Quercetin induces apoptosis by inhibiting MAPKs and TRPM7 channels in AGS cells

  • Authors:
    • Min Chul Kim
    • Hee Jung Lee
    • Bora Lim
    • Ki-Tae Ha
    • Sung Young Kim
    • Insuk So
    • Byung Joo Kim
  • View Affiliations

  • Published online on: March 18, 2014     https://doi.org/10.3892/ijmm.2014.1704
  • Pages: 1657-1663
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The worldwide incidence and mortality rate of gastric cancer remain high, and thus, novel treatment concepts are required. Quercetin, a bioflavonoid, has been proposed to have anti-cancer properties. The aim of this study was to determine the nature of the apoptotic mechanisms responsible for the effects of quercetin on AGS cells (a commonly used human gastric adenocarcinoma cell line). AGS cell viability was assessed by MTT assay and flow cytometric analysis, mitochondrial membrane depolarization was assessed, and caspase-3 was used to determine the involvement of apoptosis. Whole-cell configuration patch-clamp experiments were used to regulate the transient receptor potential melastatin (TRPM)7 channels. To investigate the signaling pathway of quercetin-induced apoptosis in the AGS cells, western blot analysis and MTT assay were performed. Quercetin was found to induce the apoptosis of these cells, and this apoptosis was inhibited by SB203580 (a p38 kinase inhibitor), SP600125 (a JNK inhibitor) and PD98059 (an ERK inhibitor). In addition, quercetin inhibited TRPM7 currents in the AGS cells and in human embryo kidney (HEK)293 cells which overexpress TRPM7 channels. Furthermore, treatment with quercetin increased the apoptosis of HEK293 cells, which overexpress TRPM7, indicating that the upregulation of TRPM7 channels underlies quercetin-induced cell death. These results suggest that quercetin plays an important pathophysiological role in AGS cells through mitogen‑activated protein kinase (MAPK) signaling pathways and TRPM7 channels, and that quercetin has potential as a pharmacological agent for the treatment of gastric cancer.

Introduction

Gastric cancer is responsible for almost one million deaths worldwide per year. Although the age-adjusted mortality of gastric cancer has decreased over the last few decades, gastric cancer remains the second leading cause of cancer-related mortatlity (1). Gastric cancer is an asymptomatic disease during the early stages, and therefore, is often detected at a late stage. The 5-year survival rate of gastric cancer is only 20–30% (2). For these reasons, intensive research is required, in order to devise new approaches to its diagnosis and treatment. Apoptosis is a key factor that contributes to the antitumor activity of chemotherapeutic drugs and therefore, plays a key role in the search for new approaches for the treatment of cancer. In addition, as cell numbers can be regulated by factors that influence cell survival, proliferation and differentiation, apoptosis is a gene-directed program that has had profound implications for our understanding of developmental biology and tissue homeostasis (3). Most anticancer agents now in use were developed using empirical screens designed to identify agents that selectively kill tumor cells (4). It is now well-established that anticancer agents induce apoptosis, and that the disruption of apoptotic programs can reduce treatment sensitivity (4). Anticancer agents induce apoptosis in normal tissues, as well as in tumors. In fact, many of the pathologists who identified apoptosis in tumors realized that apoptotic cell death was induced in a subset of normal tissues (e.g., bone marrow and the intestines), and it was suggested that the process may contribute to the ‘toxicity’ associated with chemotherapy (3). Although there is much more to learn, our current understanding of apoptosis provides new avenues for cancer diagnostics, prognosis and therapy. It seems likely that rational strategies to manipulate cell suicide programs will produce novel therapies that are less toxic and mutagenic than current treatment regimens (3).

Quercetin (3,3′,4′,5,7-pentahydroxyflavone) is a bioflavonoid, and is one of the most widely distributed dietary polyphenolics; for example, it is found in vegetables, fruits, tea and wine (5). Research into the anticarcinogenic potential of flavonoids with animal and cellular model systems supports a protective role against the progression of colorectal cancer (69). Apart from other flavonoids, quercetin has been proposed to have anti-atherogenic, anti-inflammatory, antihypertensive, and anticancer effects (3). Quercetin has been reported to protect against important diseases, including lung and colon cancers (1012). In addition, quercetin has been shown to inhibit human breast cancer cell proliferation and to induce apoptosis through the regulation of Bcl-2 and Bax (13).

