-8 expression in all the treatment groups [1,25(OH)2D3 or cisplatin treatment alone or combined treatment (P>0.05)]. Bax expression was significantly upregulated by 1,25(OH)2D3 treatment alone (P<0.05) or the combined treatment with 1,25(OH)2D3 and cisplatin (P<0.01).

Regulation of AKT and ERK1/2 phosphorylation by co-treatment with 1,25(OH)2D3 and cisplatin

Cell apoptosis is regulated by multiple pathways. AKT and ERK1/2 are two important kinases involved in cell proliferation and apoptosis in gastric cancer (15,16). In the present study, we wished to explore the effects of 1,25(OH)2D3 or cisplatin treatment alone, as well as the effects of co-treatment with both agents on the phosphorylation levels of AKT and ERK1/2. As illustrated in Fig. 3, treatment with 1,25(OH)2D3 (P<0.05) or cisplatin (P<0.01) alone, as well as the combined treatment (P<0.01) significantly reduced the phosphorylation level of AKT. Furthermore, co-treatment with 1,25(OH)2D3 and cisplatin further reduced the phosphorylation level of AKT compared to treatment with cisplatin alone (P<0.05).

ERK1/2 phosphorylation was also observed following treatment with 1,25(OH)2D3 or cisplatin alone or the combined treatment. Both agents decreased the phosphorylation levels of ERK1/2 (P<0.01). Similarly, co-treatment with 1,25(OH)2D3 and cisplatin significantly reduced the phosphorylation levels of ERK1/2 compared to treatment with cisplatin alone (P<0.05). These results indicated that co-treatment with 1,25(OH)2D3 and cisplatin further enhanced the anti-proliferative effects of cisplatin on BGC-823 gastric cancer cells.

Effects of co-treatment with 1,25(OH)2D3 and cisplatin on cell cycle distribution of BGC-823 gastric cancer cells

The evaluation of the cell cycle distribution was carried out following treatment with 1,25(OH)2D3 or cisplatin alone or the comined treatment. Co-treatment with 1,25(OH)2D3 and cisplatin significantly increased the percentage of cells in the G0/G1 phase when compared to the group treated with ETOH (vehicle control), 1,25(OH)2D3 or cisplatin alone (P<0.05) (Fig. 4). We observed that the cells treated with both 1,25(OH)2D3 and cisplatin had accumulated in the G0/G1 phase and the number of cells was decreased in the G2/M phase, when compared to the cells treated with cisplatin or 1,25(OH)2D3 alone. However, the percentage of cells in the G2/M phase was not significantly affected by treatment with 1,25(OH)2D3 alone. The percentage of cells in the different cell cycle phases is shown in Table II. 1,25(OH)2D3, in combination with cisplatin, significantly increased the percentage of cells in the G0/G1 phase and decreased the percentage of cells in the G2/M phase, when compared to treatment with cisplatin or 1,25(OH)2D3 alone (P<0.05).

Table II

Effects of treatment with 1,25(OH)2D3 alone or in combination with cisplatin on cell cycle distribution of BGC-823 cells.

Table II

Effects of treatment with 1,25(OH)2D3 alone or in combination with cisplatin on cell cycle distribution of BGC-823 cells.

Percentage of cells in each cell cycle phase (means ± SEM)

G0/G1S G2/M
Control (ETOH)33.17±1.2751.37±1.8214.27±0.73
1,25(OH)2D330.77±0.7356.03±0.5813.17±0.59
Cisplatin33.57±1.1759.14±1.217.60±0.44
1,25(OH)2D3 + cisplatin38.87±1.14a55.83±0.795.28±0.35a

a P<0.05 compared to control, or treatment with 1,25(OH)2D3 or cisplatin alone.

{ label (or @symbol) needed for fn[@id='tfn5-ijmm-33-05-1177'] } 1,25(OH)2D3, 1,25-dihydroxyvitamin D3; SEM, standard error of the mean.

