Open Access

Corosolic acid reduces 5‑FU chemoresistance in human gastric cancer cells by activating AMPK

  • Authors:
    • Jun Beom Park
    • Jin Sun Lee
    • Myung Sun Lee
    • Eun Young Cha
    • Soyeon Kim
    • Ji Young Sul
  • View Affiliations

  • Published online on: July 3, 2018     https://doi.org/10.3892/mmr.2018.9244
  • Pages: 2880-2888
  • Copyright: © Park et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

5‑Fluorouracil (5‑FU) is one of the most commonly used chemotherapeutic agents for gastric cancer. Resistance to 5‑FU‑based chemotherapy remains the major obstacle in the treatment of gastric cancer. A growing body of evidence has suggested that adenosine monophosphate‑activated protein kinase (AMPK) is pivotal for chemoresistance. However, the mechanism by which AMPK regulates the chemosensitivity of gastric cancer remains unclear. In the present study, how corosolic acid enhanced the chemosensitivity of gastric cancer cells to 5‑FU via AMPK activation was investigated. A 5‑FU‑resistant gastric cancer cell line (SNU‑620/5‑FUR) was established, which had a marked increase in thymidine synthase (TS) expression but reduced AMPK phosphorylation when compared with the parental cell line, SNU‑620. AMPK regulation by 5‑aminoimidazole‑4‑carboxamide ribonucleotide or compound c was revealed to be markedly associated with TS expression and 5‑FU‑resistant cell viability. In addition, corosolic acid activated AMPK, and decreased TS expression and the phosphorylation of mammalian target of rapamycin/4E‑binding protein 1 in a dose‑dependent manner. Corosolic acid treatment significantly reduced cell viability while compound c reversed corosolic acid‑induced cell growth inhibition. The 5‑FU‑resistance sensitization effect of corosolic acid was determined by the synergistic reduction of TS expression and inhibition of cell viability in the presence of 5‑FU. The corosolic acid‑induced AMPK activation was markedly increased by additional 5‑FU treatment, while compound c reversed AMPK phosphorylation. In addition, compound c treatment reversed corosolic acid‑induced apoptotic markers such as capase‑3 and PARP cleavage, and cytochrome c translocation to cytosol, in the presence of 5‑FU. Corosolic acid treatment in the presence of 5‑FU induced an increase in the apoptotic cell population based on flow cytometry analysis. This increase was abolished by compound c. In conclusion, these results implied that corosolic acid may have therapeutic potential to sensitize the resistance of gastric cancer to 5‑FU by activating AMPK.

Introduction

Gastric cancer is one of the most common malignant diseases and is the second leading cause of cancer-related death worldwide. The prognosis for advanced gastric cancer remains poor (1). Early stage gastric cancer patients are asymptomatic or experience minor symptoms (2). Most patients with symptoms that can be noticed clinically have reached an advanced stage (2). Surgery is the only potentially curative treatment for gastric cancer. To improve upon low survival outcomes one of major obstacles in cancer therapy, the development of tumor chemoresistance, needs to be overcome. Therefore, it is necessary to investigate the mechanisms involved in chemoresistance, to develop strategies that sensitize cancer cells to chemotherapeutic agents.

5-Fluorouracil (5-FU) is one of the most commonly used chemotherapeutic agents for gastric cancer. Despite its many advantages, clinical applications of 5-FU have been limited by drug resistance that arises due to several factors, including altered drug influx and efflux, enhancement of drug inactivation, and mutations to the drug target (3). Several intracellular enzymes, thymidine synthase (TS), dihydropyrimidine dehydrogenase (DPD), methylenetetrahydrofolate reductase (MTHFR) and thymidine phosphorylase (TP), are considered important predictors for 5-FU sensitivity or resistance (4). However, mechanisms underlying 5-FU anti-tumor activity and drug resistance have not been fully revealed yet.

TS expression and activity are increased in several tumor tissues, including lung, cervical, breast, and gastric cancers. They are considered to be indicators of cell proliferation and are associated with poor prognosis. TS has been used as an important target in chemotherapy (5,6). Several studies have shown that TS expression is a key regulator of 5-FU resistance/sensitivity (7,8).

