Inhibition of glutamine utilization sensitizes lung cancer cells to apigenin-induced apoptosis resulting from metabolic and oxidative stress

  • Authors:
    • Yoon-Mi Lee
    • Gibok Lee
    • Taek-In Oh
    • Byeong Mo Kim
    • Do-Wan Shim
    • Kwang-Ho Lee
    • Young Jun Kim
    • Beong Ou Lim
    • Ji-Hong Lim
  • View Affiliations

  • Published online on: November 11, 2015     https://doi.org/10.3892/ijo.2015.3243
  • Pages: 399-408
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Recent studies have shown anticancer activity of apigenin by suppressing glucose transporter 1 (GLUT1) expression in cultured cancer cells; however, it is not clear whether apigenin can suppress glucose metabolism in lung cancer cells or sensitize them to inhibition of glutamine utilization-mediated apoptosis through metabolic and oxidative stress. We show that apigenin significantly decreases GLUT1 expression in mice. Furthermore, we demonstrate that apigenin induces growth retardation and apoptosis through metabolic and oxidative stress caused by suppression of glucose utilization in lung cancer cells. The underlying mechanisms were defined that the anticancer effects of apigenin were reversed by ectopic GLUT1 overexpression and galactose supplementation, through activation of pentose phosphate pathway-mediated NADPH generation. Importantly, we showed that severe metabolic stress using a glutaminase inhibitor, compound 968, was involved in the mechanism of sensitization by apigenin. Taken together, the combination of apigenin with inhibitors of glutamine metabolism may provide a promising therapeutic strategy for cancer treatment.

Introduction

The flavone apigenin (4′,5,7,-trihydroxyflavone), is a plant secondary metabolite that occurs widely in numerous fruits. It is a potent anticancer drug that can suppress cancer growth, angiogenesis, and metastasis in multiple types of cancer (1). Mechanistically, several studies have shown that apigenin suppresses the activation of nuclear transcription factor-κB (NF-κB), phosphoinositide 3-kinase (PI3K), epidermal growth factor receptor (EGFR), hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor (VEGF), cyclin D1, and vascular cell adhesion molecule 1 (VCAM-1), which play critical roles in the regulation of cell growth, cell cycle, angiogenesis, metastasis, and apoptosis (2). In addition, several studies have shown that apigenin causes accumulation of intracellular reactive oxygen species (ROS) and oxidative stress through the depletion of cellular glutathione (GSH), an important factor in redox balance (36). Nevertheless, the precise molecular mechanism by which decreased cellular GSH levels and increased levels of ROS relate to apigenin-mediated apoptosis is not fully understood.

Cancer cells use large amounts of glucose and glutamine to meet the increased energetic and anabolic demands associated with unlimited cell growth and survival (79). Increased glucose utilization in cancer cells is achieved primarily by upregulation of the expression of glucose transporter 1 (GLUT1), which is widely expressed in many tumors, including hepatic, pancreatic, breast, brain, lung, colorectal, and cervical cancers (10). Indeed, several reports have shown that GLUT1 inhibitors, such as fasentin, phloretin, and WZB117, suppress growth and induce cell death in cancer cells (1113). Upregulation of GLUT1 expression is closely associated with chemoresistance, tumor aggressiveness, and decreased survival among patients (14,15). Therefore, downregulation of GLUT1 expression could be a promising therapeutic strategy for cancer treatment.

The pentose phosphate pathway (PPP), which is required for biosynthesis of ribonucleotides and NADPH, can be initiated by glucose-6-phosphate generated by hexokinase as an initial key enzyme in glucose metabolism (16). As a cellular reducing power, NADPH can remove ROS through regulation of the GSH and thioredoxin (TRX) systems that directly scavenge ROS and repair ROS-induced cellular damage (7). Thus, inhibition of NADPH production has been proposed as a clinical intervention for cancer treatment. Indeed, preclinical studies have shown that inhibition of glucose-6-phosphate dehydrogenase (G6PD), an enzyme that initiates the PPP, is sufficient to decrease cell growth in leukemia, glioblastoma, and lung cancer cells (17).

Glutamine is a critical energy source required to maintain cell growth and survival. Growing cancer cells are commonly dependent upon a supply of glutamine to support the tricarboxylic acid (TCA) cycle. Mechanistically, glutamine can be converted to glutamate and α-ketoglutarate (α-KG) via the enzymes glutaminase (GLS) and glutamate dehydrogenase (GDH) (18). Previous studies have shown that inhibition of glutamine metabolism by GLS inhibitors, including 6-diazo-5-oxo-L-norleucine (DON), Bis-2-[5-phenylacetamido-1,2,4-thiadiazol-2-yl] ethyl sulfide (BPTES), and compound 968, or genetic targeting of GLS or GDH using small interfering RNA (siRNA), can suppress cell viability and increase apoptosis in cancer cells. This suggests that inhibition of glutamine utilization might cause cancer cells to become more sensitive to the effects of conventional anticancer drugs (1820). However, there is no available information addressing synergistic effects of apigenin on targeting cancer specific metabolism, such as glutamine metabolism.

