The role of quercetin and vitamin C in Nrf2‑dependent oxidative stress production in breast cancer cells

  • Authors:
    • Zohreh Mostafavi‑Pour
    • Fatemeh Ramezani
    • Fatemeh Keshavarzi
    • Nasser Samadi
  • View Affiliations

  • Published online on: January 18, 2017     https://doi.org/10.3892/ol.2017.5619
  • Pages: 1965-1973
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The balance between the production and elimination of reactive oxygen species (ROS) is essential in determining whether cells survive or undergo apoptosis. Nuclear factor erythroid 2‑related factor 2 (Nrf2) may act as a sensor for electrophilic stress, thus regulating the intracellular antioxidant response. The present study investigated the role of vitamin C (VC) and quercetin (Q) in the induction of Nrf2‑mediated oxidative stress in cancer cells. An MTT assay was conducted to examine the anti‑proliferative effects of VC and Q. Reverse transcription‑quantitative polymerase chain reaction and western blot analysis were performed to determine the messenger RNA (mRNA) and protein expression of Nrf2, respectively. The activity of nicotinamide adenine dinucleotide phosphate dehydrogenase quinone 1, heme oxygenase 1, glutathione peroxidase, glutathione reductase and reduced glutathione were measured by spectrophotometric analysis. Intracellular generation of ROS was determined using 2'‑7'‑dichlorodihydrofluorescein diacetate fluorescent probes. The results demonstrated that the cytotoxicity (50% inhibitory concentration) of VC and Q were 271.6‑480.1 and 155.1‑232.9 µM, respectively. Additionally, there was a significant decrease in the expression of Nrf2 mRNA and protein levels following the treatment of breast cancer cells with VC and Q (P=0.024). Following treatment with VC and Q, the nuclear/cytosolic Nrf2 ratio was reduced by 1.7‑fold in MDA‑MB 231 cells, 2‑fold in MDA‑MB 468 cells, 1.4‑fold in MCF‑7 cells and 1.2 fold in A549 cells. Sequential treatment with VC and Q decreased endogenous production of ROS in a dose‑dependent manner (P=0.027). The results of the current study suggest that VC and Q treatment may be developed as an adjuvant for patients with cancer and overexpression of Nrf2.

Introduction

Phytochemicals have been applied as a multi-targeting approach to cancer medicine due to their potential to improve the efficiency of chemotherapy protocols (1). Tumor cells produce high levels of antioxidants that neutralize free radicals, thus creating a negative balance of intracellular reactive oxygen species (ROS) levels, which facilitates the survival of cancer cells. Antioxidants help to alleviate the toxic effects of free radical-producing drugs and preserve the health of normal tissues patients with cancer (2). However, it has been demonstrated that the up regulation of antioxidant systems may provide the same protection to tumor cells against oxidative damage, and subsequently, may stimulate tumor progression by increasing the aggressiveness and chemoresistance of tumor cells (2).

Nuclear factor erythroid 2-related factor 2 (Nrf2) has been recognized as a member of the cap ‘n’ collar subfamily, and regulates the intracellular antioxidant response through the controlled activation of a series of genes, including phase-II detoxifying enzymes, endogenous antioxidants and transporters that shield cells from the harmful effects of carcinogens and environmental toxins (35). Overexpression of Nrf2 and its downstream target genes has been identified in numerous primary tumors, and may protect cancer cells against the cytotoxic effects of chemotherapeutic agents (6,7). Therefore, understanding the signaling pathway of Nrf2 is essential in tumor biology, and the application of Nrf2 inhibitors may be a useful method of treating tumors (811).

Vitamin C (VC) is known as one of the most prominent antioxidative components, which may exert chemopreventive effects without perceptible toxic side effects (12). VC produces cytotoxic levels of hydrogen peroxide and kills cancer cells at pharmacological concentrations, as tumor cells are often catalase-deficient and more susceptible to hydrogen peroxide than normal cells (12). It has been demonstrated that ascorbic acid (a form of VC) protects normal cells against oxidative stress in mice, suggesting that VC may be used as an adjuvant for cancer treatment (1315). Additionally, ascorbic acid inhibits Nrf2 activation by interfering with Nrf2 nuclear localization and it's binding to the antioxidant response element (ARE) sequence (16).

One of the most abundant flavonoids in fruits and vegetables is quercetin (Q), which has been shown to exert anticancer actions, such as the blocking of tumor initiation (17), in addition to exerting anti-oxidative (18) and anti-apoptotic activities in different cancer cell lines (19,20). Notably, Q has the capacity to act either as an antioxidant or as a pro-oxidant, depending on its concentration and the period of exposure (21,22). It has been demonstrated that high doses of Q decrease cell survival rates and diminish the levels and activities of cellular antioxidants, thus enhancing antitumor effects (23). By contrast, low doses of Q augment the total antioxidant capacity of cancer cells and counteract the cytotoxic effects of antineoplastic drugs in lung cancer A549, colorectal cancer HCT116 and ovarian cancer cells (22). Q rapidly stimulates Nrf2 phosphorylation and translocation to the cytosol (24). However, long-term treatment of the cells with Q inhibits both these effects and transiently induces the activation of p38 mitogen-activated protein kinases (24).

In the present study, it was hypothesized that the combined effects of Q (a multiple signaling inhibitor) and VC (an antioxidant agent with antineoplastic activity) could exert a synergistic effect on ROS levels in cancer cells via inhibition of Nrf2. The cytotoxicity of Q and VC in various cancer cells was examined, and the effectiveness of the Nrf2 pathway was investigated at the gene and protein levels.