Adenosine monophosphate-activated protein kinase (AMPK) is a heterodimeric enzyme consisting of a catalytic α-subunit and two non-catalytic β and γ subunits. AMPK functions in the cellular metabolism of glucose, lipid, and protein (9). The role of metformin, an AMPK activator, as a chemosensitizer has been investigated in Bel-7402/5-FU cells (hepatocellular carcinoma) and MCF7/5-FU cells (breast cancer) (10,11). It has been reported that AICAR, another AMPK activator, enhanced the pro-apoptotic effect of 5-FU in 5-FU-resistant SGC-7901 cells (gastric cancer) (12). Additionally, it has been reported that phosphorylated AMPK level is reduced in 5-FU-resistant gastric cancer cells while glucose metabolism is increased in 5-FU-resistant HepG2 cells (13). However, it remains unknown as to whether AMPK can increase chemosensitization in gastric cancer cells.

Corosolic acid (2a-hydroxyursolic acid), one of the main triterpenoids, has been discovered in many medicinal plants such as Lagerstroemia speciosa (banaba) and Weigela subsessilis (14,15). Corosolic acid not only displays remarkable hypoglycemic effects in some animal experiments and clinical trials (16,17), but has also been shown to possess antitumor effects against several cancers, including liver, colon, lung, and gastric cancer (1821). Previous studies have reported that corosolic acid can enhance the anticancer effect of 5-FU in SNU-620 and NCI-N87 gastric cancer cells, suggesting that it might act as an AMPK activator (2125). Among natural chemicals, curcumin, epigallocatechin gallate (EGCG), and sinomenine have been found to be able to sensitize 5-FU resistance in gastric cancers (2628). However, whether corosolic acid can do the same for 5-FU resistance in cancers remains unclear.

Therefore, the objective of this study was to determine the effect of corosolic acid on the response of gastric cancer to 5-FU. We used 5-FU resistant human gastric cancer cells (SNU-620/5-FUR) and treated them with corosolic acid in the presence or absence of 5-FU to investigate the effect of corosolic acid on 5-FU resensitization, and determine the mechanism of action.

Materials and methods

Materials

RPMI-1640, fetal bovine serum (FBS) and penicillin/streptomycin were obtained from HyClone (GE Healthcare Life Sciences, Logan, UT, USA). Trypsin/EDTA was purchased from Gibco (Thermo Fisher Scientific, Inc., Waltham, MA, USA). The following primary antibodies were used: Rabbit polyclonal anti-human thymidylate synthase (1:1,000; no. 3766), rabbit polyclonal anti-human caspase-3 (1:1,000; no. 9662), rabbit polyclonal anti-human poly-(ADP-ribose) polymerase (PARP) (1:1,000; no. 9542), rabbit polyclonal anti-human AMPK (1:1,000; no. 2532), rabbit monoclonal anti-human phospho-AMPK (Thr172) (1:1,000; no. 2535), rabbit polyclonal anti-human mTOR (1:1,000; no. 2972), rabbit polyclonal anti-human phospho-mTOR (Ser2448) (1:1,000; no. 2971), rabbit polyclonal anti-human 4E-binding protein 1 (4EBP1) (1:1,000; no. 9452) and rabbit polyclonal anti-human phospho-4EBP1 (Thr70) (1:1,000; no. 9455) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA), and rabbit polyclonal anti-human GAPDH (1:1,000; sc-25778) were obtained from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Horseradish peroxidase-conjugated anti-mouse and anti-rabbit antibodies were obtained from Transduction Lab (Lexington, KY, USA). SuperSignal® West Pico Chemiluminescent Substrate was purchased from Pierce (Thermo Fisher Scientific, Inc., Waltham, MA, USA) and 5-FU was provided by Choongwae Pharmaceutical Co., Ltd. (Seoul, Korea). Cell Counting Kit-8 (CCK-8) was purchased from Dojindo Laboratories (Kumamoto, Japan) and the EzWay Annexin-V-FITC Apoptosis Detection kit was purchased from KomaBiotech, Inc. (Seoul, Korea). A Mitochondrial Apoptosis Staining kit was purchased from PromoKine® (PromoCell GmbH, Heidelberg, Germany). Corosolic acid, compound c, AICAR and all other reagents were obtained from Sigma-Aldrich (Merck KGaA, Darmstadt, Germany).

Cell culture

Human gastric carcinoma SNU-620 cells were purchased from Korean Cell Line Bank (Seoul, Korea). Cells were grown in RPMI-1640 media supplemented with 10% (v/v) FBS, penicillin (100 U/ml)/streptomycin (100 µg/ml) at 37°C in a humidified CO2 (5%)-controlled incubator. 5-FU-resistant SNU-620/5-FUR cells were established by repeated cultures of SNU-620 with constant treatment with 7.5 µM 5-FU.