In this study, we examined whether apigenin: i) inhibits GLUT1 expression and glucose utilization, thereby suppressing cell growth and inducing apoptosis; ii) downregulates NADPH and GSH production by inhibition of glucose metabolism, thereby resulting in increased oxidative stress; and iii) sensitizes lung cancer cells to inhibition of glutamine utilization, thereby causing apoptosis and growth retardation.

Materials and methods

Reagents and antibodies

N-acetyl-L-cysteine (NAC), apigenin, 2′-7′-dichlorodihydrofluorescein diacetate (DCF-DA) and compound 968 were purchased from Sigma-Aldrich (St. Louis, MO, USA) or Millipore (Billerica, MA, USA). Antibodies recognizing cleaved caspase-9, cleaved caspase-3, PARP, and HK1 (catalog nos. 9502, 9661, 9546, and 2024, respectively) were purchased from Cell Signaling Technology (Danvers, MA, USA). GLUT1 (sc-7903), GLS1 (156876) and β-tubulin (sc-9104) were purchased from Santa Cruz Biotechnology (Dallas, TX, USA) and Abcam (Cambridge, MA, USA).

Cell culture, plasmids, shRNA, and generation of stable cell lines

Lung cancer cells were cultured in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% fetal bovine serum and 25 mM glucose. The lentiviral vector pLenti-GIII-CMV- SLC2A1, expressing human GLUT1 was obtained from Applied Biological Materials Inc. (Richmond, BC, Canada). A short hairpin RNA construct targeting GLS1 was purchased from Sigma-Aldrich. The hairpin sequence was: 5′-CCGGG CCCTGAAGCAGTTCGAAATACTCGAGTATTTCGAAC TGCTTCAGGGCTTTTTG-3′. Lentiviral particles were produced in HEK293T cells, harvested 48 h after transfection, and then used to infect lung cancer cells. Infected lung cancer cells were selected by growth on media containing 2 μg/ml of puromycin (Invitrogen, Carlsbad, CA, USA) for 6 days.

Western blotting

Cells were lysed in buffer containing 1% IGEPAL, 150 mM NaCl, 50 mM Tris-HCl (pH 7.9), 10 mM NaF, 0.1 mM EDTA, and protease inhibitor cocktail. Protein samples were separated by SDS-PAGE, and the separated proteins were then transferred to a PVDF membrane (Millipore). Membranes were incubated in the presence of primary antibodies (1:1,000) at 4°C overnight, and HRP-conjugated secondary antibodies (1:10,000) were incubated for 1 h at room temperature. Proteins were visualized using an enhanced chemiluminescence (ECL) Prime kit (GE Healthcare, Pittsburgh, PA, USA).

ROS measurement

To measure ROS levels, cells were incubated with Hank's balanced salt solution (HBSS), containing 30 μM DCF-DA, for 30 min. After incubation, cells were detached and resuspended in phosphate-buffered saline (PBS) in preparation for fluorescence-activated cell sorting (FACS) analysis.

RNA isolation and RT-qPCR

Total RNA was extracted from cells using TRIzol (Invitrogen), and 2 μg of total RNA was used for cDNA synthesis using a high capacity cDNA reverse transcription kit (Applied Biosystems). Quantitative PCR was performed using SYBR Green PCR Master Mix (Applied Biosystems). The sequences of the PCR primers (5′-3′) were: GAGAGTTTCATCGGAGAGCC and CAGCGAGAATCGG CTACAG for HK1; GGCATTGATGACTCCAGTGTT and ATGGAGCCCAGCAGCAA for GLUT1; CTTGGGACAG CAGCCTTAAT and CAAGCTGGACGTTAAAGGGA for PGK1; AGCCCTTTCTCCATCTCCTT and AACCATGAC CAAGTGCAGAA for HK2; TACAGGCACAGTCGCAG AGT and CACTTCCTGGATGCTTGCTG for ALDOA; CCTGGCATGGATCTTGAGAA and TACGTTCACCT CGGTGTCTG for ENO1.

Glucose consumption, ATP, and lactate production assays

As previously described (9), lactate and glucose levels in cultured media were measured using lactate colorimetric assay kit and glucose colorimetric assay kit, respectively (BioVision, USA). Briefly, cells were cultured in DMEM without phenol red for 24 h, and then cultured media were mixed with assay solution. To measure intracellular ATP levels, cells were cultured in 6-well plates, and cell lysates were mixed with reaction buffer. ATP levels in the cell lysates were then determined using a luciferin-luciferase-based assay kit (Invitrogen), used according to the manufacturer's instructions. All values were normalized relative to cellular protein concentration.