Materials and methods

Reagents

RPMI-1640 medium and 10% fetal bovine serum (FBS) were obtained from Invitrogen (Thermo Fisher Scientific, Inc., Waltham, MA, USA). Primary rabbit polyclonal anti-Nrf2 (sc-722) and anti-β-actin antibodies (sc-47778) were purchased from Santa Cruz Biotechnology, Inc. (Dallas, TX, USA). Q and VC were purchased from Sigma-Aldrich (Merck Millipore, Darmstadt, Germany). The human breast cancer cell lines MDA-MB 231, MDA-MB 468 and MCF-7, and the human lung cancer cell line A549, were all obtained from the National Cell Bank of Iran, Pasteur Institute of Iran (Tehran, Iran).

Cell cytotoxicity study

Cancer cells were seeded at a density of 1×104 cells/well in a 96-well plate and cultured in RPMI-1640 medium containing 10% FBS, 100 U/ml penicillin and 100 µg/ml streptomycin at 37°C in a humidified 5% CO2 atmosphere. Cells at passages 3–5 were used in subsequent experiments after reaching 70% confluence. Cells were exposed to varying concentrations of Q and VC (0.1–1,000 µM) for 24 h. Subsequently, 100 µl 0.5 mg/ml MTT solution in PBS was added per well, and the plate was incubated at 37°C for 3 h in the dark. The absorbance was then measured at 570 nm in an ELISA reader (Mikura Ltd., Horsham, UK).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Cancer cells were cultured at a density of 5×105 cells/well in a 6-well plate, and following incubation for 18–24 h, cells were treated with 200 or 100 µM VC for 24 h. Then, 50 or 75 µM Q, respectively, was added for 6 h. Following isolation of RNA using BioZOL RNA extraction reagent (BioFlux Corporation, Tokyo, Japan), the amount of RNA was determined with a NanoDrop 1000 spectrophotometer (NanoDrop Technologies; Thermo Fisher Scientific, Inc., Wilmington, DE, USA). Total RNA was immediately reverse transcribed to generate first-strand complementary DNA using an RT kit (Fermentas; Thermo Fisher Scientific, Inc., Pittsburgh, PA, USA), according to the manufacturer's protocol. Specific primers for Nrf2 were used to detect Nrf2 expression: Forward, 5′-ACACGGTCCACAGCTCATC-3′ and reverse, 5′-TGTCAATCAAATCCATGTCCTG-3′. The levels of β-actin and ribosomal protein lateral stalk subunit P0 (RPLP0) were also analyzed as reference genes. The primers used were as follows: β-actin forward, 5′-AATCGTGCGTGACATTAAG-3′ and reverse, 5′-GAAGGAAGGCTGGAAGAG-3′; and RPLPO forward, 5′-GAAGGCTGTGGTGCTGATGG-3′ and reverse, 5′-CCGGATATGAGGCAGCAGTT-3′. qPCR was performed using SYBR® Premix Ex Taq™ II (Tli RNaseH Plus) (Takara Bio, Inc., Otsu, Japan) and analyzed using the software provided in the StepOnePlus™ Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.). qPCR amplification was carried out for 25 cycles using the following protocol: 95°C for 10 min, 94°C for 30 sec, 55°C for 30 sec, 72°C for 30 sec and 72°C for 10 min. The Pfaffl method was used for the relative mRNA quantification, as described previously (25,26).

Western blot analysis

To detect Nrf2 protein levels, cells at a density of 12×105 cells/T75 flask were cultured for 18–24 h. Firstly, they were exposed to 200 or 100 µM VC for 24 h. Subsequently, 50 or 75 µM Q (respectively) was added for 6 h. Cells were then lysed at 4°C in a buffer containing 50 mM Tris, 20 mM NaCl and 200 µl NP-40 in a final volume of 20 ml (pH 8.0). A total of 10 µl 7X protease inhibitor cocktail (P8340; Sigma-Aldrich; Merck Millipore) was mixed with 750 µl lysis buffer, and then 1X lysis buffer was added to each flask. Cells were removed by a scrapper and placed on a rotator for 30 min, followed by centrifugation at 12,000 × g for 20 min at 4°C. The supernatant was then collected, and protein concentrations were determined using the Pierce BCA Protein Assay kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA). To produce a cytosolic fraction, cells were re-suspended at 4°C in 10 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (1.5 mM MgCl2, 10 mM KCl, 0.5 mM dithiothreitol and 0.2 mM phenylmethane sulfonyl fluoride; pH 7.9), placed on ice for 10 min and vortexed for 10 sec. Samples were centrifuged at 10,000 × g for 2 min at 4°C, and the supernatant containing the cytosolic fraction was stored at −80°C. Protein concentrations were measured using the aforementioned Pierce BCA Protein Assay kit. Equal amounts of protein (30 µg per sample) were separated by 12.5% SDS-PAGE and transferred to a nitrocellulose membrane. Following blocking with 10% skimmed milk for 1 h, proteins were incubated with rabbit polyclonal antibodies against Nrf2 (dilution, 1:700) and β-actin (dilution, 1:5,000) at 4°C overnight. Upon washing three times, the membranes were further incubated with horseradish peroxidase-conjugated secondary antibodies (rabbit anti-mouse immunoglobulin G; dilution, 1:10,000; ab97046; Abcam, Cambridge, UK) for 2 h at room temperature. Finally, immunoreactive protein bands were developed using enhanced chemiluminscence (123072; Sigma-Aldrich; Merck Millipore). Normalization of western blot analysis was ensured by using β-actin as a loading control. Western blot quantification was performed using ImageJ software version 1.48 (https://imagej.nih.gov/ij/download.html).