Cell growth inhibition assay

Cells were seeded at 5×103 cells/ml in 96-well microplates and allowed to attach for 24 h. 5-FU (~750 µM) or corosolic acid (~25 µM) were added to the medium at various concentrations. Following treatment, the cell cytotoxicity and/or proliferation was assessed using the CCK-8 assay. Briefly, highly water-soluble tetrazolium salt [2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium, monosodium salt], produced an orange-colored water-soluble product, formazan. The quantity of formazan dye generated by dehydrogenases in the cells was directly proportional to the number of living cells. CCK-8 (10 µl) was added to each well and incubated for 3 h at 37°C; cell proliferation and cytotoxicity were assessed by measuring the absorbance at 450 nm using a microplate reader (Corning Incorporated, Corning, NY, USA). Three replicated wells were used per experimental condition.

Annexin V/Propidium iodide staining

Cells were cultured at a 106 density and treated with corosolic acid and/or compound c for 24 h. Cells were centrifuged and washed three times with phosphate-buffered saline (PBS), and centrifuged. The supernatant was discarded and resuspended in 0.5 ml of cold PBS. The cells were processed and labeled according to the EzWay Annexin V-FITC Apoptosis Detection Kit that was used for this assay. The labeled cells were analyzed in a flow cytometer (BD FACSCanto™II; BD Biosciences, Franklin Lakes, NJ, USA).

Western blotting analysis

Cells were harvested using Trypsin-EDTA, washed twice with cold PBS, lysed with lysis buffer (10 mM Tris, pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% Triton X-100, 0.5% NP-40, 1 mM PI, 1 mM DTT, 1 mM PMSF), and placed on ice for 1 h with occasional vortexing. Centrifugation followed at 13,000 × g for 10 min at 4°C to collect the supernatant. A Pierce BCA Protein Assay kit (Pierce; Thermo Fisher Scientific, Inc.) was used to determine the protein concentration. The cell lysate (50 µg) was subjected to SDS-polyacrylamide gel electrophoresis (PAGE) and transferred to polyvinylidene difluoride (PVDF) membrane. Blots were blocked with 5% skim milk in PBS containing 0.05% Tween-20 for 1 h at 25°C, then incubated with primary antibodies (1:1,000) overnight at 4°C, followed by incubation with anti-rabbit horseradish peroxidase-conjugated IgG (1:3,000) for 2 h at room temperature and visualized with enhanced chemiluminescence.

Statistical analysis

All results presented were confirmed in at least three independent experiments. Data were presented as the mean ± standard deviation. Statistical differences were analyzed by one-way analysis of variance followed by Tukey's post hoc test using SPSS version 24.0 software (IBM Corp., Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Establishment of 5-FU resistant SNU-620 gastric cancer cells

We established a 5-FU-resistant gastric cancer cell line (SNU-620/5-FUR) by continuous exposure of the parental cells SNU-620 to 7.5 µM 5-FU for approximately 6 months. TS protein level was markedly increased in SNU-620/5-FUR cells compared to that in the 5-FU-sensitive parental cells based on Western blot analysis, indicating that SNU-620/5-FUR cells were resistant to 5-FU (Fig. 1A and B). To confirm that SNU-620/5-FUR cells were resistant to 5-FU, 5-FU was added at various concentrations (~750 µM). Cell viability was then determined by CCK-8 assay. 5-FU decreased cell viabilities of SNU-620 cells in a dose-dependent manner. However, it was not cytotoxic to SNU-620/5-FUR cells (Fig. 1C).

AMPK phosphorylation level was reduced in 5-FU-resistant SNU-620/5-FUR gastric cancer cells

AMPK phosphorylation was found to be reduced by 32.7% in 5-FU-resistant SNU-620/5-FUR gastric cancer cells compared to the SNU-620 cells (Fig. 2A and B). To confirm the role of AMPK in cell resensitization to 5-FU, cells were treated with AMPK activator AICAR and the AMPK inhibitor compound c. TS protein expression and cell viability were then determined by western blotting analysis and CCK-8 assay. AICAR dramatically decreased TS expression (Fig. 2C and D) while AMPK inhibition by compound c treatment increased TS protein expression in SNU-620/5-FUR cells (Fig. 2C and D). Activation of AMPK by AICAR decreased viability of SNU-620/5-FUR cells. However, inhibition of AMPK by compound c did not decrease the viability of SNU-620/5-FUR cells (Fig. 2E). These results suggest that 5-FU resistance is strongly regulated by AMPK. Therefore, AMPK phosphorylation might be a therapeutic target for overcoming 5-FU-resistance in gastric cancers.