Effects of co-treatment with 1,25(OH)2D3 and cisplatin on p21 and p27 protein expression in BGC-823 gastric cancer cells

As the anti-proliferative effects of 1,25(OH)2D3 commonly involve the upregulation of p21 and/or p27 (17,18), we wished to determine the effects of 1,25(OH)2D3 and cisplatin on the protein expression of p21 and p27 in BGC-823 cells. p21 and p27 are important cell cycle regulators (19) and they were also examined in our study. As shown in Fig. 5, co-treatment with 1,25(OH)2D3 and cisplatin significantly increased the expression of p21 and p27 (P<0.01) compared to treatment with ETOH, 1,25(OH)2D3 or cisplatin alone. The effects on p21 and p27 expression induced by treatment with 1,25(OH)2D3 or cisplatin alone differed. Treatment with cisplatin alone significantly increased p27 expression (P<0.05), whereas 1,25(OH)2D3 had no such effect. Treatment with 1,25(OH)2D3 or cisplatin alone did not upregulate p21 expression. Co-treatment with 1,25(OH)2D3 and cisplatin further increased the expression of p21 and p27 compared to treatment with cisplatin alone (P<0.05).

Discussion

Gastric cancer remains the second most common cause of cancer-related mortality worldwide (20). Surgery remains as the most common curative treatment. However, the majority of patients with gastric cancer develop local or distant recurrence (21). Meta-analyses have indicated that certain patients treated with chemotherapy following surgery benefit from this treatment strategy, while other patients have undergone expensive and potentially toxic therapy without any beneficial effects (22). Cisplatin is widely used and has been demonstrated to be effective in the palliative treatment of gastric cancer (23). Oxaliplatin, the third generation platinum compound, plus 5-fluorouracil modulated with leucovorin (FOLFOX) has been widely used as the first-line treatment in advanced gastric cancer (24,25). However, resistance to oxaliplatin and cisplatin remains a major obstacle to further increasing the treatment response rate. In the present study, to the best of our knowledge, we demonstrate for the first time the biological effects of combined treatment with 1,25(OH)2D3 and cisplatin against gastric cancer cell growth. We observed that 1,25(OH)2D3 induced the apoptosis of BGC-823 cells as shown by TUNEL assay. However, when used in combination with cisplatin, the apoptotic signal significantly increased compared to treatment with cisplatin alone. Previous studies have indicated that the anticancer effects of vitamin D are limited in certain types of cancer (8). Preclinical experiments have suggested that vitamin D exerts minor effects on the prevention or therapy of cancer in vivo, although it inhibits cell proliferation and induces cell apoptosis in vitro (26,27). However, vitamin D and its analogues are still being focused on, as they exert synergistic anticancer effects when used in combination with chemotherapeutic drugs, such as platinum (28). The mechanisms of cancer progression have been clarified and a number of target proteins have been identifed to be important in the treatment of cancer (29).

In the present study, we demonstrated the differential effects of treatment with vitamin D3 alone or in combination with cisplatin on the apoptosis of BGC-823 gastric cancer cells. Treatment with 1,25(OH)2D3 and cisplatin alone induced the apoptosis of BGC-823 cells, as shown by TUNEL assay. Furthermore, enhanced apoptosis was observed following co-treatment with both agents, and the fluorescence intensity of apoptotic cells markedly increased by 5-fold of the control, and by 1.6-fold that of cisplatin (Table I). We also observed the changes in protein expression following co-treatment with 1,25(OH)2D3 and cisplatin. The caspase pathway is involved in vitamin D-induced cell apoptosis (18,30). In the present study, caspase-3 expression was reduced following co-treatment with 1,25(OH)2D3 and cisplatin, which indicated a greater apoptotic status in the BGC-823 cells. In addition, the significantly increased cleavage of PARP and the expression of pro-apoptotic Bax were also observed following co-treatment with 1,25(OH)2D3 and cisplatin when compared to treatment with cisplatin alone.

Having demonstrated the synergism between 1,25(OH)2D3 and cisplatin, we sought to explore the underlying mechanisms. Caspases play a crucial role in apoptotic cell death induced by vitamin D3 (30). The apoptosis of gastric cancer cells can be triggered by the extrinsic pathway activated by death receptor and the intrinsic pathway regulated by Bcl-2 family members and caspase cascades in the mitochondrion (31). Previous studies have indicated that 1,25(OH)2D3-mediated apoptosis is caspase-dependent and appears to act through the mitochondrial pathway of cytochrome c release, caspase-9 activation, and subsequent caspase-3 activation, finally the processing of PARP (32). In our study, we found that caspase-3 expression was significantly decreased following treatment with 1,25(OH)2D3 and cisplatin, while caspase-8 expression remained unaltered following the combined treatment [1,25(OH)2D3 and cisplatin] or treatment with 1,25(OH)2D3 and cisplatin alone. This suggests that the mitochondrial pathway is involved in vitamin D-mediated apoptosis, although the involvement of other pathways cannot be ruled out.