However, the effects of quercetin on human gastric adenocarcinoma cells have not yet been fully elucidated. Therefore, in this study, we investigated the effects of quercetin in human gastric adenocarcinoma cells and the mechanisms responsible for its activities.

Materials and methods

Materials

Quercetin was purchased from Alexis Biochemicals (Vinci Biochem, Florence, Italy) and PD98059 (ERK inhibitor), SB203580 (p38 kinase inhibitor), and SP600125 (JNK inhibitor) were purchased from Tocris Bioscience (Bristol, UK). All other agents were purchased from Sigma-Aldrich (St. Louis, MO, USA).

Cells

The AGS cell line (a human gastric adenocarcinoma cell line) was established at the Cancer Research Center, College of Medicine, Seoul National University, Seoul, Korea, and propagated in RPMI-1640 medium (Gibco-BRL, Carlsbad, CA, USA) supplemented with 10% heat-inactivated fetal bovine serum containing 20 μg/ml penicillin and streptomycin in a 5% CO2 atmosphere at 37°C.

3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay

Cell viability was assessed by MTT assay. The AGS or human embryo kidney (HEK)293 cells were seeded into each well of 12-well culture plates and then cultured in RPMI-1640 supplemented with 10% heat-inactivated fetal bovine serum containing 20 μg/ml penicillin and streptomycin for 72 h. MTT solution [100 μl; 5 mg/ml in phosphate-buffered saline (PBS)] was then added to each well, and the plates were incubated at 37°C for 4 h. After removing the supernatant and shaking with 200 μl of dimethyl sulfoxide (Jersey Lab Supply, Livingston, NJ, USA) for 30 min, and absorbances were measured at 570 nm. All experiments were repeated at least 3 times.

Flow cytometric analysis

In order to investigate the effects of treatment on the cell cycle of the AGS cells, flow cytometric analysis was performed with propidium iodine (PI) staining, as previously described (14,15). The cells (1×106) were placed in an Eppendorf tube, and 700 μl of ice-cold fixation buffer (ethyl alcohol) was slowly added with vortexing. The tubes were sealed with parafilm, incubated at 4°C overnight, spun for 3 min at 106 × g at 4°C, and the supernatants were aspirated and discarded. Cell pellets were resuspended in 200 μl of PI staining solution [2 μl of PI (5 mg/ml) and 2 μl of RNase in a total of 196 μl of PBS] and centrifuged at 2,0817 × g for 5 sec. After being kept for 30 min in the dark at room temperature, the samples were analyzed using a fluorescence-activated cell sorter (FACScan; Becton-Dickinson, Mountain View, CA, USA) at λ=488 nm using Cell-Quest software (Becton-Dickinson). The DNA content distribution of normally growing AGS cells is characterized by 2 peaks, representing the G1/G0 and G2/M phases. The G1/G0 phase is composed of cells in the resting state of the cell cycle and has the greatest diploid DNA content, whereas the DNA content in the G2/M phase is more than diploid. Cells in the sub-G1 phase have the least DNA content, which is termed hypodiploid (a hypoploid DNA content represents DNA fragmentation) (16).

Assessment of mitochondrial membrane depolarization

Mitochondrial membrane depolarization was evaluated using the JC-1 fluorescence probe according to the manufacturer’s instructions (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). The AGS cells were labeled with 2 μM JC-1 for 30 min at 37°C and then analyzed by flow cytometry at excitation and emission wavelengths of 530/30 or 585/42 nm and 488 nm, respectively. The cells without red fluorescence were regarded to manifest mitochondrial membrane depolarization.

Caspase assay

Caspase-3 kits (Cellular Activity Assay kit PLUS) were purchased from Biomol International (Plymouth Meeting, PA, USA). After the treatments, the cells were centrifuged (10,000 × g, 4°C, 10 min), washed with PBS, resuspended in ice-cold cell lysis buffer, incubated on ice for 10 min, centrifuged at 10,000 × g (4°C, 10 min), and the supernatants were removed. The supernatants (10 μl) were then incubated with 50 μl of substrate (400-μM Ac-DEVD-pNA) in 40 μl of assay buffer at 37°C. Absorbance was read at 405 nm at several time points. The pNA concentration in the samples was determined using a standard pNA concentration vs. absorbance plot. The pan-caspase inhibitor, zVAD-fmk (Calbiochem, La Jolla, CA, USA), was used to validate the assay method.