Corosolic acid activates AMPK and suppresses mTOR/4EBP1 phosphorylation in SNU-620/5-FUR cells

Previous studies have reported that pharmacological activators of AMPK such as AICAR and metformin can induce apoptosis of gastric cancers (12,29). Moreover, corosolic acid, (2α,3β)-2,3-dihydroxyurs-12-en-28-oic acid (Fig. 3A) can activate AMPK and induce apoptosis in gastric cancers (25). Corosolic acid can also inhibit inflammation in adipose tissues (23). To evaluate the effect of corosolic acid on AMPK activation in SNU-620/5-FUR cells, western blot analysis was performed. Results revealed that treatment with 10 and 25 µM corosolic acid dramatically increased AMPK phosphorylation (Fig. 3B). However, corosolic acid failed to activate AMPK in 5-FU sensitive SNU-620 gastric cancer cells (data not shown). We also tested the status of mTOR/4EBP1, a downstream molecular marker of AMPK signaling. Activated AMPK inhibited the activation of mTOR/4EBP1 in corosolic acid treated SNU-620/5-FUR cells (Fig. 3B). Corosolic acid decreased the level of TS expression in a dose-dependent manner, with a pattern similar to that of AMPK activation (Fig. 3B). This suggests possible cross-talk between AMPK and 5-FU resistance after corosolic acid treatment. To determine whether corosolic acid-induced AMPK activation was associated with enhanced growth rate of SNU-620/5-FUR cells after treatment with compound c (40 µM) and/or corosolic acid (1, 10, 25 and 50 µM), cell viability was measured by CCK-8 assay. Results are shown in Fig. 3C. Compared to solo treatment with 10 or 25 µM corosolic acid, additional compound c treatment increased cell viabilities by 27.1 and 40%, respectively. These results suggest that corosolic acid-induced AMPK activation might be a mechanism involved in 5-FU resistance.

Corosolic acid resensitizes SNU-620/5-FUR gastric cancer cells to 5-FU

To investigate the sensitization effect of corosolic acid on 5-FU-resistant gastric cancer cells, SNU-620/5-FUR cells were treated with 5-FU (150 µM, 50% inhibitory concentration in 5-FU sensitive SNU-620 cells) and/or corosolic acid and growth rates were measured by CCK-8 assay. Results showed that single treatment with 150 µM of 5-FU or 25 µM of corosolic acid decreased the growth rate SNU-620/5-FUR cells by 93.4 and 42.7%, respectively. However, combination treatment significantly enhanced sensitivity of SNU-620/5-FUR cells to 5-FU (Fig. 4A). To estimate the sensitization effect of corosolic acid, 5-FU-sensitive cells were also exposed to 5-FU (150 µM) with or without corosolic acid. However, the combination effect observed in SNU-620/5-FUR cells was not evident in SNU-620 cells (Fig. 4A). In addition, TS expression in SNU-620/5-FUR cells was diminished by treatment with corosolic acid alone. It was drastically reduced following treatment with corosolic acid and 5-FU in combination (Fig. 4B and C). These results suggest that corosolic acid can probably sensitize 5-FU resistance.

Corosolic acid enhances sensitivity of SNU-620/5-FUR gastric cancer cells to 5-FU by upregulating AMPK

The effect of corosolic acid on AMPK activation in the presence or absence of 5-FU was tested in SNU-620/5-FUR cells. 5-FU (150 µM) moderately increased AMPK activation. A combination of corosolic acid (25 µM) with 5-FU was more effective. However, compound c significantly blocked AMPK phosphorylation (Fig. 5A and B). Compared to treatment with corosolic acid or 5-FU alone, the combination of corosolic acid and 5-FU resulted in increased caspase-3 and PARP cleavage and cytochrome c translocation, while compound c partly blocked 5-FU+corosolic acid-induced apoptotic activities (Fig. 5C). Apoptotic cell percentages were examined after treated with corosolic acid, 5-FU, or compound c, by flow cytometric analysis. A significant increase in the percentage of apoptotic cells was observed in SNU-620/5-FUR gastric cancer cells after treatment with corosolic acid in the presence of 5-FU. However, compound c significantly blocked apoptosis compared to a combination of corosolic acid and 5-FU (Fig. 5D). Percentages of early and late apoptotic/necrotic cells were increased from 0.5±0.1 to 21.7±2.0% in corosolic acid (25 µM) treated cells, 68.6±13.7% in cells treated with a combination of corosolic acid (25 µM) and 5-FU (150 µM), and 39.7±2.8% in cells treated with combination of corosolic acid (25 µM), 5-FU (150 µM), and compound c (40 µM) (Fig. 5E). Compound c significantly decreased the percentage of apoptotic cells compared to corosolic acid or 5-FU (Fig. 5D and E). These results suggest that corosolic acid-induced AMPK activation plays a key role in enhancing sensitivity of 5-FU-resistant gastric cancer cells to 5-FU.