We also found that the pro-apoptotic protein, Bax, was upregulated following co-treatment with 1,25(OH)2D3 and cisplatin, while treatment with cisplatin alone did not increase the expression of Bax. The translocation of Bax to the mitochondria has been shown to be of particular importance for the induction of vitamin D-mediated apoptosis in certain cell types. The treatment of MCF-7 breast cancer cells with 1,25(OH)2D3 has been shown to result in the redistribution of Bax from the cytosol to the mitochondria (33,34). Changes in the expression or cellular distribution of Bcl-2 anti-apoptotic proteins are a possible mechanism of 1,25(OH)2D3-mediated apoptosis (35). However, Bcl-2 expression did not show a downregulation in the BGC-823 gastric cancer cells treated with 1,25(OH)2D3 or cisplatin. Cisplatin cytotoxicity results from the formation of bifunctional, intrastrand DNA adducts (36). Cisplatin activates p53 and then results in the increased transcription of p53 target genes, including Bax and p21, as well as in cell cycle arrest and apoptosis (37). In our study, the combined use of cisplatin and 1,25(OH)2D3 enhanced the apoptosis of BGC-823 cells.

In our previous study, we reported that 1,25(OH)2D3-mediated apoptosis is associated with the downregulation of the AKT and ERK survival signaling pathways (18). Activated AKT phosphorylates a host of proteins that affect cell growth, cell cycle entry and cell survival. The decreased phosphorylation of AKT may contribute to the anti-proliferative effects of 1,25(OH)2D3. siRNA-AKT has been shown to promote 1,25(OH)2D3-induced apoptosis in SCC cells through the caspase-10-caspase-3 pathway, whereas caspase-8 and caspase-9 are not involved (38). Akt may regulate apoptosis through a number of different mechanisms depending on the apoptotic stimuli and cell types, which involve the regulation of phosphorylation and protein expression (39,40). In our study, as compared to treatment with cisplatin alone, AKT phosphorylation was further decreased in the cells that became apoptotic following combined treatment with 1,25(OH)2D3 and cisplatin. These data indicate that 1,25(OH)2D3 further enhances the cisplatin-induced loss of survival signaling, and thus further inhibits the proliferation of gastric cancer cells. The AKT pathway presents an attractive target for anticancer therapies, which may be applied in future anticancer chemotherapy.

It has been demonstrated that the MAPK-ERK pathway is one of the most significant signal transduction pathways (41), and several key growth factors and genes promote tumor growth by activating this signaling cascade. The downregulation of ERK phosphorylation is a contributing factor to cellular apoptosis in gastric cancer (42). The vitamin D analog, Gemini, has been shown to suppress ErbB2-positive mammary tumor growth through the inhibition of ErbB2/AKT/ERK signaling (43), and the knockdown of ZFX has been shown to inhibit gastric cancer cell growth in vitro and in vivo by downregulating the MAPK-ERK pathway (44). In our previous study, we observed that the pERK/ERK ratio was decreased in 1,25(OH)2D3-treated BGC-823 cells (18), and we found a further reduction in pERK/ERK in BGC-823 cells co-treated with 1,25(OH)2D3 and cisplatin. Therefore, the promotion of gastric cancer cell apoptosis or inhibition of cell growth due to the combined effects of vitamin D and cisplatin may be explained, at least in part, by the inhibition of the ERK and AKT pathway. However, the direct link between vitamin D and the ERK or AKT pathway requires further investigation.

The anti-proliferative effects of 1,25(OH)2D3 commonly involve the cell cycle arrest of different cancer cells. 1,25(OH)2D3 inhibits cell proliferation, induces cell cycle arrest and promotes the accumulation of cells in the G0/G1 phase in multipotent mesenchymal cells (MMCs) (45). The vitamin D analogue, EB1089, has been shown to significantly reduce cell growth in human hepatoma cells (Hep-G2) and block Hep-G2 cell-associated tumor formation in nude mice through cell cycle G1 phase arrest by the accumulation of p27 (46). In this study, we demonstrated that 1,25(OH)2D3 alone did not induce cell cycle G0/G1 arrest or G2/M cell cycle change. However, 1,25(OH)2D3, in conjunction with cisplatin, induced G0/G1 cell cycle arrest or a decrease in the number of cells in the G2/M phase in BGC-823 cells, and we observed that the effects of this combined treatment were more potent compared to the effects induced by cisplatin alone.