Patch-clamp experiments

Whole-cell configuration patch-clamp experiments were performed at room temperature (22–25°C). The AGS cells were transferred to a small chamber on an inverted microscope stage (IX70; Olympus, Tokyo, Japan), and constantly perfused with a solution containing (mmol/l) KCl 2.8, NaCl 145, CaCl2 2, glucose 10, MgCl2 1.2 and HEPES 10 (adjusted to pH 7.4 with NaOH). The pipette solution contained (mmol/l) Cs-glutamate 145, NaCl 8, Cs-2-bis(2-aminophenoxy)-ethane-N,N,N′,N′-tetraacetic acid 10 and HEPES-CsOH 10 (adjusted to pH 7.2 with CsOH). An Axopatch I-D (Axon Instruments, Foster City, CA, USA) was used to amplify the membrane currents and potentials. pClamp software version 9.2 and Digidata 1322A (Axon Instruments) were used for data acquisition and to apply command pulses. The results were analyzed using pClamp and Origin software (Microcal Origin version 6.0).

Transient receptor potential melastatin (TRPM)7 expression in HEK293 cells

The HEK293 cells were transfected with Flag-murine LTRPC7/pCDNA4-TO construct and grown on glass coverslips in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal bovine serum, blasticidin (5 μg/ml) and zeocin (0.4 mg/ml). TRPM7 (LTRPC7) expression was induced by the addition of 1 μg/ml tetracycline to the culture medium. Whole-cell patch-clamp experiments were performed at 21–25°C using cells grown on glass coverslips.

Sodium dodecyl sulfate-polyacrylamide gel electrophoresis and western blot analysis

Western blot analysis was performed using AGS cell lysates. Proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis by using 8% polyacrylamide gels, transferred onto polyvinylidene difluoride membranes, and quantified by incubating the membranes with the indicated antibodies. Antibodies against poly(ADP-ribose) polymerase (PARP), p-p38, p-JNK, p-ERK, p38, ERK and β-actin were purchased from Cell Signaling Technology (Danvers, MA, USA). Antibodies against JNK and GAPDH were obtained from Santa Cruz Biotechnology. The data are representative of at least 3 independent experiments.

Statistical analysis

Data are expressed as the means ± SEM. The Student’s t-test was used to determine the significances of the differences. Statistical significance was set at P<0.05.

Results

Quercetin induces AGS cell death

To ascertain whether quercetin kills AGS cells, we performed MTT assays. Viable cell populations were gradually reduced by quercetin in a concentratio- and a time-dependent manner at 24, 48 and 72 h with an IC50 value of 267 μM (Fig. 1).

Quercetin induces apoptosis of AGS cells

To determine whether the AGS-induced cell death was due to apoptosis, we conducted sub-G1 analysis, as previously described (17,18). Briefly, the cells were incubated with quercetin and stained with a fluorescent DNA stain (PI). Endogenous endonucleases in apoptotic cells cleave DNA into endonucleosomal fragments. The loss of DNA is detected by FACS analysis, as a reduction of nuclear staining in apoptotic cells, which results in a novel (sub-G1) fluorescence peak to the left of the regular fluorescence peak. Flow cytometric analysis revealed that the percentage of cells in the sub-G1 phase markedly increased in a dose-dependent manner in the cells treated with quercetin (Fig. 2A). In addition, quercetin elevated mitochondrial membrane depolarization, an early event of intrinsic apoptotic signaling (Fig. 2B). Thus, our findings suggest that quercetin induces apoptosis through intrinsic apoptotic mechanism(s). In addition, using a synthetic substrate, we examined caspase-3 activity (caspase-3 activation is a hallmark of apoptotic cell death) in the AGS cells. Quercetin increased the activity of caspase-3, which was suppressed by zVAD-fmk (a pan-caspase inhibitor) (Fig. 2C). We also examined changes in caspase-3 activity by western blot analysis for its natural substrate, PARP, which has been shown to function as a cellular target of caspase-3 and other caspases. During apoptosis, PARP is proteolytically cleaved from its 116-kDa-intact form into 85 and 25 kDa fragments. Following incubation with quercetin for 72 h, the AGS cells showed increased PARP cleavage (Fig. 2D).