Determination of glutathione peroxidase (GPx) activity

The procedures defined by Fecondo and Augusteyn (27), which screen continuous regeneration of reduced glutathione (GSH) from oxidized glutathione (GSSG) in the presence of glutathione reductase (GR; Sigma-Aldrich; Merck Millipore) and disodium (Na2) salt of reduced nicotinamide adenine dinucleotide phosphate (NADPH; Sigma-Aldrich; Merck Millipore) were used to determine the GPx activity, with minor modifications. After culturing cells at a density of 12×105 in a T75 flask for 18–24 h, cancer cells were treated with VC and Q as mentioned in the preceding paragraph. The enzyme activity in the clear supernatant of tumor cell lysates was expressed as µmol of NADPH oxidized/min/mg cell protein, using a molar extinction coefficient of 6.22×106 M−1 cm−1 for NADPH. The GPx activity is defined as mU/mg of cell protein.

Determination of GR activity

The activity of GR was assessed by the method elucidated by Maiani et al (28), with minor modifications. Cancer cells were cultured at a density of 12×105 cells/T75 flask, and after 18–24 h of incubation, they were treated with 200 or 100 µM VC for 24 h. Then, 50 or 75 µM Q, respectively, was added for 6 h. The GR assay was performed in a cuvette in a total volume of 1 ml, containing 60 µM buffer, 5 mM EDTA (pH 8.0), 0.033 M GSSG, 2 mM NADPH and sample. The decrease in absorbance, which represents the oxidation of NADPH during the reduction of GSSG by the GR present in the sample, was monitored spectrophotometrically at 340 nm for 3 min. Results were based on a molar extinction coefficient for NADPH of 6.22×106 M−1 cm−1. The GR activity was defined as mU/mg cell protein.

Determination of NADPH dehydrogenase quinone 1 (NQO1) activity

Similarly, to the determination of GR activity, following the seeding of cancer cells at a density of 12×105 cells/T75 flask and treatment with VC and Q, cells were washed with FBS and resuspended in 2 ml 25 mM Tris-HCl buffer (pH 7.4) and 250 mM sucrose (1:1). Then, cells were sonicated on ice for 10 sec (twice) using a probe sonicator. The resultant sonicate was centrifuged at 10,000 × g at 4°C for 30 sec to remove large particles. The activity of NQO1 was determined spectrophotometrically as the dicoumarol-inhibitable fraction of the NADH-dependent reduction of dichloroindophenol (DCPIP). DCPIP was used as an electron acceptor, as it loses color upon reduction. Briefly, 100 ml extract was placed in an acid-cleaned quartz cuvette containing 2.7 ml buffer [25 mM Tris-HCl (pH 7.4), 700 mg/ml bovine serum albumin (BSA) (A1933; Sigma-Aldrich; Merck Millipore)], 100 ml NADH (6 mM) and 100 ml DCPIP (1.2 mM). The cuvette was rapidly agitated, and the absorbance at 600 nm was recorded over 2 min using a sample with no enzyme as a reference. The assay was then repeated with a fresh sample containing 10 ml dicoumarol inhibitor (10 mM in dimethyl sulfoxide). Dicoumarol sensitive activity [rate of optical density (OD) change without inhibitor/rate of OD change with inhibitor] was used to measure NQO1 activity. The final activities were calibrated against protein concentration and expressed as nM/min/mg protein. Protein concentration was determined using the Pierce BCA Protein Assay kit (Thermo Fisher Scientific, Inc.).

Heme oxygenase 1 (HO1) activity assay

HO1 activity was measured in microsomal preparations from cells. Similar to the previous test, following cell culture at a density of 12×105 cells/T75 flask and treatment with VC and Q for 30 h, cells were homogenized in 0.5 ml ice-cold 0.25 M sucrose solution containing 50 mM potassium phosphate buffer (pH 7.4). Homogenates were centrifuged at 200 × g for 10 min. The supernatants were then centrifuged at 10,000 × g for 20 min, and further centrifuged at 30,000 × g for 60 min at 4°C. The resultant pellet was resuspended in 50 mM potassium phosphate buffer (pH 7.4) and the protein concentration was determined using the aforementioned Pierce BCA Protein Assay kit. The extract (containing 40 mM protein) was mixed with 20 µM hemin (H2250; Sigma-Aldrich; Merck Millipore), 15 mM BSA, 1 mM NADPH, 0.1 M potassium phosphate buffer (pH 7.4) and 1.5 unit purified biliverdin reductase (B3687; Sigma-Aldrich; Merck Millipore). After 1 h of incubation at 37°C, the reaction was stopped with 0.6 ml chloroform. Following the extraction of cells, bilirubin concentrations in the chloroform cell extracts were determined by spectrophotometry at an absorbance wavelength of 464–530 nm. HO1 activity was calculated as nM bilirubin/mg protein/min, assuming an extinction coefficient of 40/(mmol/l)/cm in chloroform.

Determination of GSH

GSH assay using 5,5-dithio-bis-(2-nitrobenzoic acid) (DTNB) was performed according to the Ellman's method (29). Standard curves were constructed from 1 mM GSH. Following the seeding of cancer cells at a density of 12×105 cells/T75 flask and treatment with VC and Q, clear supernatant of cell lysate was analyzed for GSH levels. A total of 2.3 ml potassium phosphate buffer (0.2 M, pH 7.6) was added to 0.2 ml cell lysate supernatant, and then 0.5 ml DTNB (0.001 M) was added to the solution. The absorbance was measured 5 min later at 412 nm.