Glutathione and NADPH assays

To prepare samples for determination of glutathione levels, cells were cultured in 6-well tissue culture dishes, and then cell lysates were obtained in extraction buffer without dithiothreitol (DTT) or β-mercaptoethanol. The levels of reduced glutathione (GSH) and oxidized glutathione (GSSG) were measured using a Glutathione Colorimetric Detection kit (BioVision) and an OxiSelect Glutathione Assay kit (Cell Biolabs), respectively, as previously described (21). The levels of NADPH in cultured cells were measured using a NADPH quantitation kit (BioVision). Briefly, cell lysates were extracted in NADPH extraction buffer, and then NADPH levels were determined by measurement of absorption at a wavelength of 450 nm.

Clonogenic cell survival assays

To measure clonogenic cell survival, 1×104 lung cancer cells were seeded in 6-well tissue culture dishes, and incubated for 7 days. After incubation, cells were fixed in 10% paraformaldehyde, and then stained with crystal violet for 10 min.

Apoptosis assays

A kit utilizing Annexin V-FITC and propidium iodide (BD Biosciences, San Jose, CA, USA) was used to quantify the numbers of apoptotic cells. Briefly, cells were detached from the culture dishes, washed with cold PBS, and then incubated with anti-Annexin V-FITC antibody and propidium iodide. Apoptotic cell numbers were determined by FACS analysis.

Statistical analysis

All data were analyzed using the unpaired Student's t-test for two experimental comparisons and one-way ANOVA with Tukey post-test for multiple comparisons. Data are represented as means ± standard deviations (SD). A p-value <0.05 was considered statistically significant.

Results

Apigenin reduces GLUT1 expression levels

Because it has been reported that apigenin exerts an anticancer effect through suppression of GLUT1 expression (2225), we examined whether apigenin also downregulates gene expression related to glycolysis, including GLUT1, HK1, PGK1, HK2, ALDOA, and ENO1. The results were consistent with those of previous reports (2225) that apigenin significantly decreased GLUT1 mRNA (Fig. 1A) and protein (Fig. 1B) levels, but did not decrease the levels of other glycolytic enzymes in H1299 or H460 lung carcinoma cells. In addition, GLUT1 mRNA levels were also decreased by apigenin treatment in other cancer cell lines, including A549 (lung carcinoma), H460 (lung carcinoma), H2030 (lung adenocarcinoma), HCT116 (colorectal carcinoma), SW480 (colorectal adenocarcinoma), and A375 (melanoma) (Fig. 1C). To elucidate whether apigenin suppresses GLUT1 expression in mice, we measured GLUT1 and GLUT4 expression levels in skeletal muscle, white adipose tissue, the heart, brain, liver, lung, and pancreas obtained from apigenin-treated mice. Fig. 1D shows that apigenin significantly suppressed GLUT1 but not GLUT4 expression in the brain, liver, lung, and pancreas. These results indicate that apigenin decreases GLUT1 mRNA and protein levels in various human cancer cell lines, and in mice. All experiments were performed according to the guidelines of the Animal Care and Use Committee, Konkuk University, Korea.

Apigenin causes cellular metabolic and oxidative stress

Cancer cells use large amounts of glucose to generate glycolytic intermediates and end products, including lactate, ATP, nucleotides, lipids, amino acids, and NADPH, all of which contribute to rapid cell proliferation (8). Therefore, we investigated whether apigenin can block glucose utilization in cancer cells. Interestingly, glucose consumption, lactate production, and ATP production were all strongly decreased by apigenin treatment (Fig. 2A). Glucose-6-phosphate, which is produced from glucose, can be shunted into the oxidative branch of the PPP by glucose-6-phosphate dehydrogenase, thereby generating NADPH and promoting redox homeostasis (26,27). Here, we demonstrated that NADPH generation by the oxidative branch of the PPP is decreased by apigenin treatment. Fig. 2B shows that, in apigenin-treated lung cancer cells, NADPH and GSH levels were significantly decreased, whereas GSSG levels were increased. Since NADPH maintains redox homeostasis through generation of GSH, which is necessary for the elimination of hydrogen peroxide (H2O2) by glutathione peroxidase (GPx) (27), we measured intracellular ROS levels. As we had hypothesized, intracellular ROS levels were strongly increased in apigenin-treated H1299 cells (Fig. 2C). Fig. 2D shows significantly decreased NADPH levels in the brain, liver, lung, and pancreatic tissues derived from apigenin-treated mice. These results suggest that apigenin induces oxidative stress through destruction of glucose utilization-mediated redox homeostasis.

Apigenin causes oxidative stress leading to apoptosis

Because apoptotic signal transduction cascades involving caspase-9, -3 and PARP cleavage can be activated by increased ROS levels (9,28), we examined whether apigenin could likewise increase apoptosis through this pathway. Fig. 3A shows that apoptotic cell numbers were increased by ~30% in apigenin-treated H1299, H460, and H2030 lung cancer cells. Furthermore, in H1299 and H460 cells, apigenin increased caspase-9, -3 and PARP cleavage in a dose-dependent manner (Fig. 3B). In addition, apigenin-induced apoptosis may be mediated by elevated ROS levels in H1299 and H460 cells, because treatment with the antioxidant, N-acetyl-L-cysteine (NAC), significantly rescued cell viability (Fig. 3C), and suppressed cell apoptosis (Fig. 3D). Taken together, these results show that accumulation of ROS mediates apigenin-induced apoptosis.