Involvement of mitogen-activated protein kinases (MAPKs) in quercetin-induced apoptosis of AGS cells

To investigate the signaling pathway of quercetin-induced apoptosis in AGS cells, we assessed the effects of quercetin on MAPKs, as they play critical roles in the apoptosis-related signaling pathway. As shown in Fig. 3, quercetin markedly increased the phosphorylation levels of p38, JNK and ERK (Fig. 3). As the role of MAPK signaling in the quercetin induced apoptosis of AGS cells has been little characterized in this study (Fig. 3), we performed MTT assays using MAPK inhibitors to assess the effects of MAPK signaling on quercetin signaling. MTT assays revealed that exposure to quercetin with SB203580 (Fig. 4A), SP600125 (Fig. 4B) and PD98059 (Fig. 4C) resulted in an increase in the viable cell populations.

Effects of quercetin in TRPM7 currents on AGS cells and HEK293 cells overexpressing TRPM7

In a previous study, we found that AGS cells express the TRPM7 channel, which is known to be essential for cell survival and has been suggested as a pharmacological target for the treatment of gastric cancer (19). Therefore, in this study, we investigated whether quercetin influences TRPM7 currents in AGS cells. To confirm the effects of quercetin on TRPM7 currents, we investigated the effects of quercetin in AGS cells using patch-clamp techniques. First, to examine the effects of internal Mg2+ on TRPM7-like currents, Mg2+ was added to the pipette solution at different concentrations from 0 to 2 mM. Current amplitudes were indeed larger at the lower concentration, and the presence of 2 mM Mg2+ in the pipette solution significantly inhibited the current (n=4; Fig. 5A). Whole cell voltage-clamp recordings were then taken to investigate the effects of quercetin on TRPM7-like currents in AGS cells. A voltage ramp from +100 to −100 mV evoked small inward currents at negative potentials, whereas larger outward currents were evoked at positive potentials, showing outwardly rectifying cation currents (n=6; Fig. 5B). However, in the presence of 500 μM quercetin, the amplitudes of these currents were inhibited outwardly by 79.3±1.3% and inwardly by 89.2±1.4% (n=6; Fig. 5B). Furthermore, similar results were obtained withthe HEK293 cells overexpressing TRPM7 channels (Fig. 5C). To provide additional evidence supporting the positive contribution made by TRPM7 channels to quercetin toxicity, we investigated the effects of different TRPM7 channel levels on quercetin-mediated cell death. For this purpose, we used HEK293 cells with inducible TRPM7 channel expression (20,21). In the absence of induced TRPM7 channel expression [TRPM7(−) cells, Tet(−)], quercetin did not induce HEK293 cell death, as shown by MTT assay (n=6; Fig. 5D). However, when TRPM7 channel overexpression was induced by the addition of tetracycline [TRPM7(+) cells, Tet(+)], quercetin markedly induced more HEK293 cell death, indicating that TRPM7 channel levels are positively associated with quercetin-induced cell death.

Discussion

Quercetin is a polyphenolic flavonoid that is found in fruits and vegetables, such as, apples, blueberries, broccoli, grapes, leeks, lettuce, onions and tomatoes. The mechanisms underlying the cancer-preventive effects of quercetin and other flavonoids are believed to be associated with antioxidant effects, the inhibition of enzymes that activate carcinogens, the modification of signal transduction pathways, and interactions with receptors and/or proteins (22,23). However, the effects of quercetin on AGS human adenocarcinoma cells have not yet been fully elucidated. Quercetin has been shown to exert antiproliferative effects on the parental EPG85-257P cell line and its daunorubicin-resistant variant, EPG85-257RDB, mainly through the induction of apoptosis (24). Quercetin has also been shown to induce the apoptosis of BGC-823 cells and decrease the Bcl-2/Bax ratio with the increased expression of caspase-3 (25). As previously demonstrated, quercetin induced pronounced pro-apoptotic effects by activating the mitochondrial pathway in AGS and MKN28 gastric cancer cells. Treatment with quercetin induced the appearance of autophagic vacuoles, the formation of acidic vesicular organelles, the conversion of LC3-I to LC3-II, the recruitment of LC3-II to the autophagosomes, as well as the activation of autophagy genes, suggesting that quercetin initiates autophagic progression in gastric cancer cells (26). However, to our knowledge, there are no studies available on the association of MAPK and TRP channels in quercetin-induced apoptosis in gastric cancer cells.

