Open Access

Assessment of the antioxidant activity of an olive oil total polyphenolic fraction and hydroxytyrosol from a Greek Olea europea variety in endothelial cells and myoblasts

  • Authors:
    • Paraskevi Kouka
    • Alexandros Priftis
    • Dimitrios Stagos
    • Apostolis Angelis
    • Panagiotis Stathopoulos
    • Nikos Xinos
    • Alexios‑Léandros Skaltsounis
    • Charalampos Mamoulakis
    • Aristides M. Tsatsakis
    • Demetrios A. Spandidos
    • Demetrios Kouretas
  • View Affiliations

  • Published online on: July 20, 2017     https://doi.org/10.3892/ijmm.2017.3078
  • Pages: 703-712
  • Copyright: © Kouka et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Olive oil (OO) constitutes the basis of the Mediterranean diet, and it seems that its biophenols, such as hydroxytyrosol (HT) may scavenge free radicals, attracting distinct attention due to their beneficial effects in many pathological conditions, such as cancer. To the best of our knowedge, this is the first study in which the functional properties of an OO total polyphenolic fraction (TPF) and pure HT were examined in order to determine their antioxidant effects at a cellular level in endothelial cells and myoblasts. The test compounds were isolated using a green gradient‑elution centrifugal partition chromatography‑based method that allows the isolation of large volumes of OO in a continuous extraction procedure and with extremely low solvent consumption. For the isolation of HT, a combination of two chromatographic techniques was used, which is effective for the recovery of pure compounds from complex natural extracts. Moreover, TPF and HT exhibited potent free radical scavenging activity in vitro. The cells were treated with non‑cytotoxic concentrations and their redox status [in terms of glutathione (GSH) and reactive oxygen species (ROS) levels] was assessed. TPF extract was less cytotoxic than HT, and the observed differences between the two cell lines used suggest a tissue‑specific activity. Finally, flow cytometric analysis revealed that both TPF and HT improved the redox status by increasing the levels of GSH, one of the most important antioxidant molecules, in both endothelial cells and myoblasts, while the ROS levels were not significantly affected.

Introduction

Aerobic organisms are exposed to reactive oxygen species (ROS) due to aerobic metabolism. ROS, such as superoxide anion (O2•−), hydrogen peroxide (H2O2) and hydroxyl radicals (OH), have coalescent chemical properties that confer reactivity to a number of biological targets (1). ROS are often associated with oxidative stress, a condition characterized by the imbalance between the production of free radicals and the endogenous antioxidant mechanisms in the favor of the first (1). ROS are capable of reversibly or irreversibly damaging biological macromolecules, such as nucleic acids, proteins, lipids and carbohydrates (1). Thus, oxidative stress may lead to the development of a number of pathological conditions, such as cancer, diabetes, obesity, and neurodegenerative and autoimmune diseases (2). However, ROS also serve as signaling molecules which regulate biological and physiological processes, such as the regulation of signaling pathways, gene expression and apoptosis (1). In living organisms, there is a wide range of endogenous defensive mechanisms against free radicals, including enzymatic and non-enzymatic antioxidants (2). Aside from the endogenous mechanisms, an organism may also acquire antioxidant components through diet (3,4). Polyphenols, products of secondary metabolism in plants, are some of the most important antioxidants (5,6).

The entire vascular system is comprised of a monolayer of endothelial cells. Endothelial cell integrity is necessary to the maintenance of the vessel wall and circulatory function (7,8). Moreover, the endothelium regulates the homeostasis, immune and inflammatory responses (8). Oxidative stress may cause damage to the vascular endothelium due to the loss of its integrity, leading to senescence and its detachment into the circulation (9). Thus, it consists one of the most important factors of pathological conditions of vessels, including atherosclerosis and thrombosis (8). In addition, oxidative stress may cause damage to the endothelium through leukocyte adhesion (10,11). Moreover, the interplay between ROS and nitric oxide sets off a vicious circle which leads to further endothelial activation and inflammation (8).

Oxidative stress also occurs in muscle tissue. Specifically, during intense exercise, a high rate of O2 consumption takes place in skeletal muscle that can cause incomplete O2 reduction and electron leakage from the electron transfer chain, leading to ROS generation and oxidative stress. Moreover, exercise induces oxidative stress through xanthine oxidase mechanisms (12). Oxidative stress, in turn, results in muscle fatigue, cell damage and apoptosis (13,14).