Galactose supplementation confers resistance to apigenin-induced apoptosis by re-activating antioxidant capacity

Because galactose supports NADPH production and cancer cell proliferation largely through its metabolism in the pentose phosphate pathway (26,29), we examined whether galactose supplementation could attenuate apigenin-mediated oxidative stress. Interestingly, the decreased levels of NADPH and GSH in apigenin-treated H1299 cells were strongly reversed in the presence of 20 mM galactose (Fig. 4A). Moreover, galactose decreased ROS levels by ~50% (Fig. 4B). Fig. 4C shows that galactose increased cell viability by ~40% in apigenin-treated H1299 and H460 cells. Consistent with these results, the numbers of apoptotic cells were also markedly decreased by ~20% in the presence of apigenin with galactose supplemented cells (Fig. 4D). Fig. 4E shows that apigenin-mediated activation of caspase-9, -3 and PARP apoptotic pathways (left panel) and decreased clonogenic cell survival (right panel) were completely blocked by 20 mM galactose in H1299 lung cancer cells. Together, these results indicate that galactose confers resistance to apigenin-mediated apoptosis by activating NADPH production through the pentose phosphate pathway.

Cancer cells expressing high levels of GLUT1 are resistant to apigenin-induced apoptosis through metabolic compensation of glucose utilization

Because apigenin induces cancer cell death by suppressing GLUT1 expression and glucose utilization in multiple types of cancer cell lines, we examined whether GLUT1-overexpressing cancer cells could confer resistance to apigenin-induced apoptosis. For this purpose, we generated H1299 cell stably expressing a GLUT1 lentiviral vector, or an empty vector (Fig. 5A). Fig. 5B shows that, even in the presence of apigenin, glucose consumption increased dramatically in GLUT1-overexpressing H1299 cells compared to cells harboring the empty vector. Furthermore, GLUT1 overexpression produced high levels of lactate in apigenin-treated H1299 cells compared to levels of lactate in cells carrying the empty vector. To determine whether GLUT1 overexpression could block apigenin-mediated suppression of NADPH and GSH production, we measured intracellular NADPH and GSSG levels in GLUT1-overexpressing H1299 cells in the presence or absence of apigenin. Interestingly, the effects of apigenin on NADPH and GSH levels were significantly rescued by GLUT1 overexpression (Fig. 5C). Furthermore, GLUT1 overexpression attenuated the effect of apigenin on ROS, resulting in a clear decrease in ROS levels of ~50% (Fig. 5D). Because increased oxidative stress is a critical step for apigenin-mediated apoptosis, we examined whether GLUT1-overexpressing cells were resistant to apigenin-mediated cell death. Fig. 5E shows that 50 μM apigenin was required to decrease cell viability in GLUT1-overexpressing cells, while 10 μM apigenin was sufficient to decrease cell viability in cells harboring the empty vector. Fig. 5F shows that 30 μM apigenin induced 20% of the cells to undergo apoptosis in GLUT1-overexpressing H1299 cells. In contrast, 30 μM apigenin induced 50% of cells harboring the empty vector to undergo apoptosis. Furthermore, GLUT1 overexpression dramatically reversed the apigenin-mediated suppression of clonogenic cell survival (Fig. 5G, left panel) and attenuated the activation of caspase-9 and caspase-3 in H1299 cells (Fig. 5G, right panel). These results suggest that GLUT1 overexpression causes resistance to apigenin-mediated apoptosis through suppression of metabolic and oxidative stress.

Figure 5

GLUT1 overexpression confers resistance to apigenin-induced apoptosis. (A) Ectopically expressed GLUT1 was detected by immunoblotting. (B) Glucose consumption and lactate production were measured in apigenin-treated H1299 lung cancer cells harboring either the empty vector or the GLUT1 expression vector. Values represent mean ± SD of three independent experiments performed in triplicate; *p<0.05, #p<0.05. (C) Intracellular NADPH and GSH levels were determined in apigenin-treated H1299 lung cancer cells harboring either the empty vector or the GLUT1 expression vector. Values represent mean ± SD of three independent experiments performed in duplicate; *p<0.05, #p<0.05. (D) Intracellular ROS levels. Values represent mean ± SD of three independent experiments performed in duplicate; **p<0.01, #p<0.05. (E) Cell viability was determined in apigenin-treated H1299 lung cancer cells harboring either the empty vector or the GLUT1 expression vector. Note the dose-dependent effects of apigenin treatment. Values represent mean ± SD of three independent experiments performed in duplicate; *p<0.05. (F) Apoptotic cell numbers were determined, after treatment at the indicated concentrations of apigenin, in H1299 lung cancer cells harboring either the empty vector or the GLUT1 expression vector. Values represent mean ± SD of three independent experiments performed in duplicate; *p<0.05. (G) Clonogenic cell survival was performed after treatment with 10 μM apigenin in H1299 cell cultures harboring either the empty vector or the GLUT1 expression vector. The cleaved caspase-9 and caspase-3 proteins were detected by immunoblotting.