Determination of intracellular generation of ROS

2′-7′-Dichlorodihydrofluorescein diacetate (DCFH-DA) fluorescent probes were used to measure the intracellular generation of hydrogen peroxide (H2O2) and superoxide anions (O2˙), respectively (30). These probes are stable nonpolar compounds that readily diffuse into cells. Once inside the cells, the acetate groups of DCFH-DA are cleaved from the molecule by intracellular esterases to yield DCFH, which is trapped within the cells. Intracellular H2O2 or low-molecular weight peroxides, oxidize DCFH to dichloride, which is a highly fluorescent compound. Thus, the fluorescence intensity is proportional to the quantity of peroxide produced by the cells. Briefly, solid tumor cells were seeded at a density of 1×104 cells/well in a 96-well plate. Following cell treatments with VC and Q, the media of each well were removed, and 100 µl 10 µM DCFH-DA was added to the plate, which was then incubated for 30 min at 37°C in a humidified 5% CO2 atmosphere. Extracellular DCFH-DA was subsequently replaced with 200 µl PBS (PBS without calcium or magnesium), and the fluorescence intensity was determined with a fluorimeter, using 480 and 530 nm as the excitation and emission wavelengths, respectively.

Statistical analysis

Data were collected and expressed as the mean ± standard error of the mean from three independent experiments. Statistical analysis was performed by applying one-way analysis of variance and Tukey's test to compare the control and vehicle groups against the treated groups. P<0.05 was considered to indicate a statistically significant difference. The 50% inhibitory concentration (IC50) values for VC and Q were calculated using GraphPad Prism software version 5 (GraphPad Software, Inc., La Jolla, CA, USA).

Results

Evaluating the cytotoxic effects of VC and Q against solid tumor cell lines

VC exhibited cytotoxicity against the cancer cell lines, with an IC50 of 271.6–480.1 µM. Q displayed comparable cytotoxic profiles against the tumor cell lines, with an IC50 of 155.1–232.9 µM (Table I). VC and Q exhibited a growth-inhibitory effect in a dose-dependent manner. Dose-response values were measured (Fig. 1A and D). The results indicated a concentration-dependent decrease in cell viability following treatment with VC and Q. VC and Q significantly decreased cell viability of tumor cells at concentrations >100 µM (Fig. 1) (P=0.045). Based on this observation, subtoxic concentrations of Q (50 and 75 µM) and VC (100 and 200 µM) were selected to investigate the effect of Q and VC on Nrf2 signaling.

Table I.

IC50 values of vitamin C and quercetin in different tumor cell lines. Data are expressed as the mean ± standard deviation of three independent experiments (n=3).

Table I.

IC50 values of vitamin C and quercetin in different tumor cell lines. Data are expressed as the mean ± standard deviation of three independent experiments (n=3).

A549MCF-7MDA-MB 468MDA-MB 231 IC50
232.90±17.75155.10±33.80183.20±22.50196.70±40.90Quercetin (µM)
480.10±25.05271.60±31.40365.90±24.95382.10±8.69Vitamin C (µM)

[i] IC50, 50% inhibitory concentration.

Roles of VC and Q on Nrf2 expression

In a preliminary study, MDA-MB 231 cells were seeded and cultured for 24 h prior to treatment with various concentrations of VC (50, 100 and 200 µM) or Q (25, 50 and 75 µM), individually or in combination. With sequential treatment, the most significant decrease in Nrf2 mRNA expression was observed following the incubation of cells with 200 µM VC in combination with 50 µM Q, or 100 µM VC with 75 µM Q (data not shown). Therefore, the same combinations of VC and Q were applied for subsequent treatments of the other cancer cell lines. The results indicated that incubating breast cancer cells with VC and Q induced a marked decrease in the mRNA and protein expression of Nrf2 in a dose-dependent manner (Figs. 2 and 3). Despite an aggressive genotype, MDA-MB 231 cells exhibited a greater reduction in the mRNA and nuclear fraction of Nrf2 than other cells, suggesting that MDA-MB 231 cells with higher Nrf2 levels are more sensitive to the suppressive effect of VC and Q than cells with lower levels of Nrf2 (P=0.024 for all cell lines). Treatment with VC and Q decreased nuclear Nrf2 levels (Fig. 2B) and the nuclear/cytosolic Nrf2 ratio (Fig. 3C) in all cell lines. The nuclear/cytosolic Nrf2 ratio decreased by 1.7-fold in MDA-MB 231 cells, 2-fold in MDA-MB 468 cells, 1.4-fold in MCF-7 cells and 1.2-fold in A549 cells following treatment of tumor cells with 200 µM VC and 50 µM Q. This ratio was much lower in cells treated with 100 µM VC and 75 µM Q: 3.4-fold in MDA-MB 231 cells, 6-fold in MDA-MB 468 cells, 3.1-fold in MCF-7 cells and 1.2-fold in A549 cells (P=0.027 for breast cancer cell lines and P=0.505 for A549 cells). These results suggest that 100 and 200 µM VC, as well as 50 and 75 µM Q, have a prominent effect on the modulation of Nrf2 expression in tumor cells.

Effects of sequential treatment with VC and Q on xenobiotic metabolizing enzymes and thiol content

To investigate how treatment with VC and Q affects Nrf2-regulated genes, the levels of xenobiotic metabolizing enzymes and thiol content in solid tumor cells were determined. There were no significant changes in GPx or GR activities in MDA-MB 231, MCF7 or A549 cells following exposure of cells to sequential treatment of VC and Q (Fig. 4A and B). However, both these parameters were significantly decreased in MDA-MB-468 cells (P=0.027), indicating a significant reduction in the level of antioxidant enzymes. In the MDA-MB 231 and MCF-7 cell lines, HO1 was significantly suppressed following treatment with VC and Q (Fig. 4C). Additionally, NQO1 activity was significantly decreased in all treated cells (Fig. 4D). The most prominent changes were observed in MDA-MB 231 cells, suggesting that this cell line, which has higher levels of Nrf2 expression, is more sensitive to the suppressive effects of VC and Q than the other cell lines evaluated.