Discussion

5-FU is a heterocyclic aromatic organic compound with a structure similar to that of pyrimidine molecules of DNA and RNA. It is an analogue of uracil with a fluorine atom at the C-5 position in place of hydrogen (3). 5-FU is used for treating gastric cancer. It can increase overall survival by 6% and reduce the risk of mortality by 18% (30). However, the occurrence of resistance to 5-FU treatment is a major problem for most gastric cancer patients, resulting in limited overall efficacy (31). High-level expression of TS, increased activity of deoxyuridine triphosphatase, methylation of MLH1 gene, and overexpression of Bcl-2, Bcl-XL, and Mcl-1 proteins have been reported to be associated with cancer resistance to 5-FU (3). Although the precise mechanism involved in gastric cancer resistance to 5-FU remains unknown, several reports have suggested that AMPK might be a biological predictor and beneficial target for cancer treatment through metabolic alteration (32). Previous clinical studies have shown that increases in phosphorylated AMPK is associated with tumor grade and prognosis for several solid tumors (33). More recently, phosphorylated AMPK levels have been reported as being significantly reduced in 5-FU-resistant gastric cancer cells (AGS cells) compared to 5-FU-sensitive cells (SGC-7901 cells) (12). In this study, we demonstrated that AMPK phosphorylation level was significantly decreased in 5-FU resistant gastric cancer cells (SNU-620/5-FUR) compared to sensitive gastric cancer cells (SNU-620). The present study aimed to identify alterative therapeutic approaches for enhancing 5-FU sensitivity by activating AMPK. TS expression level was decreased by AICAR, an AMPK activator. Consistent with this finding, treatment with the AMPK inhibitor, compound c, increased TS expression. AICAR significantly decreased viability of 5-FU resistant cells without altering viability of 5-FU sensitive cells (data not shown). Even though compound c is widely known as an AMPK inhibitor, this compound is involved in killing cancer cells by multiple mechanisms (Calpain/Cathepsin pathway; AKT; mTORC1/C2; cell cycle block; necroptosis; autophagy) (34). Liu et al (34) suggests that compound c kills cancer cells by an AMPK-independent mechanism. Therefore, we need to perform further research to find out why compound c did not increase cell viability in gastric cancer cells. These data suggest that AMPK might be a potent regulator of 5-FU resistance in gastric cancers.

Several studies have demonstrated that corosolic acid can activate AMPK in adipose tissue, endothelial cells, and gastric cancer cells (2325). A recent clinical study revealed that metformin, an AMPK activator, can reduce gastric cancer risk in patients with type 2 diabetes mellitus (35). In addition, metformin can reverse multidrug resistance in breast cancer cells and hepatocellular carcinoma Bel-7402/5-fluorourscil cells (10,11). In the present study, we found that corosolic acid activated AMPK phosphorylation in 5-FU resistant SNU-620/5-FUR cells followed by decreased phosphorylation levels of mTOR/4EBP1. Corosolic acid-induced AMPK activation down-regulated cell viability in a dose-dependent manner. However, AMPK activity was significantly inhibited by compound c in 5-FU resistant SNU-620/5-FUR cells. Therefore, corosolic acid-induced AMPK activation plays an important role in overcoming 5-FU-resistance in gastric cancer. In Fig. 3C, 1 µM corosolic acid + compound c (Lane 3) and 50 µM corosolic acid + compound c (Lane 9) did not preserve cell viability. 1 µM corosolic acid and 50 µM corosolic acid did not activate AMPK in our experiments, which might be a possible reason corosolic acid-treated cells could not be reversed by compound c. To investigate the sensitization effect of corosolic acid to 5-FU, resistant or sensitive cells were treated with corosolic acid (25 µM) in the presence or absence of 5-FU (150 µM). Viability of SNU-620/5-FUR cells treated with 5-FU in the presence of corosolic acid was significantly inhibited. However, no difference in viability of SNU-620 cells was found after such treatment. Corosolic acid in combination with 5-FU significantly decreased TS expression level in SNU-620/5-FUR cells compared to treatment with corosolic acid or 5-FU alone. Because TS is a major marker of 5-FU resistance status, these results indicate that corosolic acid might be able to reverse and/or sensitize 5-FU resistance of SNU-620/5-FUR cells.