Inhibition of glutamine utilization sensitizes apigenin-induced apoptosis through exacerbation of metabolic stress

In cancer cells, glutamine is a nutrient critical for promotion of tumor growth and survival (30,31). Thus, inhibitors targeting glutamine utilization have shown potential as anticancer drugs (1820). We therefore hypothesized that inhibition of glutamine utilization might sensitize cancer cells to apigenin-mediated cell death, and that the mechanism would involve suppression of the major pathways of glucose and glutamine metabolism required for tumor growth and survival. In support of this idea, Fig. 6A shows that compound 968, a GLS inhibitor, synergized with apigenin, resulting in an ~75% reduction of ATP levels in H1299 and H460 cells. Because glutamine utilization supports NADPH production needed for redox control (31), we additionally examined whether combination treatment of apigenin and compound 968 is sufficient to increase severe oxidative stress by decreasing NADPH and increasing intracellular ROS levels in cancer cells. Here, we found that compound 968 hugely decreased NADPH (Fig. 6B) and increased intracellular ROS levels (Fig. 6C) in the presence of apigenin. Furthermore, apigenin-treated cells, compound 968 exacerbated the reduction in cell viability (Fig. 6D), increased the induction of apoptosis (Fig. 6E, upper panel), and reduced clonogenic cell survival (Fig. 6E, bottom panel). In addition, we used a short hairpin RNA (shRNA) to knock down GLS1 expression by >70% in H1299 and H460 cell (Fig. 6F). Fig. 6G shows that apigenin caused a large decrease in cell viability of ~80% in GLS1 knocked-down H1299 and H460 cells. Consistent with this, caspase-9 and PARP were more strongly activated in these cells (Fig. 6H), suggesting that decreased GLS1 expression or activity would make cancer cells more susceptible to apigenin-mediated apoptosis. Taken together, these results indicate that inhibition of glutamine utilization should sensitize lung cancer cells to apigenin-mediated apoptosis.

Figure 6

Inhibition of glutamine metabolism sensitizes lung cancer cells to apigenin-induced apoptosis. (A) Intracellular ATP levels were determined in H1299 and H460 lung cancer cells treated with 10 μM apigenin in the absence or presence of 5 μM compound 968. Values represent mean ± SD of three independent experiments performed in duplicate; *p<0.05, **p<0.01. (B) NADPH levels were determined in H1299 and H460 lung cancer cells treated with 10 μM apigenin in the absence or presence of 5 μM compound 968. Values represent mean ± SD of three independent experiments performed in triplicate; *p<0.05, **p<0.01. (C) Intracellular ROS levels were measured in H1299 and H460 lung cancer cells treated with 10 μM apigenin in the absence or presence of 5 μM compound 968. Values represent mean ± SD of three independent experiments performed in duplicate; *p<0.05, **p<0.01. (D) Cell viability was determined in H1299 and H460 lung cancer cell cultures treated with 10 μM apigenin in the absence or presence of 5 μM compound 968. Values represent mean ± SD of three independent experiments performed in duplicate; *p<0.05, **p<0.01. (E) Apoptotic cell numbers were determined in H1299 and H460 lung cancer cell cultures treated with 10 μM apigenin in the absence or presence of 5 μM compound 968. Values represent mean ± SD of three independent experiments performed in duplicate; *p<0.05, **p<0.01. (F) GLS1 protein levels in H1299 and H460 cells were determined by immunoblotting. (G) Cell viability was determined in H1299 and H460 lung cancer cells expressing either GLS1 shRNA or scrambled shRNA in absence or presence of apigenin. Values represent mean ± SD of three independent experiments performed in triplicate; *p<0.05, **p<0.01. (H) Cleaved caspase-9 and PARP were detected by immunoblotting in H1299 and H460 lung cancer cells expressing GLS1 shRNA or scrambled shRNA in the absence or presence of apigenin (n=3). Protein levels were quantified by ImageJ.