Inhibitory effect of sequential treatment with VC and Q on intracellular ROS levels

The baseline DCF florescence measurement indicated that 30 h of sequential treatment with VC and Q significantly decreased endogenous ROS levels in a dose-dependent manner (Fig. 5A). By contrast, sequential treatment of cells with VC and Q did not modulate cellular thiol levels in tumor cells (Fig. 5B).

Discussion

Identifying novel and potential molecular targets for cancer therapy is the goal of current studies that aim to decrease the side effects of chemotherapy agents and overcome chemo-resistance. Nrf2 and downstream target genes serve a pivotal role in cellular redox homeostasis, elimination of ROS and xenobiotic metabolism (5). Persistent Nrf2-mediated antioxidant responses promote malignant progression, development of acquired apoptotic resistance and chemo-resistance in cancer cells (30,31). Therefore, the identification of stable, safe and potent Nrf2 inhibitors to decrease the antioxidant response and reduce drug metabolism in tumor cells is urgently required. In a pilot study in our laboratory (Recombinant Protein Laboratory, School of Medicine, Shiraz University of Medical Sciences), it was demonstrated that treatment of MDA-MB 231 cells with 25 µM Q increased the expression of Nrf2, while 50 and 75 µM Q decreased the mRNA levels of Nrf2. In addition, suppression of Nrf2 mRNA was detected when the cells were treated with 50–400 µM VC (data not shown).

It has been reported that treatment of HepG2 cells with 50 µM Q inhibited Nrf2 activation by decreasing the nuclear translocation of Nrf2 and the nuclear content of phosphorylated Nrf2 (24). Furthermore, changes in the redox state caused by antioxidants such as VC inhibited Nrf2-mediated gene expression and overcame resistance to imatinib (16). Therefore, the doses of VC and Q used in the current study were pharmacological and selected according to their low levels of toxicity, as well as their efficacy at inhibiting Nrf2 expression. The results of the present study demonstrated that treatment with Q, a tumor-active phytochemical, and VC, an antioxidant agent with antineoplastic activity, resulted in a significant decrease in Nrf2 expression and induced oxidative stress in cancer cells. Sequential treatment of solid tumor cells with VC and Q reduced the mRNA and protein levels of Nrf2. Suppression of Nrf2 protein expression was notable, and the overall response indicated that the aforementioned treatment decreased Nrf2 mRNA and protein expression, as well as reducing the stability of Nrf2 protein, leading to the suppression of Nrf2.

In various types of cancer cells, Q inhibits cell growth and induces apoptosis; however, it also induces the expression of antioxidant proteins involved in the elimination of ROS, thus protecting cells against oxidative damage (32,33). Flavonoids, including VC and Q, have emerged as an effective adjuvant in cancer therapy, due to acting as free radical scavengers and immune system modulators, in addition to exhibiting antioxidant properties (34). It has been demonstrated that treatment of MSTO-211H lung cancer cells with 20 µM Q stimulated an increase in the levels of cellular Nrf2, upregulation of Nrf2 mRNA and protein and an increase in the affinity of Nrf2 for binding to ARE-driven reporter sequences, consequently boosting the expression of downstream genes compared with untreated cells (35). However, at doses of Q ≥60 µM, the levels of Nrf2 protein were not affected (35). It has been suggested that Q may act as a ‘double-edged sword’ due to its unique properties, since it behaves as an antioxidant and/or pro-oxidant depending on its concentration and the duration of exposure (35). Treatment of HepG2 cells with 50 µM Q induced activation of p38 following 4 h of treatment. By contrast, following18 h of incubation, the level of p38 expression detected was similar to that of the control cells. Nrf2 expression was inhibited at both incubation times, and Q (50 µM) induced a time-dependent activation of p38, in parallel with a transient stimulation of Nrf2, provoking its inhibition later (24).