Although a number of 1,25(OH)2D3 responsive genes are known, the exact mechanisms of growth regulation by 1,25(OH)2D3 have not been completely defined. However, an increase in p21 and/or p27 expression is an almost universal feature (47). In our study, p21 protein expression increased significantly following co-treatment with 1,25(OH)2D3 and cisplatin, and p27 was upregulated to a much higher degree following the combined treatment compared to treatment with cisplatin alone. This indicates that 1,25(OH)2D3 promotes the effects of cisplatin, inducing cell cycle arrest in the G0/G1 phase.

In conclusion, to the best of our knowledge, the present study demonstrates for the first time that 1,25(OH)2D3 plays a synergistic role in cisplatin-mediated growth inhibition and apoptosis in gastric cancer cells. The combined use of 1,25(OH)2D3 and cisplatin may be used as a strategy to overcome resistance to cisplatin and dose limitations, and to improve the anticancer effects of chemotherapy.

Acknowledgements

This study was supported by grants from the National Clinical Key Specialty Construction Project.

References

1 

Deeb KK, Trump DL and Johnson CS: Vitamin D signalling pathways in cancer: potential for anticancer therapeutics. Nat Rev Cancer. 7:684–700. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Eelen G, Verlinden L, De Clercq P, Vandewalle M, Bouillon R and Verstuyf A: Vitamin D analogs and coactivators. Anticancer Res. 26:2717–2721. 2006.PubMed/NCBI

3 

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

4 

Wadhwa R, Song S, Lee JS, Yao Y, Wei Q and Ajani JA: Gastric cancer-molecular and clinical dimensions. Nat Rev Clin Oncol. 10:643–655. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Abnet CC, Chen Y, Chow WH, et al: Circulating 25-hydroxyvitamin D and risk of esophageal and gastric cancer: Cohort Consortium Vitamin D Pooling Project of Rarer Cancers. Am J Epidemiol. 172:94–106. 2010. View Article : Google Scholar

6 

Fleet JC: Molecular actions of vitamin D contributing to cancer prevention. Mol Aspects Med. 29:388–396. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Feldman DR, Bosl GJ, Sheinfeld J and Motzer RJ: Medical treatment of advanced testicular cancer. JAMA. 299:672–684. 2008. View Article : Google Scholar

8 

Krishnan AV, Trump DL, Johnson CS and Feldman D: The role of vitamin D in cancer prevention and treatment. Rheum Dis Clin North Am. 38:161–178. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Krege S, Beyer J, Souchon R, et al: European consensus conference on diagnosis and treatment of germ cell cancer: a report of the second meeting of the European Germ Cell Cancer Consensus group (EGCCCG): part I. Eur Urol. 53:478–496. 2008. View Article : Google Scholar

10 

Tanida S, Mizoshita T, Ozeki K, et al: Mechanisms of cisplatin-induced apoptosis and of cisplatin sensitivity: potential of BIN1 to act as a potent predictor of cisplatin sensitivity in gastric cancer treatment. Int J Surg Oncol. 2012:8628792012.

11 

Moffatt KA, Johannes WU and Miller GJ: 1Alpha, 25dihydroxyvitamin D3 and platinum drugs act synergistically to inhibit the growth of prostate cancer cell lines. Clin Cancer Res. 5:695–703. 1999.PubMed/NCBI

12 

Cho YL, Christensen C, Saunders DE, et al: Combined effects of 1,25-dihydroxyvitamin D3 and platinum drugs on the growth of MCF-7 cells. Cancer Res. 51:2848–2853. 1991.PubMed/NCBI

13 

Ma Y, Yu WD, Trump DL and Johnson CS: 1,25D3 enhances antitumor activity of gemcitabine and cisplatin in human bladder cancer models. Cancer. 116:3294–3303. 2010.PubMed/NCBI

14 

Light BW, Yu WD, McElwain MC, Russell DM, Trump DL and Johnson CS: Potentiation of cisplatin antitumor activity using a vitamin D analogue in a murine squamous cell carcinoma model system. Cancer Res. 57:3759–3764. 1997.