Discussion

In recent years, apigenin, which is abundant in common fruits and vegetables, has shown considerable promise for development as an anticancer and chemopreventive agent (2,3,6). In fact, several recent reports have shown that apigenin is capable of selectively suppressing growth and inducing apoptosis in cancer cells by affecting the activities of protein tyrosine kinases, including epidermal growth factor receptor (EGFR), Src tyrosine kinase, phosphatidylinositol 3-kinase (PI3K), mitogen-activated protein kinase (MAPK), and hypoxia-inducible factor-1α (HIF-1Kα) (3236). It has been reported that apigenin suppresses glucose uptake by a mechanism involving decreased GLUT1 expression, leading to increased apoptosis and chemosensitivity in pancreatic carcinoma, laryngeal carcinoma, and head and neck cancer (2225). However, there is no previous evidence to indicate that a combination of apigenin with other inhibitors of cancer metabolism would show a synergistic effect in the suppression of cancer growth and survival. In this study, we observed that downregulation of GLUT1 by apigenin led to suppression of glucose utilization through decreased glycolysis and pentose phosphate metabolism, thereby suppressing the generation of ATP, macromolecules, and NADPH required for cancer cell growth, proliferation, and survival. Furthermore, our findings indicate that ectopic overexpression of GLUT1, combined with galactose supplementation to enhance PPP-mediated NADPH and biomass generation, confers resistance to apigenin-mediated apoptosis in several lung cancer cell lines. GLUT1 is upregulated in multiple types of cancer, and is closely correlated with cancer grade, radio-resistance, and chemo-resistance (10,37). Indeed, several GLUT1 inhibitors, including WZB117 and fasentin, have been developed as anticancer drugs (12,13). Thus, apigenin treatment might provide a selective anticancer strategy, acting through suppression of GLUT1-mediated cancer growth and survival.

Growing cancer cells use large amounts of glucose and glutamine to meet the bioenergetics and biosynthetic demands of increased cell growth and survival (7,8). It is becoming clear that altered metabolic pathways in cancer cells, such as aerobic glycolysis, fatty acid synthesis, and glutamine utilization are closely linked to therapeutic resistance in cancer treatment (7,8,26). Therefore, treatment of cancer using combinations of chemotherapeutic drugs and selective cancer metabolism inhibitors could represent a promising strategy to overcome chemoresistance. Indeed, several reports have shown that inhibition of glycolysis enhances the susceptibility of lung cancer, multiple myeloma, and breast cancer to apoptosis during treatment with anticancer drugs, such as cisplatin, doxorubicin, and trastuzumab (18). Previous reports have shown that GLS inhibitors, such as CB-839 and compound 968, can suppress cancer growth and survival in vitro and in vivo (20,38). However, these studies did not address whether the combined inhibition of glucose and glutamine utilization might represent a promising strategy for cancer treatment. In this study, we demonstrated that the combined suppression of glucose and glutamine utilization, using a combination of apigenin and compound 968, markedly decreased cell viability and increased apoptosis in lung cancer cells. Thus, our findings suggest that the simultaneous inhibition of multiple cancer metabolic pathways may provide a promising strategy for cancer treatment.

Mechanistically, increased ROS is involved in apigenin-induced apoptosis in prostate and colorectal cancer cells (3,6). However, the molecular mechanism by which apigenin increases intracellular ROS levels is not well understood. Because suppression of glucose utilization is reported to induce oxidative stress through inhibition of the PPP and NADPH generation (27), we hypothesized that apigenin might induce oxidative stress by the same mechanism. Indeed, we found that apigenin significantly decreased glucose utilization through suppression of GLUT1 expression, and consequently decreased NADPH production, which led to increased ROS levels. In addition, activation of NADPH production by galactose supplementation, which can be entered into PPP, significantly reversed the apigenin-induced ROS accumulation. These results therefore suggest a possible mechanism by which apigenin induces oxidative stress through inhibition of glucose utilization and PPP-mediated NADPH production.

In conclusion, the major findings are that: i) apigenin decreases glucose utilization, via glycolysis and the PPP, through suppression of GLUT1 expression, thereby resulting in the inhibition of cancer cell growth and survival; ii) activation of glucose utilization and the PPP by GLUT1 overexpression and galactose supplementation confer resistance to apigenin-induced apoptosis by compensating NADPH production, which suppresses ROS accumulation; iii) targeting glutamine utilization sensitizes lung cancer cells to apigenin-induced apoptosis under conditions of severe metabolic stress. These results may provide a promising therapeutic strategy for cancer treatment.

Acknowledgements

This study was supported by the faculty research fund of Konkuk University in 2013.

Abbreviations:

PPP

pentose phosphate pathway

GLUT1

glucose transporter 1

ROS

reactive oxygen species

GLS

glutaminase

NAC

N-acetyl-L-cysteine

DCF-DA

2′-7′-dichrolodihydrofluorescein

References

1 

Li-Weber M: Targeting apoptosis pathways in cancer by Chinese medicine. Cancer Lett. 332:304–312. 2013. View Article : Google Scholar