AMPK activation is known to play an important role in enhancing chemosensitivity to certain chemotherapeutic agents such as 5-FU in breast cancer, intrahepatic cholangiocarcinoma, and gall bladder cancer (10,11,36). To investigate corosolic acid-induced AMPK activation involved in the resensitization effect of 5-FU, we determined AMPK phosphorylation level in the presence or absence of 5-FU. Our results showed that corosolic acid (10 and 25 µM) synergistically enhanced p-AMPK in the presence of 5-FU. Sensitivity of cells to chemotherapy might be estimated by examining apoptosis. The apoptotic rate was analyzed by western blot to detect caspase-3 and PARP cleavage, as well as cytochrome c translocation, and Annexin V-PI staining. The expression of cleaved caspase-3 and PARP and cytochrome c translocation in SNU-620/5-FUR cells were increased by 5-FU. The apoptotic rate in SNU-620/5-FUR cells treated with a combination of corosolic acid and 5-FU was significantly higher than when treated with corosolic acid alone. Chemically inhibited AMPK activation by compound c abolished AMPK phosphorylation and apoptotic activities in the presence of 5-FU. These findings suggest that corosolic acid might be able to enhance chemosensitivity of gastric cancer to 5-FU.

We agree with that gastric cancer is a heterogenous disease. To investigate the effect of corosolic acid on 5-FU chemoresistance in various gastric cancer cell types, several gastric cancer cell lines (SNU-1, SNU-5, AGS, SNU-484, SNU-601, and NCI-N87), have being established that are 5-FU resistant. Investigations of these are planned.

In conclusion, our study revealed that 5-FU resistant gastric cancer cells (SNU-620/5-FUR) had lower phosphorylated AMPK expression than 5-FU sensitive parental cells (SNU-620). Therefore, AMPK expression might be a possible treatment target for 5-FU resistant gastric cancers. We also found that corosolic acid could sensitize 5-FU resistance and inhibit viability of 5-FU resistant gastric cancer cells by activating the AMPK pathway. Therefore, corosolic acid could be used as an effective complimentary medicine to restore chemosensitivity of drug resistant gastric cancer cells to 5-FU. Further studies need to focus on chemotherapeutic sensitization by corosolic acid in combination with other chemotherapeutics, and to investigate the detailed molecular mechanisms involved.

Acknowledgements

The present study is based on a doctoral thesis (Chungnam National University, 2017).

Funding

The present study was supported by the Research Fund of Chungnam National University (grant no. 2015109701).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

JBP designed the study and prepared the manuscript. JSL contributed to the conception of the study, analyzed the data and drafted the manuscript. MSL, EYC and SK performed the experiments. JYS was involved in the study conception and design, and revised the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Smith JK, McPhee JT, Hill JS, Whalen GF, Sullivan ME, Litwin DE, Anderson FA and Tseng JF: National outcomes after gastric resection for neoplasm. Arch Surg. 142:387–393. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Tsai MM, Wang CS, Tsai CY, Chi HC, Tseng YH and Lin KH: Potential prognostic, diagnostic and therapeutic markers for human gastric cancer. World J Gastroenterol. 20:13791–13803. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Zhang N, Yin Y, Xu SJ and Chen WS: 5-Fluorouracil: Mechanisms of resistance and reversal strategies. Molecules. 13:1551–1569. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Scartozzi M, Maccaroni E, Giampieri R, Pistelli M, Bittoni A, Del Prete M, Berardi R and Cascinu S: 5-Fluorouracil pharmacogenomics: Still rocking after all these years? Pharmacogenomics. 12:251–265. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Johnston PG, Lenz HJ, Leichman CG, Danenberg KD, Allegra CJ, Danenberg PV and Leichman L: Thymidylate synthase gene and protein expression correlate and are associated with response to 5-fluorouracil in human colorectal and gastric tumors. Cancer Res. 55:1407–1412. 1995.PubMed/NCBI

6 

Saga Y, Suzuki M, Mizukami H, Urabe M, Fukushima M, Ozawa K and Sato I: Enhanced expression of thymidylate synthase mediates resistance of uterine cervical cancer cells to radiation. Oncology. 63:185–191. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Copur S, Aiba K, Drake JC, Allegra CJ and Chu E: Thymidylate synthase gene amplification in human colon cancer cell lines resistant to 5-fluorouracil. Biochem Pharmacol. 49:1419–1426. 1995. View Article : Google Scholar : PubMed/NCBI