Certain studies have suggested that MAPK activation plays a role in the apoptosis of cancer cells (27,28). PKI 166 induced apoptosis through the activation of the p53, MAPK and caspase pathway in epidermoid carcinoma (27). In addition, isoorientin induced mitochondrial-mediated apoptosis through MAPK signaling pathways in human hepatoblastoma cancer cells (28). Thus, in this study, we investigated whether MAPKs play a role in quercetin-induced apoptosis in AGS cells. The 3 principal ubiquitously expressed MAPKs are p38 MAPK, JNK and ERK and JNK (29), and in the present study, inhibitors of p38, JNK and ERK inhibited AGS cell apoptosis induced by quercetin.

TRP channels were first cloned from the Drosophila species, and constitute a superfamily of proteins that encode a diverse group of Ca2+-permeable non-selective cation channels (NSCCs) (30). Based on their amino acid sequences, the TRP superfamily can be divided into 7 subfamilies: the TRPC (‘Canonical’), TRPM (‘Melastatin’), TRPV (‘Vanilloid’), TRPP (‘Polycystin’), TRPML (‘Mucolipin’), TRPA (‘Ankyrin’) and the TRPN (‘NOMPC’) families (31). TRPM7, a member of the TRPM ion channel subfamily, is a widely expressed bifunctional protein with both ion channel and α-kinase domains (20,21). In a previous study [Kim et al (19)], we reported that human gastric adenocarcinoma cells express TRPM7 channels and that this channel is essential for cell survival. It was also suggested that this channel be viewed as a potential target for the pharmacological treatment of gastric cancer.

The TRPM7 channel is widely expressed in various tissues, including those of the brain, spleen, kidneys, lungs, liver and heart (20,32). Furthermore, its expression has also been reported in head and neck carcinoma (21), retinoblastoma (33), breast cancer (34,35) and gastric cancer (19). The activation of TRPM7 channels in human head and neck squamous carcinoma cell lines enhances proliferation (21), whereas TRPM7 knockdown by siRNA and the blockade of TRPM7 channels by Gd3+ or 2-APB, which exerts broad inhibitory effects on TRP superfamilies, including TRPM7, suppresses proliferation. The proliferation of human retinoblastoma cells is promoted by increasing spontaneous Ca2+ influx (33). Furthermore, the blockade of cation channel by Gd3+, La3+, LOE908, or 2-APB inhibits spontaneous Ca2+ influx and decreases the proliferation of retinoblastoma cells. TRPM7 channel-related proliferation is also associated with MCF-7 human breast cancer cells (35) and AGS human gastric adenocarcinoma cells (19). As previously demonstrated, the proliferation of both cells was prevented by TRPM7 protein knockout by siRNA transfection and by blocking TRPM7 channels with La3+ or 2-APB. Thus, TRPM7 channels may play an important role in valuable diagnostic and/or prognostic markers, as well as targets for pharmaceutical intervention and more in depth investigations are required to elucidate the role TRPM7 channels in cancer (21).

In the present study, we found that quercetin inhibited TRPM7-like currents in AGS cells (Fig. 5B). To investigate the the effects of quercetin on TRPM7 channels, we overexpressed TRPM7 channels in HEK293 cells. It was found that quercetin inhibited the TRPM7 currents (Fig. 5C), and that its inhibitory effects were similar to those in the AGS cells. Therefore, we hypothesized that TRPM7 is involved in the apoptotic activity of quercetin in AGS cells.

TRPM7 has also been suggested to play a central role in: cellular Mg2+ homeostasis (36), central nervous system ischemic injury (37), skeletogenesis in zebrafish (38), defecation rhythm in Caenorhabditis elegans (39), cholinergic vesicle fusion with the plasma membrane (40), phosphoinositide-3-kinase signaling in lymphocytes (41), and in osteoblast proliferation (42).

Apoptotic pathways are induced by a variety of signals, and a certain mechanism consistently implicated in apoptosis is the activation of a series of cytosolic proteases, that is, the caspases (43). Caspases are synthesized as inactive pro-enzymes that are processed in cells undergoing apoptosis by self-proteolysis and/or cleavage by another protein. Functionally, active caspases form a proteolytic cascade, which is capable of cleaving and activating specific substrates, including PARP (an enzyme involved in DNA repair and genomic maintenance). Cleavage events of this type may result in important alterations to normal homeostatic cellular processes (44). Furthermore, PARP has been shown to be a cellular target of caspase-3 and of other caspases. In our study, After 72 h of incubation with quercetin, PARP became proteolytically cleaved in AGS cells from its 116-kDa intact form into 85-kDa fragments (Fig. 2D), and caspase-3 activity increased accordingly (Fig. 2C).