2 

Gupta SC, Kim JH, Prasad S and Aggarwal BB: Regulation of survival, proliferation, invasion, angiogenesis, and metastasis of tumor cells through modulation of inflammatory pathways by nutraceuticals. Cancer Metastasis Rev. 29:405–434. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Banerjee K and Mandal M: Oxidative stress triggered by naturally occurring flavone apigenin results in senescence and chemotherapeutic effect in human colorectal cancer cells. Redox Biol. 5:153–162. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Chan LP, Chou TH, Ding HY, Chen PR, Chiang FY, Kuo PL and Liang CH: Apigenin induces apoptosis via tumor necrosis factor receptor- and Bcl-2-mediated pathway and enhances susceptibility of head and neck squamous cell carcinoma to 5-fluorouracil and cisplatin. Biochim Biophys Acta. 1820:1081–1091. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Kachadourian R and Day BJ: Flavonoid-induced glutathione depletion: Potential implications for cancer treatment. Free Radic Biol Med. 41:65–76. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Shukla S and Gupta S: Apigenin-induced prostate cancer cell death is initiated by reactive oxygen species and p53 activation. Free Radic Biol Med. 44:1833–1845. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Cairns RA, Harris IS and Mak TW: Regulation of cancer cell metabolism. Nat Rev Cancer. 11:85–95. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Vander Heiden MG: Targeting cancer metabolism: A therapeutic window opens. Nat Rev Drug Discov. 10:671–684. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Vazquez F, Lim JH, Chim H, Bhalla K, Girnun G, Pierce K, Clish CB, Granter SR, Widlund HR, Spiegelman BM, et al: PGC1α expression defines a subset of human melanoma tumors with increased mitochondrial capacity and resistance to oxidative stress. Cancer Cell. 23:287–301. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Szablewski L: Expression of glucose transporters in cancers. Biochim Biophys Acta. 1835:164–169. 2013.

11 

Kim MS, Kwon JY, Kang NJ, Lee KW and Lee HJ: Phloretin induces apoptosis in H-Ras MCF10A human breast tumor cells through the activation of p53 via JNK and p38 mitogen-activated protein kinase signaling. Ann NY Acad Sci. 1171:479–483. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Liu Y, Cao Y, Zhang W, Bergmeier S, Qian Y, Akbar H, Colvin R, Ding J, Tong L, Wu S, et al: A small-molecule inhibitor of glucose transporter 1 downregulates glycolysis, induces cell-cycle arrest, and inhibits cancer cell growth in vitro and in vivo. Mol Cancer Ther. 11:1672–1682. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Wood TE, Dalili S, Simpson CD, Hurren R, Mao X, Saiz FS, Gronda M, Eberhard Y, Minden MD, Bilan PJ, et al: A novel inhibitor of glucose uptake sensitizes cells to FAS-induced cell death. Mol Cancer Ther. 7:3546–3555. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Hatanaka M: Transport of sugars in tumor cell membranes. Biochim Biophys Acta. 355:77–104. 1974.PubMed/NCBI

15 

Ulanovskaya OA, Cui J, Kron SJ and Kozmin SA: A pairwise chemical genetic screen identifies new inhibitors of glucose transport. Chem Biol. 18:222–230. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Patra KC and Hay N: The pentose phosphate pathway and cancer. Trends Biochem Sci. 39:347–354. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Budihardjo II, Walker DL, Svingen PA, Buckwalter CA, Desnoyers S, Eckdahl S, Shah GM, Poirier GG, Reid JM, Ames MM, et al: 6-Aminonicotinamide sensitizes human tumor cell lines to cisplatin. Clin Cancer Res. 4:117–130. 1998.PubMed/NCBI

18 

Zhao Y, Butler EB and Tan M: Targeting cellular metabolism to improve cancer therapeutics. Cell Death Dis. 4:e5322013. View Article : Google Scholar : PubMed/NCBI

19 

Seltzer MJ, Bennett BD, Joshi AD, Gao P, Thomas AG, Ferraris DV, Tsukamoto T, Rojas CJ, Slusher BS, Rabinowitz JD, et al: Inhibition of glutaminase preferentially slows growth of glioma cells with mutant IDH1. Cancer Res. 70:8981–8987. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Wang JB, Erickson JW, Fuji R, Ramachandran S, Gao P, Dinavahi R, Wilson KF, Ambrosio AL, Dias SM, Dang CV, et al: Targeting mitochondrial glutaminase activity inhibits oncogenic transformation. Cancer Cell. 18:207–219. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Lim JH, Luo C, Vazquez F and Puigserver P: Targeting mitochondrial oxidative metabolism in melanoma causes metabolic compensation through glucose and glutamine utilization. Cancer Res. 74:3535–3545. 2014. View Article : Google Scholar : PubMed/NCBI

22 

Bao YY, Zhou SH, Fan J and Wang QY: Anticancer mechanism of apigenin and the implications of GLUT-1 expression in head and neck cancers. Future Oncol. 9:1353–1364. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Bao YY, Zhou SH, Lu ZJ, Fan J and Huang YP: Inhibiting GLUT-1 expression and PI3K/Akt signaling using apigenin improves the radiosensitivity of laryngeal carcinoma in vivo. Oncol Rep. 34:1805–1814. 2015.PubMed/NCBI

24 

Melstrom LG, Salabat MR, Ding XZ, Milam BM, Strouch M, Pelling JC and Bentrem DJ: Apigenin inhibits the GLUT-1 glucose transporter and the phosphoinositide 3-kinase/Akt pathway in human pancreatic cancer cells. Pancreas. 37:426–431. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Xu YY, Wu TT, Zhou SH, Bao YY, Wang QY, Fan J and Huang YP: Apigenin suppresses GLUT-1 and p-AKT expression to enhance the chemosensitivity to cisplatin of laryngeal carcinoma Hep-2 cells: An in vitro study. Int J Clin Exp Pathol. 7:3938–3947. 2014.PubMed/NCBI