The medicinal properties of Olea europaea (olive tree) and its main product, olive oil (OO), has been known since ancient times (15). OO is the most important component of the Mediterranean diet (16), characterized by the intake of plant foods, such as fruits, vegetables, nuts and seeds (17). Epidemiological evidence indicates an inverse association with proper adherence to this diet and death due to coronary artery disease and various types of cancer, such as urinary bladder cancer, suggesting a potential protective role of regularly consumed OO and its hydrophilic fraction physiologically excreted through urine (1820). The results of in vitro studies further support this evidence by revealing a protective effect of OO against various types of cancer (2124), including urogenital neoplasms (2527), paving the way towards research for the development of novel anticancer strategies. It has been shown in human prostate cell cultures for example, that exposure to OO induces an antioxidant effect on benign prostatic hyperplasia-1 cells and a pro-oxidant effect on malignant cells, suggesting that OO may potentially be used for prostate cancer prevention (25). Furthermore, it has been reported that increased OO consumption raises the concentration of phenol glucuronide conjugates in a dose-dependent manner in human urine (28) and that extra virgin OO (EVOO) extract suppresses the migration and invasion of T24 human bladder cancer cells by significantly inhibiting proliferation/motility in a dose-dependent manner (26). These promising results have been replicated in another study showing that EVOO extract significantly inhibited the proliferation and clonogenic ability of T24 and 5637 human bladder cancer cells in a dose-dependent manner by blocking the cell cycle, and that EVOO modulated the chemotherapeutic toxicity in bladder cancer cells (27).

Over the past 25 years, the consumption of OO worldwide has increased by >30% due to its two key characteristics, namely its nutritional and organoleptic properties (16,29). The nutritional properties of OO are provided by its fatty acid (FA) profile and high monounsaturated/saturated FA ratio (MUFA/SFA), as well as by its rich antioxidant content, particularly that of biophenols which are believed to play a role in the prevention of diseases (16,30). OO biophenols act as oxidation chain-breakers, reacting with free radicals and forming inactive radicals. Some of the most important biophenols found in OO with marked biological activities, are oleocanthal (OLEO), oleacein (OLEA), elenoic acid, oleuropein and its derivatives, tyrosol (T) and hydroxytyrosol (HT) (31). These compounds have the ability to scavenge free radicals by donating them an electron or hydrogen atom or by chelating metals (16). Among the OO biophenols, HT has attracted distinct attention due to its potent antioxidant activity attributed mainly to its orthodiphenolic structure (32). It is mainly found in olive tree leaves, olive pulp and OO (33). It is worth mentioning that HT can also be synthesized endogenously in the human body, as a product of dopamine oxidative metabolism, known as 3,4-dihydroxyphenyl ethanol (DOPET) (34). HT has been shown to exert preventive effects in several pathological conditions, such as metabolic syndrome, neurodegenerative diseases and cancer (34).

The aim of the present study was to examine the antioxidant properties of a EVOO total polyphenolic fraction (TPF) rich in biophenols, from a Greek endemic variety of Olea europea, grown on Mount Athos, as well as that of pure HT. It is worth noting, that to date, at least to the best of our knowledge, there are no studies available investigating the antioxidant properties of an OO TPF in cell cultures. The antioxidants effects were assessed at a cellular level, particularly in EA.hy926 endothelial cells and C2C12 myoblasts. The obtained results will provide important information on the possible use of the TPF as an antioxidant food supplement.

Materials and methods

Reagents and materials

n-Hexane, ethyl acetate, ethanol (etOH) and acetonitrile were purchased from Carlo Erba Reactifs SDS (Val de Reuil, France). Methanol (MeOH), dichloromethane and sulfuric acid (H2SO4 >95%) were obtained from Fisher Scientific UK (Leicestershire, UK). All solvents were of analytical grade. Deionized water was used to prepare all aqueous solutions. Vanillin standard were purchased from Sigma-Aldrich (Poole, UK) with purity of >95%.