VC has been employed as an adjuvant for the treatment of cancer patients, as it acts as pro-oxidant by generating ascorbate radicals and hydrogen peroxide against the growth of tumor cells but not against that of normal cells (36). It has been indicated that VC significantly inhibits tumor growth in Lewis lung carcinoma (LLC)-bearing mice at low and high doses (37). Addition of 0.125 mM ascorbic acid to KCL22/SR cells markedly reduced their peroxide levels and inhibited the formation of the Nrf2/DNA complex in KCL22/SR cells, without any changes in the level of Nrf2 protein in the total cell lysate, suggesting that ascorbic acid represses the translocation of Nrf2 into the nucleus (16). Since oxidative stress stimulated the translocation of Nrf2 into the nucleus, a shift in intracellular redox balance towards a reduced condition may hinder the movement of Nrf2 in KCL22/SR cells. The results of the current study demonstrated that HO1 activity was reduced in MDA-MB 231 and MCF-7 cells, and that the activity of NQO1 diminished significantly in all tumor cell lines following treatment with VC and Q. To maintain homeostasis during oxidative stress, cells enhance the GSH concentration and upregulate glutathione-related enzymes to prevent potential oxidative insults and suppress oxidative-stress induced injuries (38,39). NQO1 and HO-1 are two major downstream targets of Nrf2, and serve a pivotal role in the maintenance of cellular redox homeostasis, thus preventing the transformation of normal cells to precancerous or malignant ones by counteracting ROS-mediated carcinogenesis (40). However, it was demonstrated that NQO1, in parallel with Nrf2 overexpression, aberrantly elevated the levels of HO1 in different types of cancer (40,41). Minaei et al demonstrated the effectiveness of nano-Qin decreasing the levels of NQO1 and multidrug resistance-associated protein 1without altering Nrf2 expression (41). Ren et al reported that Q decreased the half-life of Nrf2 by means of ubiquitination systems, which led to a reduction in the gene expression levels. It was also demonstrated that treatment of human keratinocytes with 50 µM VC had no effect on NQO1 or HO1 activities (42). Furthermore, it was identified that administering injections of VC to tumor cells increased the carbonyl levels in the liver, but reduced the GSH/GSSG ratio in the liver and kidney (43). Therefore, it was suggested that high-dose VC has a bifunctional role: i) Pro-oxidant activity against tumor growth; and ii) antioxidant activity against oxidative stress and nephrotoxicity induced by cisplatin in LLC-bearing mice (37). It was demonstrated that treatment of human keratinocytes with 50 µM VC did not affect NQO1 or HO1 activity (43). Additionally, it has been demonstrated that treatment of HepG2 cells with Q for 4 h enhanced the activity of GPx and GR, and increased GSH levels as well as GCS expression, therefore suggesting that Q can mediate the expression of GSH-related enzymes (44). This may be associated with the fact that Nrf2 activation is a master regulator upstream the GPx and GR genes. Similarly, following acute stress, GSH levels may be temporarily suppressed and subsequently recovered, due to an increase in GCS activity and mRNA levels. GSH levels therefore may be a signal of cellular self-protection against a sub-lethal toxic insult. High levels of Q, which induce toxicity, overcome the defense mechanisms of the cell, such as the Nrf2 response (45). By contrast, the results of the current study suggest that unchanged levels of GPx, GR and GSH may imply impairment in the machinery involved in the gene transcription and mRNA synthesis of antioxidant enzymes. It was demonstrated that ROS levels significantly increased in tumor cells treated with VC and Q. The capability of Q to reduce the levels of accumulated intracellular ROS indicated that the protective effects of flavonoids are not only limited to their antioxidant properties, but they can also act as ROS scavengers in the extracellular medium (46). Treatment with 1 or 5 mM ascorbate increased the levels of intracellular ROS in ReN cells but not in mesothelium cells. Additionally, it was demonstrated that malignant mesothelioma cells enhanced superoxide production and induced overexpression of the superoxide-producing NADPH oxidase 4 (47). This discrepancy between data collected in the current study and previous studies may be due to different doses of agents used, duration of treatments and the sequential treatments that were employed.

The results of the present study indicate that targeting Nrf2 may be a promising strategy to induce oxidative stress, which in turn represents a potential efficient method of sensitizing tumor cells. Furthermore, Nrf2 may be a determining factor for inhibition in chemotherapy protocols. The sequential treatment of solid tumor cells with VC and Q reduced the expression of Nrf2 at the mRNA and protein levels. Therefore, further studies to establish the efficacy and safety of antioxidant adjuvants in vivo and in humans are required to establish evidence-based guidelines on their use in cancer therapy, in order to obtain optimal therapeutic outcomes in patients with cancer.

Acknowledgements

The present study was performed both as a part of a PhD student thesis by Miss Fatemeh Ramezani (Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran) and an MSc student thesis by Mrs. Fatemeh Keshavarzi (Department of Biochemistry, Recombinant Protein Laboratory, School of Medicine, Shiraz University of Medical Sciences), who received grants (grant nos. 92–6659 and 94–7441, respectively) from the office of Vice Chancellor for Research and the Committee for Advanced Biomedical Sciences, Shiraz University of Medical Sciences.

References

1 

Ramos S: Effects of dietary flavonoids on apoptotic pathways related to cancer chemoprevention. J Nutr Biochem. 18:427–442. 2007. View Article : Google Scholar : PubMed/NCBI

2 

No JH, Kim YB and Song YS: Targeting nrf2 signaling to combat chemoresistance. J Cancer Prev. 19:111–117. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Wu T, Harder BG, Wong PK, Lang JE and Zhang DD: Oxidative stress, mammospheres and Nrf2-new implication for breast cancer therapy? Mol Carcinog. 54:1494–1502. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Jaramillo MC and Zhang DD: The emerging role of the Nrf2-Keap1 signaling pathway in cancer. Genes Dev. 27:2179–2191. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Kensler TW, Wakabayashi N and Biswal S: Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 47:89–116. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Foygel K, Sekar TV and Paulmurugan R: Monitoring the antioxidant mediated chemosensitization and ARE-signaling in triple negative breast cancer therapy. PloS One. 10:e01419132015. View Article : Google Scholar : PubMed/NCBI

7 

Wang XJ, Sun Z, Villeneuve NF, Zhang S, Zhao F, Li Y, Chen W, Yi X, Zheng W, Wondrak GT, et al: Nrf2 enhances resistance of cancer cells to chemotherapeutic drugs, the dark side of Nrf2. Carcinogenesis. 29:1235–1243. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Amadori D, Frassineti G, Zoli W, Milandri C, Serra P, Tienghi A, Ravaioli A, Gentile A and Salzano E: Doxorubicin and paclitaxel (sequential combination) in the treatment of advanced breast cancer. Oncology (Williston Park). 11:(4 Suppl 3) 30–33. 1997.PubMed/NCBI

9 

Danesi R, Conte PF and Del Tacca M: Pharmacokinetic optimisation of treatment schedules for anthracyclines and paclitaxel in patients with cancer. Clin Pharmacokinet. 37:195–211. 1999. View Article : Google Scholar : PubMed/NCBI