In addition, we found that quercetin inhibited cell viability and that MAPK signaling pathways were involved in AGS cell apoptosis (Figs. 3 and 4). Furthermore, TRPM7 currents in AGS cells and in HEK293 cells which overexpress TRPM7 were inhibited by quercetin (Fig. 5B and C). Furthermore, in TRPM7-overexpressing HEK293 cells, quercetin induced cell death at a greater rate, suggesting that the upregulation of TRPM7 channels increases quercetin-induced cell death. These results suggest that quercetin plays an important pathophysiological role in AGS cells through MAPK signaling and TRPM7 channels. Accordingly, we suggest that quercetin be viewed as a potential drug for the pharmacological treatment of gastric cancer.

Acknowledgements

This study was supported by the Basic Science Research Program of the Korean National Research Foundation (NRF) funded by the Ministry of Education, Science and Technology (Grant no. 2010-0021347).

References

1 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Bosman FT, Carneiro F, Hruban RH and Theise ND: WHO Classification of Tumours of the Digestive System. 3. 4th edition. IARC Press; Geneva: 2010

3 

Lowe SW and Lin AW: Apoptosis in cancer. Carcinogenesis. 21:485–495. 2000. View Article : Google Scholar

4 

Schmitt CA and Lowe SW: Apoptosis and therapy. J Pathol. 187:127–137. 1999. View Article : Google Scholar

5 

Ishizawa K, Yoshizumi M, Kawai Y, Terao J, Kihira Y, Ikeda Y, Tomita S, Minakuchi K, Tsuchiya K and Tamaki T: Pharmacology in health food: metabolism of quercetin in vivo and its protective effect against arteriosclerosis. J Pharmacol Sci. 115:466–470. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Williams RJ, Spencer JPE and Rice-Evans C: Flavonoids: antioxidants or signaling molecules? Free Rad Biol Med. 36:838–849. 2004. View Article : Google Scholar : PubMed/NCBI

7 

Daskiewicz JB, Depeint F, Viornery L, Bayet C, Comte-Sarrazin G, Comte G, Gee JM, Johnson IT, Ndjoko K, Hostettmann K and Barron D: Effects of flavonoids on cell proliferation and caspase activation in a human colonic cell line HT29: an SAR study. J Med Chem. 48:2790–2804. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Surh YJ, Kundu JK, Na HK and Lee JS: Redox-sensitive transcription factors as prime targets for chemoprevention with anti-inflammatory and antioxidative phytochemicals. J Nutr. 135:S2993–S3001. 2005.PubMed/NCBI

9 

Kyle JA, Sharp L, Little J, Duthie GG and McNeill G: Dietary flavonoid intake and colorectal cancer: a case-control study. Br J Nutr. 103:429–436. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Stefani ED, Boffetta P, Deneo-Pellegrini H, Mendilaharsu M, Carzoglio JC, Ronco A and Olivera L: Dietary antioxidants and lung cancer risk: a case-control study in Uruguay. Nutr Cancer. 34:100–110. 1999. View Article : Google Scholar : PubMed/NCBI

11 

van Erk MJ, Roepman P, van der Lende TR, Stierum RH, Aarts JM, van Bladeren PJ and van Ommen B: Integrated assessment by multiple gene expression analysis of quercetin bioactivity on anticancer-related mechanisms in colon cancer cells in vitro. Eur J Nutr. 44:143–156. 2005.PubMed/NCBI

12 

Dihal AA, de Boer VC, van der Woude H, Tilburgs C, Bruijntjes JP, Alink GM, Rietjens IM, Woutersen RA and Stierum RH: Quercetin, but not its glycosidated conjugate rutin, inhibits azoxymethane-induced colorectal carcinogenesis in F344 rats. J Nutr. 136:2862–2867. 2006.PubMed/NCBI

13 

Duo J, Ying GG, Wang GW and Zhang L: Quercetin inhibits human breast cancer cell proliferation and induces apoptosis via Bcl-2 and Bax regulation. Mol Med Rep. 5:1453–1456. 2012.PubMed/NCBI

14 

Schönherr R: Clinical relevance of ion channels for diagnosis and therapy of cancer. J Membr Biol. 205:175–184. 2005.PubMed/NCBI