Extraction of TPF from EVOO

EVOO was procured from Northern Greece (Mount Athos) which was freshly produced in January 2015). The extraction of TPF was achieved following a previously described procedure (35). Liquid-liquid extraction was carried out on a laboratory-scales Fast Centrifugal Partition Extractor FCPE300® (Rousselet-Robatel Kromaton, Anonay, France; column capacity of 300 ml) connected to a LabAlliance preparative pump. The extraction process consisted of several 'Extraction-Recovery' cycles (multi-dual-mode method) using as 'mobile phase' a mixure of n-hexane/OO in proportion of 3:2 (v/v) and as 'stationary phase' etOH/water in proportion of 3:2 (v/v). The procedure starts by filling the CPE column with 0.3 liters of the stationary phase in ascending mode and at 200 rpm. The rotation speed was increased up to 1,000 rpm and the mobile phase (feed oil phase) was pumped at 60 ml/min in ascending mode. After passing 2.5 liters of mobile phase (extraction step) the pumping mode was switched to 'descending' and a volume of 0.3 liters of aqueous extraction phase was pumped (at the same flow rate and rotation speed) in order to replace the concentrated in biophenols stationary phase with fresh one (recovery step). This cycle of extraction-recovery was repeated 5 more times extracting totally 15 liters of feed oil phase corresponding to 6 liters of OO. The 6 collected stationary phases were evaporated under a vacuum and the obtained viscous extract was defatted using the biphasic system n-hexane/acetonitrile (1/1 v/v) and stored at 4°C for further use. Finally, the extraction of 6 liters of OO resulted in the recovery of 6.35 g of OO biophenol extract.

Purification of HT from TPF

High purity HT was recovered from the treatment of TPF in a two steps separation process. Initially, the extract was fractionated by gradient-elution centrifugal partition chromatography (CPC) and the enriched in the HT fraction was further analyzed by using Sephadex LH-20 (Sigma-Aldrich) column chromatography. The CPC experiment was performed on a FCPC1000® apparatus (Rousselet-Robatel Kromaton) equipped with a preparative column (1,000 ml total column capacity) and connected to a LabAlliance Preparative pump. The system was coupled to a SPECTRA SYSTEM UV 2000 detector set at 254, 280 and 366 nm (Thermo Fisher Scientific, Inc., Waltham, MA, USA), while fractions were collected using a Büchi B-684 fraction collector (Büchi Labortechnik AG, Flawil, Switzerland).

The TPF was fractionated in a step-gradient elution mode following a previously described method (35). For this purpose, a series of 4 biphasic systems consisting of the solvents, n-hexane, ethyl acetate, etOH, and water in proportions 4/1/2/3 (S1), 3/2/2/3 (S2), 2/3/2/3 (S3) and 1/4/2/3 v/v/v/v (S4) were used. The solvents were thoroughly mixed in a separating funnel at room temperature prior to use, and the 2 phases of each system were separated after equilibration of the mixture. The column was initially filled with the lower phase of the first biphasic system (stationary phase) in ascending mode at a flow rate of 30 ml/min and 200 rpm. The rotation speed was increased to 1,000 rpm and the mobile phase I (upper phase of S1) was pumped at a flow rate of 15 ml/min. When the hydrodynamic equilibrium of the two liquid phases inside the CPC column was established (Sf=67%), 5 g of sample (diluted in 20 ml of lower and 10 ml of upper phase of S1) was injected into the column via a 30 ml sample loop. A sequential pumping of the upper mobile phases of S1, S2, S3 and S4 was then performed in volumes of 500, 1,100, 1,400 and 1,000 ml, respectively (gradient elution step). The experiment completed by pumping the stationary phase in ascending mode (at the same flow rate and rotation speed) and collection of 30 more fractions (extrusion step). Fraction collector was set to collect 25 ml fractions during the all experiment.

All collected fractions were evaluated using thin layer chromatography (TLC). The plates were coated (Merck Millipore, Billerica, MA, USA) with silica gel 60 F254 and developed in dichloromethane/MeOH in different proportions. After detection at UV254 and UV366, the plates were sprayed with vanillin-sulfuric acid and heated. Based on the TLC qualitative results, the fractions containing HT (195 fractions of 25 ml) were combined and evaporated to dryness resulting to 95 mg of an enriched HT fraction (86% purity). For further purification, Sephadex LH-20 column chromatography was incorporated.

Specifically, 90 mg of the sample (diluted in 1 ml EtOH) were subjected to column chromatography (Sephadex® LH-20, 10×285 mm) and eluted with EtOH. All obtained fractions were initially analyzed by TLC and those of similar chemical content were combined giving finally three main fractions (Fractions A-C). Fraction B contained 71.3 mg HT in high purity (>97%). The purity was determined by quantitative high-performance liquid chromatography (HPLC) analysis.