10 

Ibrahim T, Fabbri M, Frassineti GL, Zoli W, Monti M, Ricotti L and Amadori D: Doxorubicin, paclitaxel and gemcitabine: A Phase I study of a new sequential treatment in stage III B-IV breast cancer. J Chemother. 15:488–494. 2003.PubMed/NCBI

11 

Chou TC: Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev. 58:621–681. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Lee KW, Lee HJ, Surh YJ and Lee CY: Vitamin C and cancer chemoprevention: Reappraisal. Am J Clin Nutr. 78:1074–1078. 2003.PubMed/NCBI

13 

Zielinski CC: Gemcitabine, anthracycline, and taxane combinations for advanced breast cancer. Oncology (Williston Park). 17:(12 Suppl 14) 36–40. 2003.PubMed/NCBI

14 

Chao MW, Lai MJ, Liou JP, Chang YL, Wang JC, Pan SL and Teng CM: The synergic effect of vincristine and vorinostat in leukemia in vitro and in vivo. J Hematol Oncol. 8:822015. View Article : Google Scholar : PubMed/NCBI

15 

Weiss RB, Woolf SH, Demakos E, Holland JF, Berry DA, Falkson G, Cirrincione CT, Robbins A, Bothun S, Henderson IC, et al: Natural history of more than 20 years of node-positive primary breast carcinoma treated with cyclophosphamide, methotrexate, and fluorouracil-based adjuvant chemotherapy: A study by the cancer and leukemia group B. J Clin Oncol. 21:1825–1835. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Tarumoto T, Nagai T, Ohmine K, Miyoshi T, Nakamura M, Kondo T, Mitsugi K, Nakano S, Muroi K, Komatsu N and Ozawa K: Ascorbic acid restores sensitivity to imatinib via suppression of Nrf2-dependent gene expression in the imatinib-resistant cell line. Exp Hematol. 32:375–381. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Khanduja KL, Gandhi RK, Pathania V and Syal N: Prevention of N-nitrosodiethylamine-induced lung tumorigenesis by ellagic acid and quercetin in mice. Food Chem Toxicol. 37:313–318. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Saw CL, Guo Y, Yang AY, Paredes-Gonzalez X, Ramirez C, Pung D and Kong AN: The berry constituents quercetin, kaempferol, and pterostilbene synergistically attenuate reactive oxygen species: Involvement of the Nrf2-ARE signaling pathway. Food Chem Toxicol. 72:303–311. 2014. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Granado-Serrano AB, Martín MA, Bravo L, Goya L and Ramos S: Quercetin induces apoptosis via caspase activation, regulation of Bcl-2, and inhibition of PI-3-kinase/Akt and ERK pathways in a human hepatoma cell line (HepG2). J Nutr. 136:2715–2721. 2006.PubMed/NCBI

21 

Li N, Sun C, Zhou B, Xing H, Ma D, Chen G and Weng D: Low concentration of quercetin antagonizes the cytotoxic effects of anti-neoplastic drugs in ovarian cancer. PloS One. 9:e1003142014. View Article : Google Scholar : PubMed/NCBI

22 

Robaszkiewicz A, Balcerczyk A and Bartosz G: Antioxidative and prooxidative effects of quercetin on A549 cells. Cell Biol Int. 31:1245–1250. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Samuel T, Fadlalla K, Mosley L, Katkoori V, Turner T and Manne U: Dual-mode interaction between quercetin and DNA-damaging drugs in cancer cells. Anticancer Res. 32:61–71. 2012.PubMed/NCBI

24 

Granado-Serrano AB, Martín MA, Bravo L, Goya L and Ramos S: Quercetin modulates Nrf2 and glutathione-related defenses in HepG2 cells: Involvement of p38. Chem Biol Interact. 195:154–164. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Pfaffl MW: Quantification strategies in real-time PCR. A-Z of quantitative PCR. 3:87–112. 2004.

26 

Sabzichi M, Samadi N, Mohammadian J, Hamishehkar H, Akbarzadeh M and Molavi O: Sustained release of melatonin: A novel approach in elevating efficacy of tamoxifen in breast cancer treatment. Colloids Surf B Biointerfaces. 145:64–71. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Fecondo JV and Augusteyn RC: Superoxide dismutase, catalase and glutathione peroxidase in the human cataractous lens. Exp Eye Res. 36:15–23. 1983. View Article : Google Scholar : PubMed/NCBI

28 

Maiani G, Mobarhan S, Nicastro A, Virgili F, Scaccini C and Ferro-Luzzi A: Determination of glutathione reductase activity in erythrocytes and whole blood as an indicator of riboflavin nutrition. Acta Vitaminol Enzymol. 5:171–178. 1982.(In Italian).

29 

Ellman GL: Tissue sulfhydryl groups. Arch Biochem Biophys. 82:70–77. 1959. View Article : Google Scholar : PubMed/NCBI

30 

Furfaro A, Traverso N, Domenicotti C, Piras S, Moretta L, Marinari UM, Pronzato MA and Nitti M: The Nrf2/HO-1 axis in cancer cell growth and chemoresistance. Oxid Med Cell Longev. 2016:19581742016. View Article : Google Scholar : PubMed/NCBI

31 

van der Wijst MG, Brown R and Rots MG: Nrf2, the master redox switch: The Achilles' heel of ovarian cancer? Biochim Biophys Acta. 1846:494–509. 2014.PubMed/NCBI