15 

Almhanna K, Strosberg J and Malafa M: Targeting AKT protein kinase in gastric cancer. Anticancer Res. 31:4387–4392. 2011.PubMed/NCBI

16 

Yao J, Qian CJ, Ye B, Zhang X and Liang Y: ERK inhibition enhances TSA-induced gastric cancer cell apoptosis via NF-κB-dependent and Notch-independent mechanism. Life Sci. 91:186–193. 2012.PubMed/NCBI

17 

Chen S, Law CS and Gardner DG: Vitamin D-dependent suppression of endothelin-induced vascular smooth muscle cell proliferation through inhibition of CDK2 activity. J Steroid Biochem Mol Biol. 118:135–141. 2010. View Article : Google Scholar

18 

Bao A, Li Y, Tong Y, Zheng H, Wu W and Wei C: Tumor-suppressive effects of 1,25-dihydroxyvitamin D3 in gastric cancer cells. Hepatogastroenterology. 60:943–948. 2013.PubMed/NCBI

19 

Mitrea DM, Yoon MK, Ou L and Kriwacki RW: Disorder-function relationships for the cell cycle regulatory proteins p21 and p27. Biol Chem. 393:259–274. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Leung WK, Wu MS, Kakugawa Y, et al: Screening for gastric cancer in Asia: current evidence and practice. Lancet Oncol. 9:279–287. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Macdonald JS: Treatment of localized gastric cancer. Semin Oncol. 31:566–573. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Carrato A, Gallego-Plazas J and Guillen-Ponce C: Adjuvant therapy of resected gastric cancer is necessary. Semin Oncol. 32(Suppl 9): S105–S108. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Topuz E, Basaran M, Saip P, et al: Adjuvant intraperitoneal chemotherapy with cisplatinum, mitoxantrone, 5-fluorouracil, and calcium folinate in patients with gastric cancer: a phase II study. Am J Clin Oncol. 25:619–624. 2002. View Article : Google Scholar

24 

Cavanna L, Artioli F, Codignola C, et al: Oxaliplatin in combination with 5-fluorouracil (5-FU) and leucovorin (LV) in patients with metastatic gastric cancer (MGC). Am J Clin Oncol. 29:371–375. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Louvet C, Andre T, Tigaud JM, et al: Phase II study of oxaliplatin, fluorouracil, and folinic acid in locally advanced or metastatic gastric cancer patients. J Clin Oncol. 20:4543–4548. 2002. View Article : Google Scholar

26 

Mocellin S: Vitamin D and cancer: deciphering the truth. Biochim Biophys Actas. 1816:172–178. 2011.PubMed/NCBI

27 

Picotto G, Liaudat AC, Bohl L and Tolosa de Talamoni N: Molecular aspects of vitamin D anticancer activity. Cancer Invest. 30:604–614. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Reichrath J, Friedrich M and Vogt T: Vitamin D and its analogs in cancer prevention and therapy. Anticancer Res. 32:209–210. 2012.PubMed/NCBI

29 

Rahman N: Realizing the promise of cancer predisposition genes. Nature. 505:302–308. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Fingas CD, Altinbas A, Schlattjan M, et al: Expression of apoptosis- and vitamin D pathway-related genes in hepatocellular carcinoma. Digestion. 87:176–181. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Fulda S and Debatin KM: Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene. 25:4798–4811. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Wang W, Zhao CH, Zhang N and Wang J: Vitamin D analog EB1089 induces apoptosis in a subpopulation of SGC-7901 gastric cancer cells through a mitochondrial-dependent apoptotic pathway. Nutr Cancer. 65:1067–1075. 2013. View Article : Google Scholar

33 

Narvaez CJ and Welsh J: Role of mitochondria and caspases in vitamin D-mediated apoptosis of MCF-7 breast cancer cells. J Biol Chem. 276:9101–9107. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Koshizuka K, Koike M, Kubota T, Said J, Binderup L and Koeffler HP: Novel vitamin D3 analog (CB1093) when combined with paclitaxel and cisplatin inhibit growth of MCF-7 human breast cancer cells in vivo. Int J Oncol. 13:421–428. 1998.

35 

Wagner N, Wagner KD, Schley G, Badiali L, Theres H and Scholz H: 1,25-dihydroxyvitamin D3-induced apoptosis of retinoblastoma cells is associated with reciprocal changes of Bcl-2 and bax. Exp Eye Res. 77:1–9. 2003.