26 

Hamanaka RB and Chandel NS: Targeting glucose metabolism for cancer therapy. J Exp Med. 209:211–215. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Jeon SM, Chandel NS and Hay N: AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress. Nature. 485:661–665. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Lim JH: Zinc finger and BTB domain-containing protein 3 is essential for the growth of cancer cells. BMB Rep. 47:405–410. 2014. View Article : Google Scholar : PubMed/NCBI

29 

Weinberg F, Hamanaka R, Wheaton WW, Weinberg S, Joseph J, Lopez M, Kalyanaraman B, Mutlu GM, Budinger GR and Chandel NS: Mitochondrial metabolism and ROS generation are essential for Kras-mediated tumorigenicity. Proc Natl Acad Sci USA. 107:8788–8793. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Le A, Lane AN, Hamaker M, Bose S, Gouw A, Barbi J, Tsukamoto T, Rojas CJ, Slusher BS, Zhang H, et al: Glucose-independent glutamine metabolism via TCA cycling for proliferation and survival in B cells. Cell Metab. 15:110–121. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Wise DR and Thompson CB: Glutamine addiction: A new therapeutic target in cancer. Trends Biochem Sci. 35:427–433. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Agullo G, Gamet-Payrastre L, Manenti S, Viala C, Rémésy C, Chap H and Payrastre B: Relationship between flavonoid structure and inhibition of phosphatidylinositol 3-kinase: A comparison with tyrosine kinase and protein kinase C inhibition. Biochem Pharmacol. 53:1649–1657. 1997. View Article : Google Scholar : PubMed/NCBI

33 

Fang J, Xia C, Cao Z, Zheng JZ, Reed E and Jiang BH: Apigenin inhibits VEGF and HIF-1 expression via PI3K/AKT/p70S6K1 and HDM2/p53 pathways. FASEB J. 19:342–353. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Huang YT, Kuo ML, Liu JY, Huang SY and Lin JK: Inhibitions of protein kinase C and proto-oncogene expressions in NIH 3T3 cells by apigenin. Eur J Cancer. 32A:146–151. 1996. View Article : Google Scholar : PubMed/NCBI

35 

Llorens F, Miró FA, Casañas A, Roher N, Garcia L, Plana M, Gómez N and Itarte E: Unbalanced activation of ERK1/2 and MEK1/2 in apigenin-induced HeLa cell death. Exp Cell Res. 299:15–26. 2004. View Article : Google Scholar : PubMed/NCBI

36 

Yin F, Giuliano AE, Law RE and Van Herle AJ: Apigenin inhibits growth and induces G2/M arrest by modulating cyclin-CDK regulators and ERK MAP kinase activation in breast carcinoma cells. Anticancer Res. 21(1A): 413–420. 2001.PubMed/NCBI

37 

Adekola K, Rosen ST and Shanmugam M: Glucose transporters in cancer metabolism. Curr Opin Oncol. 24:650–654. 2012. View Article : Google Scholar : PubMed/NCBI

38 

Gross MI, Demo SD, Dennison JB, Chen L, Chernov-Rogan T, Goyal B, Janes JR, Laidig GJ, Lewis ER, Li J, et al: Antitumor activity of the glutaminase inhibitor CB-839 in triple-negative breast cancer. Mol Cancer Ther. 13:890–901. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 48 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lee Y, Lee G, Oh T, Kim BM, Shim D, Lee K, Kim YJ, Lim BO and Lim J: Inhibition of glutamine utilization sensitizes lung cancer cells to apigenin-induced apoptosis resulting from metabolic and oxidative stress. Int J Oncol 48: 399-408, 2016
APA
Lee, Y., Lee, G., Oh, T., Kim, B.M., Shim, D., Lee, K. ... Lim, J. (2016). Inhibition of glutamine utilization sensitizes lung cancer cells to apigenin-induced apoptosis resulting from metabolic and oxidative stress. International Journal of Oncology, 48, 399-408. https://doi.org/10.3892/ijo.2015.3243
MLA
Lee, Y., Lee, G., Oh, T., Kim, B. M., Shim, D., Lee, K., Kim, Y. J., Lim, B. O., Lim, J."Inhibition of glutamine utilization sensitizes lung cancer cells to apigenin-induced apoptosis resulting from metabolic and oxidative stress". International Journal of Oncology 48.1 (2016): 399-408.
Chicago
Lee, Y., Lee, G., Oh, T., Kim, B. M., Shim, D., Lee, K., Kim, Y. J., Lim, B. O., Lim, J."Inhibition of glutamine utilization sensitizes lung cancer cells to apigenin-induced apoptosis resulting from metabolic and oxidative stress". International Journal of Oncology 48, no. 1 (2016): 399-408. https://doi.org/10.3892/ijo.2015.3243