15 

Lehen’kyi V, Shapovalov G, Skryma R and Prevarskaya N: Ion channels and transporters in cancer. 5 Ion channels in control of cancer and cell apoptosis. Am J Physiol Cell Physiol. 301:C1281–C1289. 2011.PubMed/NCBI

16 

Wang BJ, Won SJ, Yu ZR and Su CL: Free radical scavenging and apoptotic effects of Cordyceps sinensis fractionated by supercritical carbon dioxide. Food Chem Toxicol. 43:543–552. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Hotz MA, Gong J, Traganos F and Darzynkiewicz Z: Flow cytometric detection of apoptosis: comparison of the assays of in situ DNA degradation and chromatin changes. Cytometry. 15:237–244. 1994. View Article : Google Scholar : PubMed/NCBI

18 

Vermes I, Haanen C and Reutelingsperger C: Flow cytometry of apoptotic cell death. J Immunol Methods. 243:167–190. 2000. View Article : Google Scholar : PubMed/NCBI

19 

Kim BJ, Park EJ, Lee JH, Jeon JH, Kim SJ and So I: Suppression of transient receptor potential melastatin 7 channel induces cell death in gastric cancer. Cancer Sci. 99:2502–2509. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Nadler MJ, Hermosura MC, Inabe K, Perraud AL, Zhu Q, Stokes AJ, Kurosaki T, Kinet JP, Penner R, Scharenberg AM and Fleig A: LTRPC7 is a Mg.ATP-regulated divalent cation channel required for cell viability. Nature. 411:590–595. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Jiang J, Li MH, Inoue K, Chu XP, Seeds J and Xiong ZG: Transient receptor potential melastatin 7-like current in human head and neck carcinoma cells: role in cell proliferation. Cancer Res. 67:10929–10938. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Daker M, Ahmad M and Khoo AS: Quercetin-induced inhibition and synergistic activity with cisplatin - a chemotherapeutic strategy for nasopharyngeal carcinoma cells. Cancer Cell Int. 12:342012. View Article : Google Scholar : PubMed/NCBI

23 

Murakami A, Ashida H and Terao J: Multitargeted cancer prevention by quercetin. Cancer Lett. 269:315–325. 2008. View Article : Google Scholar

24 

Borska S, Chmielewska M, Wysocka T, Drag-Zalesinska M, Zabel M and Dziegiel P: In vitro effect of quercetin on human gastric carcinoma: targeting cancer cells death and MDR. Food Chem Toxicol. 50:3375–3383. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Wang P, Zhang K, Zhang Q, Mei J, Chen CJ, Feng ZZ and Yu DH: Effects of quercetin on the apoptosis of the human gastric carcinoma cells. Toxicol In Vitro. 26:221–228. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Wang K, Liu R, Li J, Mao J, Lei Y, Wu J, Zeng J, Zhang T, Wu H, Chen L, Huang C and Wei Y: Quercetin induces protective autophagy in gastric cancer cells: involvement of Akt-mTOR- and hypoxia-induced factor 1α-mediated signaling. Autophagy. 7:966–978. 2011.PubMed/NCBI

27 

Das S, Dey KK, Bharti R, MaitiChoudhury S, Maiti S and Mandal M: PKI 166 induced redox signalling and apoptosis through activation of p53, MAP kinase and caspase pathway in epidermoid carcinoma. J Exp Ther Oncol. 10:139–153. 2012.

28 

Yuan L, Wang J, Xiao H, Wu W, Wang Y and Liu X: MAPK signaling pathways regulate mitochondrial-mediated apoptosis induced by isoorientin in human hepatoblastoma cancer cells. Food Chem Toxicol. 53:62–68. 2013. View Article : Google Scholar : PubMed/NCBI

29 

Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berman K and Cobb MH: Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev. 22:153–183. 2001.PubMed/NCBI

30 

Clapham DE: TRP channels as cellular sensors. Nature. 426:517–524. 2003. View Article : Google Scholar : PubMed/NCBI