8 

Fakhrejahani E, Miyamoto A and Tanigawa N: Correlation between thymidylate synthase and dihydropyrimidine dehydrogenase mRNA level and in vitro chemosensitivity to 5-fluorouracil, in relation to differentiation in gastric cancer. Cancer Chemother Pharmacol. 60:437–446. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Kim YH, Liang H, Liu X, Lee JS, Cho JY, Cheong JH, Kim H, Li M, Downey TJ, Dyer MD, et al: AMPKα modulation in cancer progression: Multilayer integrative analysis of the whole transcriptome in Asian gastric cancer. Cancer Res. 72:2512–2521. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Ling S, Tian Y, Zhang H, Jia K, Feng T, Sun D, Gao Z, Xu F, Hou Z, Li Y and Wang L: Metformin reverses multidrug resistance in human hepatocellular carcinoma Bel-7402/5-fluorouracil cells. Mol Med Rep. 10:2891–2897. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Qu C, Zhang W, Zheng G, Zhang Z, Yin J and He Z: Metformin reverses multidrug resistance and epithelial-mesenchymal transition (EMT) via activating AMP-activated protein kinase (AMPK) in human breast cancer cells. Mol Cell Biochem. 386:63–71. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Wu Y, Qi Y, Liu H, Wang X, Zhu H and Wang Z: AMPK activator AICAR promotes 5-FU-induced apoptosis in gastric cancer cells. Mol Cell Biochem. 411:299–305. 2016. View Article : Google Scholar : PubMed/NCBI

13 

Hua HW, Jiang F, Huang Q, Liao ZJ and Ding G: Re-sensitization of 5-FU resistance by SPARC through negative regulation of glucose metabolism in hepatocellular carcinoma. Tumour Biol. 36:303–313. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Hou W, Li Y, Zhang Q, Wei X, Peng A, Chen L and Wei Y: Triterpene acids isolated from Lagerstroemia speciosa leaves as alpha-glucosidase inhibitors. Phytother Res. 23:614–618. 2009. View Article : Google Scholar : PubMed/NCBI

15 

Thuong PT, Min BS, Jin W, Na M, Lee J, Seong R, Lee YM, Song K, Seong Y, Lee HK, et al: Anti-complementary activity of ursane-type triterpenoids from Weigela subsessilis. Biol Pharm Bull. 29:830–833. 2006. View Article : Google Scholar : PubMed/NCBI

16 

Fukushima M, Matsuyama F, Ueda N, Egawa K, Takemoto J, Kajimoto Y, Yonaha N, Miura T, Kaneko T, Nishi Y, et al: Effect of corosolic acid on postchallenge plasma glucose levels. Diabetes Res Clin Pract. 73:174–177. 2006. View Article : Google Scholar : PubMed/NCBI

17 

Miura T, Ueda N, Yamada K, Fukushima M, Ishida T, Kaneko T, Matsuyama F and Seino Y: Antidiabetic effects of corosolic acid in KK-Ay diabetic mice. Biol Pharm Bull. 29:585–587. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Xu Y, Zhao Y, Xu Y, Guan Y, Zhang X, Chen Y, Wu Q, Zhu G, Chen Y, Sun F, et al: Blocking inhibition to YAP by ActinomycinD enhances anti-tumor efficacy of Corosolic acid in treating liver cancer. Cell Signal. 29:209–217. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Sung B, Kang YJ, Kim DH, Hwang SY, Lee Y, Kim M, Yoon JH, Kim CM, Chung HY and Kim ND: Corosolic acid induces apoptotic cell death in HCT116 human colon cancer cells through a caspase-dependent pathway. Int J Mol Med. 33:943–949. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Nho KJ, Chun JM and Kim HK: Corosolic acid induces apoptotic cell death in human lung adenocarcinoma A549 cells in vitro. Food Chem Toxicol. 56:8–17. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Lee HS, Park JB, Lee MS, Cha EY, Kim JY and Sul JY: Corosolic acid enhances 5-fluorouracil-induced apoptosis against SNU-620 human gastric carcinoma cells by inhibition of mammalian target of rapamycin. Mol Med Rep. 12:4782–4788. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Lee MS, Cha EY, Thuong PT, Kim JY, Ahn MS and Sul JY: Down-regulation of human epidermal growth factor receptor 2/neu oncogene by corosolic acid induces cell cycle arrest and apoptosis in NCI-N87 human gastric cancer cells. Biol Pharm Bull. 33:931–937. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Yang J, Leng J, Li JJ, Tang JF, Li Y, Liu BL and Wen XD: Corosolic acid inhibits adipose tissue inflammation and ameliorates insulin resistance via AMPK activation in high-fat fed mice. Phytomedicine. 23:181–190. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Li Y, Zhou ZH, Chen MH, Yang J, Leng J, Cao GS, Xin GZ, Liu LF, Kou JP, Liu BL, et al: Inhibition of mitochondrial fission and NOX2 expression prevent NLRP3 inflammasome activation in the endothelium: The role of corosolic acid action in the amelioration of endothelial dysfunction. Antioxid Redox Signal. 24:893–908. 2016. View Article : Google Scholar : PubMed/NCBI