36 

Basu A and Krishnamurthy S: Cellular responses to cisplatin-induced DNA damage. J Nucleic Acids. 2010:2013672010. View Article : Google Scholar

37 

Wang S, Li W, Xue Z, et al: Molecular imaging of p53 signal pathway in lung cancer cell cycle arrest induced by cisplatin. Mol Carcinog. 52:900–907. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Ma Y, Yu WD, Kong RX, Trump DL and Johnson CS: Role of nongenomic activation of phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase/extracellular signal-regulated kinase kinase/extracellular signal-regulated kinase 1/2 pathways in 1,25D3-mediated apoptosis in squamous cell carcinoma cells. Cancer Res. 66:8131–8138. 2006.

39 

Vivanco I and Sawyers CL: The phosphatidylinositol 3-kinase AKT pathway in human cancer. Nat Rev Cancer. 2:489–501. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Xiao H, Shi W, Liu S, et al: 1,25-Dihydroxyvitamin D(3) prevents puromycin aminonucleoside-induced apoptosis of glomerular podocytes by activating the phosphatidylinositol 3-kinase/Akt-signaling pathway. Am J Nephrol. 30:34–43. 2009. View Article : Google Scholar

41 

Roberts PJ and Der CJ: Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene. 26:3291–3310. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Shen XJ, Wang HB, Ma XQ and Chen JH: β,β-Dimethylacrylshikonin induces mitochondria dependent apoptosis through ERK pathway in human gastric cancer SGC-7901 cells. PLoS One. 7:e417732012.

43 

Lee HJ, So JY, DeCastro A, et al: Gemini vitamin D analog suppresses ErbB2-positive mammary tumor growth via inhibition of ErbB2/AKT/ERK signaling. J Steroid Biochem Mol Biol. 121:408–412. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Wu S, Lao XY, Sun TT, et al: Knockdown of ZFX inhibits gastric cancer cell growth in vitro and in vivo via downregulating the ERK-MAPK pathway. Cancer Lett. 337:293–300. 2013. View Article : Google Scholar : PubMed/NCBI

45 

Artaza JN, Sirad F, Ferrini MG and Norris KC: 1,25(OH)2vitamin D3 inhibits cell proliferation by promoting cell cycle arrest without inducing apoptosis and modifies cell morphology of mesenchymal multipotent cells. J Steroid Biochem Mol Biol. 119:73–83. 2010.

46 

Luo W, Chen Y, Liu M, et al: EB1089 induces Skp2-dependent p27 accumulation, leading to cell growth inhibition and cell cycle G1 phase arrest in human hepatoma cells. Cancer Invest. 27:29–37. 2009. View Article : Google Scholar

47 

Banerjee P and Chatterjee M: Antiproliferative role of vitamin D and its analogs - a brief overview. Mol Cell Biochem. 253:247–254. 2003. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2014
Volume 33 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bao A, Li Y, Tong Y, Zheng H, Wu W and Wei C: 1,25-Dihydroxyvitamin D3 and cisplatin synergistically induce apoptosis and cell cycle arrest in gastric cancer cells. Int J Mol Med 33: 1177-1184, 2014
APA
Bao, A., Li, Y., Tong, Y., Zheng, H., Wu, W., & Wei, C. (2014). 1,25-Dihydroxyvitamin D3 and cisplatin synergistically induce apoptosis and cell cycle arrest in gastric cancer cells. International Journal of Molecular Medicine, 33, 1177-1184. https://doi.org/10.3892/ijmm.2014.1664
MLA
Bao, A., Li, Y., Tong, Y., Zheng, H., Wu, W., Wei, C."1,25-Dihydroxyvitamin D3 and cisplatin synergistically induce apoptosis and cell cycle arrest in gastric cancer cells". International Journal of Molecular Medicine 33.5 (2014): 1177-1184.
Chicago
Bao, A., Li, Y., Tong, Y., Zheng, H., Wu, W., Wei, C."1,25-Dihydroxyvitamin D3 and cisplatin synergistically induce apoptosis and cell cycle arrest in gastric cancer cells". International Journal of Molecular Medicine 33, no. 5 (2014): 1177-1184. https://doi.org/10.3892/ijmm.2014.1664