31 

Pedersen SF, Owsianik G and Nilius B: TRP channels: an overview. Cell Calcium. 38:233–252. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Runnels LW, Yue L and Clapham DE: TRP-PLIK, a bifunctional protein with kinase and ion channel activities. Science. 291:1043–1047. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Hanano T, Hara Y, Shi J, Morita H, Umebayashi C, Mori E, Sumimoto H, Ito Y, Mori Y and Inoue R: Involvement of TRPM7 in cell growth as a spontaneously activated Ca2+ entry pathway in human retinoblastoma cells. J Pharmacol Sci. 95:403–419. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Dhennin-Duthille I, Gautier M, Faouzi M, Guilbert A, Brevet M, Vaudry D, Ahidouch A, Sevestre H and Ouadid-Ahidouch H: High expression of transient receptor potential channels in human breast cancer epithelial cells and tissues: correlation with pathological parameters. Cell Physiol Biochem. 28:813–822. 2011. View Article : Google Scholar

35 

Guilbert A, Gautier M, Dhennin-Duthille I, Haren N, Sevestre H and Ouadid-Ahidouch H: Evidence that TRPM7 is required for breast cancer cell proliferation. Am J Physiol Cell Physiol. 297:C493–C502. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Schmitz C, Perraud AL, Johnson CO, Inabe K, Smith MK, Penner R, Kurosaki T, Fleig A and Scharenberg AM: Regulation of vertebrate cellular Mg2+ homeostasis by TRPM7. Cell. 114:191–200. 2003. View Article : Google Scholar : PubMed/NCBI

37 

Aarts M, Iihara K, Wei WL, Xiong ZG, Arundine M, Cerwinski W, MacDonald JF and Tymianski M: A key role for TRPM7 channels in anoxic neuronal death. Cell. 115:863–877. 2003. View Article : Google Scholar : PubMed/NCBI

38 

Elizondo MR, Arduini BL, Paulsen J, MacDonald EL, Sabel JL, Henion PD, Cornell RA and Parichy DM: Defective skeletogenesis with kidney stone formation in dwarf zebrafish mutant for trpm7. Curr Biol. 15:667–671. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Vriens J, Owsianik G, Voets T, Droogmans G and Nilius B: Invertebrate TRP proteins as functional models for mammalian channels. Pflugers Arch. 449:213–226. 2004.PubMed/NCBI

40 

Brauchi S, Krapivinsky G, Krapivinsky L and Clapham DE: TRPM7 facilitates cholinergic vesicle fusion with the plasma membrane. Proc Natl Acad Sci USA. 105:8304–8308. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Sahni J and Scharenberg AM: TRPM7 ion channels are required for sustained phosphoinositide 3-kinase signaling in lymphocytes. Cell Metab. 8:84–93. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Abed E and Moreau R: Importance of melastatin-like transient receptor potential 7 and cations (magnesium, calcium) in human osteoblast-like cell proliferation. Cell Prolif. 40:849–865. 2007. View Article : Google Scholar : PubMed/NCBI

43 

Patel T, Gores GJ and Kaufmann SH: The role of proteases during apoptosis. FASEB J. 10:587–597. 1996.

44 

Lin C, Holland RE Jr, Donofrio JC, McCoy MH, Tudor LR and Chambers TM: Caspase activation in equine influenza virus induced apoptotic cell death. Vet Microbiol. 84:357–365. 2002. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2014
Volume 33 Issue 6

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
Kim MC, Lee HJ, Lim B, Ha K, Kim SY, So I and Kim BJ: Quercetin induces apoptosis by inhibiting MAPKs and TRPM7 channels in AGS cells. Int J Mol Med 33: 1657-1663, 2014
APA
Kim, M.C., Lee, H.J., Lim, B., Ha, K., Kim, S.Y., So, I., & Kim, B.J. (2014). Quercetin induces apoptosis by inhibiting MAPKs and TRPM7 channels in AGS cells. International Journal of Molecular Medicine, 33, 1657-1663. https://doi.org/10.3892/ijmm.2014.1704
MLA
Kim, M. C., Lee, H. J., Lim, B., Ha, K., Kim, S. Y., So, I., Kim, B. J."Quercetin induces apoptosis by inhibiting MAPKs and TRPM7 channels in AGS cells". International Journal of Molecular Medicine 33.6 (2014): 1657-1663.
Chicago
Kim, M. C., Lee, H. J., Lim, B., Ha, K., Kim, S. Y., So, I., Kim, B. J."Quercetin induces apoptosis by inhibiting MAPKs and TRPM7 channels in AGS cells". International Journal of Molecular Medicine 33, no. 6 (2014): 1657-1663. https://doi.org/10.3892/ijmm.2014.1704