25 

Lee MS, Lee CM, Cha EY, Thuong PT, Bae K, Song IS, Noh SM and Sul JY: Activation of AMP-activated protein kinase on human gastric cancer cells by apoptosis induced by corosolic acid isolated from Weigela subsessilis. Phytother Res. 24:1857–1861. 2010. View Article : Google Scholar : PubMed/NCBI

26 

Goel A and Aggarwal BB: Curcumin, the golden spice from Indian saffron, is a chemosensitizer and radiosensitizer for tumors and chemoprotector and radioprotector for normal organs. Nutr Cancer. 62:919–930. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Tang H, Zhang X, Cui S, Wang J, Ruan Q, Huang Y and Yang D: Role and mechanism research on reversal of 5-fluorouracil resistance by epigallocatechin gallate in gastric cancer drug-resistance cells lines SGC-7901/5-FU. Zhonghua Wei Chang Wai Ke Za Zhi. 19:1170–1175. 2016.(In Chinese). PubMed/NCBI

28 

Liao F, Yang Z, Lu X, Guo X and Dong W: Sinomenine sensitizes gastric cancer cells to 5-fluorouracil in vitro and in vivo. Oncol Lett. 6:1604–1610. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Kato K, Gong J, Iwama H, Kitanaka A, Tani J, Miyoshi H, Nomura K, Mimura S, Kobayashi M, Aritomo Y, et al: The antidiabetic drug metformin inhibits gastric cancer cell proliferation in vitro and in vivo. Mol Cancer Ther. 11:549–560. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Kim DC, Park KR, Jeong YJ, Yoon H, Ahn MJ, Rho GJ, Lee J, Gong YD and Han SY: Resistance to the c-Met inhibitor KRC-108 induces the epithelial transition of gastric cancer cells. Oncol Lett. 11:991–997. 2016. View Article : Google Scholar : PubMed/NCBI

31 

Holohan C, Van Schaeybroeck S, Longley DB and Johnston PG: Cancer drug resistance: An evolving paradigm. Nat Rev Cancer. 13:714–726. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Wang W and Guan KL: AMP-activated protein kinase and cancer. Acta Physiol (Oxf). 196:55–63. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Hadad SM, Baker L, Quinlan PR, Robertson KE, Bray SE, Thomson G, Kellock D, Jordan LB, Purdie CA, Hardie DG, et al: Histological evaluation of AMPK signaling in primary breast cancer. BMC Cancer. 9:3072009. View Article : Google Scholar : PubMed/NCBI

34 

Liu X, Chhipa RR, Nakano I and Dasgupta B: The AMPK inhibitor compound C is a potent AMPK-independent antiglioma agent. Mol Cancer Ther. 13:596–605. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Tseng CH: Metformin reduces gastric cancer risk in patients with type 2 diabetes mellitus. Aging (Albany NY). 8:1636–1649. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Gao H, Xie J, Peng J, Han Y, Jiang Q, Han M and Wang C: Hispidulin inhibits proliferation and enhances chemosensitivity of gallbladder cancer cells by targeting HIF-1α. Exp Cell Res. 332:236–246. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2018
Volume 18 Issue 3

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
Park JB, Lee JS, Lee MS, Cha EY, Kim S and Sul JY: Corosolic acid reduces 5‑FU chemoresistance in human gastric cancer cells by activating AMPK. Mol Med Rep 18: 2880-2888, 2018
APA
Park, J.B., Lee, J.S., Lee, M.S., Cha, E.Y., Kim, S., & Sul, J.Y. (2018). Corosolic acid reduces 5‑FU chemoresistance in human gastric cancer cells by activating AMPK. Molecular Medicine Reports, 18, 2880-2888. https://doi.org/10.3892/mmr.2018.9244
MLA
Park, J. B., Lee, J. S., Lee, M. S., Cha, E. Y., Kim, S., Sul, J. Y."Corosolic acid reduces 5‑FU chemoresistance in human gastric cancer cells by activating AMPK". Molecular Medicine Reports 18.3 (2018): 2880-2888.
Chicago
Park, J. B., Lee, J. S., Lee, M. S., Cha, E. Y., Kim, S., Sul, J. Y."Corosolic acid reduces 5‑FU chemoresistance in human gastric cancer cells by activating AMPK". Molecular Medicine Reports 18, no. 3 (2018): 2880-2888. https://doi.org/10.3892/mmr.2018.9244