32 

Liu KC, Yen CY, Wu RS, Yang JS, Lu HF, Lu KW, Lo C, Chen HY, Tang NY, Wu CC and Chung JG: The roles of endoplasmic reticulum stress and mitochondrial apoptotic signaling pathway in quercetin-mediated cell death of human prostate cancer PC-3 cells. Environ Toxicol. 29:428–439. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Lee WJ, Hsiao M, Chang JL, Yang SF, Tseng TH, Cheng CW, Chow JM, Lin KH, Lin YW, Liu CC, et al: Quercetin induces mitochondrial-derived apoptosis via reactive oxygen species-mediated ERK activation in HL-60 leukemia cells and xenograft. Arch Toxicol. 89:1103–1117. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Bournival J, Francoeur MA, Renaud J and Martinoli MG: Quercetin and sesamin protect neuronal PC12 cells from high-glucose-induced oxidation, nitrosative stress, and apoptosis. Rejuvenation Res. 15:322–333. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Bouayed J and Bohn T: Exogenous antioxidants-double-edged swords in cellular redox state: Health beneficial effects at physiologic doses versus deleterious effects at high doses. Oxid Med Cell Longev. 3:228–237. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Carr AC, Vissers MC and Cook JS: The effect of intravenous vitamin C on cancer-and chemotherapy-related fatigue and quality of life. Front Oncol. 4:2832014. View Article : Google Scholar : PubMed/NCBI

37 

Chen MF, Yang CM, Su CM and Hu ML: Vitamin C protects against cisplatin-induced nephrotoxicity and damage without reducing its effectiveness in C57BL/6 mice xenografted with Lewis lung carcinoma. Nutr Cancer. 66:1085–1091. 2014. View Article : Google Scholar : PubMed/NCBI

38 

Wang X, Ye XL Liu R, Chen HL, Bai H, Liang X, Zhang XD, Wang Z, Li WL and Hai CX: Antioxidant activities of oleanolic acid in vitro: Possible role of Nrf2 and MAP kinases. Chem Biol Interact. 184:328–337. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Ding M, Zhao J, Bowman L, Lu Y and Shi X: Inhibition of AP-1 and MAPK signaling and activation of Nrf2/ARE pathway by quercitrin. Int J Oncol. 36:59–67. 2010.PubMed/NCBI

40 

Chun KS, Kundu J, Kundu JK and Surh YJ: Targeting Nrf2-Keap1 signaling for chemoprevention of skin carcinogenesis with bioactive phytochemicals. Toxicol Lett. 229:73–84. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Minaei A, Sabzichi M, Ramezani F, Hamishehkar H and Samadi N: Co-delivery with nano-quercetin enhances doxorubicin-mediated cytotoxicity against MCF-7 cells. Mol Biol Rep. 43:99–105. 2016. View Article : Google Scholar : PubMed/NCBI

42 

Ren D, Villeneuve NF, Jiang T, Wu T, Lau A, Toppin HA and Zhang DD: Brusatol enhances the efficacy of chemotherapy by inhibiting the Nrf2-mediated defense mechanism. Proc Natl Acad Sci USA. 108:1433–1438. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Wagner AE, Ernst I, Iori R, Desel C and Rimbach G: Sulforaphane but not ascorbigen, indole-3-carbinole and ascorbic acid activates the transcription factor Nrf2 and induces phase-2 and antioxidant enzymes in human keratinocytes in culture. Exp Dermatol. 19:137–144. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Scharf G, Prustomersky S, Knasmuller S, Schulte-Hermann R and Huber WW: Enhancement of glutathione and g-glutamylcysteine synthetase, the rate limiting enzyme of glutathione synthesis, by chemoprotective plant-derived food and beverage components in the human hepatoma cell line HepG2. Nutr Cancer. 45:74–83. 2003. View Article : Google Scholar : PubMed/NCBI

45 

Goldring CE, Kitteringham NR, Elsby R, Randle LE, Clement YN, Williams DP, McMahon M, Hayes JD, Itoh K, Yamamoto M and Park BK: Activation of hepatic Nrf2 in vivo by acetaminophen in CD-1 mice. Hepatology. 39:1267–1276. 2004. View Article : Google Scholar : PubMed/NCBI

46 

Hanneken A, Lin FF, Johnson J and Maher P: Flavonoids protect human retinal pigment epithelial cells from oxidative-stress-induced death. Invest Ophthalmol Vis Sci. 47:3164–3177. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Parrow NL, Leshin JA and Levine M: Parenteral ascorbate as a cancer therapeutic: A reassessment based on pharmacokinetics. Antioxid Redox Signal. 19:2141–2156. 2013. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2017
Volume 13 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mostafavi‑Pour Z, Ramezani F, Keshavarzi F and Samadi N: The role of quercetin and vitamin C in Nrf2‑dependent oxidative stress production in breast cancer cells. Oncol Lett 13: 1965-1973, 2017
APA
Mostafavi‑Pour, Z., Ramezani, F., Keshavarzi, F., & Samadi, N. (2017). The role of quercetin and vitamin C in Nrf2‑dependent oxidative stress production in breast cancer cells. Oncology Letters, 13, 1965-1973. https://doi.org/10.3892/ol.2017.5619
MLA
Mostafavi‑Pour, Z., Ramezani, F., Keshavarzi, F., Samadi, N."The role of quercetin and vitamin C in Nrf2‑dependent oxidative stress production in breast cancer cells". Oncology Letters 13.3 (2017): 1965-1973.
Chicago
Mostafavi‑Pour, Z., Ramezani, F., Keshavarzi, F., Samadi, N."The role of quercetin and vitamin C in Nrf2‑dependent oxidative stress production in breast cancer cells". Oncology Letters 13, no. 3 (2017): 1965-1973. https://doi.org/10.3892/ol.2017.5619