HPLC-diode-array detection (DAD) qualitative and quantitative analysis

The qualification analysis of TPF and quantification of HT, T, OLEO and OLEA were performed on a Thermo Finnigan HPLC instrument (Ontario, Canada) equipped with a SpectraSystem P4000 pump, a SpectraSystem 1000 degasser, a SpectraSystem AS3000 automated injector, and a UV SpectraSystem UV6000LP detector monitored at 235, 280 and 365 nm. The samples were analyzed in a Discovery HS C18 (15cm x 4.6mm, particle size 5 µm) analytical column (Supelco, Bellefonte, PA, USA) using two different methods. The first one (method A) was used for the qualification analysis of the stable TPF components and quantification of HT and T, and was based on a previously published method with minor modifications (36). Water acidified with 0.2% acetic acid (solvent A) and acetonitrile (solvent B) was used as a solvent in the following gradient system: 0–40 min 2–30% B, 40–45 min 30% B, 45–50 min 30–2% B. The flow rate was set at 1 ml/min, the injection volume was 20 µl (from a solution of 1 mg TPF/ml MeOH) and the total run duration was 50 min. The qualification analysis of the labile TPF components and quantification analysis of OLEA and OLEO was performed using the second method (method B). The mobile phase consisted of water (solvent A) and acetonitrile (solvent B), while the gradient elution began with 80% of solvent A decreasing to 70% in 20 min, remaining stable for 15 min and increasing again to 80% in the next 5 min (total duration, 40 min). The flow rate was set at 1 ml/min, while the injection volume was 20 µl (from a solution of 1 mg TPF/ml MeOH), as previously described (37,38).

The quantification analysis of HT and T was performed using method A. The calibration curves were created using the concentrations 2, 3, 20, 50 and 100 µg/ml and was used to calculate the percentage of the target compounds in EVOO and TPF, as well as to examine the purity of HT both in CPC fraction and in the final pure form. The peak area values (measured at 280 nm) constitute the average of 3 measurements. Linear regression analysis with the use of syringaldehyde (98% HPLC; ExtraSynthese, Genay Cedex, France) as an internal standard (ISTD) was performed.

The quantification of OLEO and OLEA was performed using method B. The calibration curves were constructed using 10 concentrations levels (50,100, 200, 300, 400, 500, 600, 700, 800, 900 and 1,000 µg/ml). ISTD was prepared in a mixture of MeOH/water 1:1 (v/v) at a concentration of 1 mg/ml and stored at −4°C. Both compounds are eluted, as double peaks and thus the quantification was based on the total area of both peaks (measured at 235 nm) as previously described by Impellizzeri and Lin (37). In the case of HT and T, the quantification was based on the ratio between the peak area of the analyte and the peak area of ISTD. The equations and the coefficient of determination (R2) of the standard curves were as follows: y=45130x+22444, R2=0.995 for OLEO, y=43653x-1E+06, R2=0.997 for OLEA, y=0.041x-0.011, R2=0.999 for HT and y=0.025x-0,014, R2=0.999 for T.

TLC and NMR analysis

The TLC analysis of TPF and CPC fractions was performed on Merck 60 F254pre-coated silica gel plates and developed with DCM/MeOH 95:5 (v/v). The TLC chromatograms were firstly revelation by using UV light at 254 and 365 nm and the TLC plates were then sprayed by a vanillin (5% w/v in ethanol)-H2SO4 (50% v/v in methanol) solution and heated at 100–120°C for 2–3 min. Structure confirmation of the isolated HT was achieved by NMR analysis. The 1H-NMR experiments were performed on a 600 MHz on a Bruker AvanceAVIII-600 spectrometer (Karlsruhe, Germany) equipped with a TXI cryoprobe (Wissembourg, France) in CD3OD.

Assessment of the total polyphenolic content (TPC) of the extracts

The TPC of the OO extract was determined using the Folin-Ciocalteu reagent, as previously described (39). A total of 20 µl of extract were added to a tube containing 1 ml distilled water, followed by the addition of 100 µl of Folin-Ciocalteu reagent and incubation for 3 min at room temperature. Subsequently, 280 µl of 25% w/v sodium carbonate solution along with 600 µl of distilled water were added to the mixture. Finally, following 1 h incubation at room temperature in the dark, the absorbance was measured at 765 nm versus a blank lacking the extract. The measurement was carried out on a Hitachi U-1900 radio beam spectrophotometer (serial no. 2023–029; Hitachi, Ltd., Tokyo, Japan). The optical density of the sample without the Folin-Ciocalteu reagent at 765 nm was also measured. The TPC was determined using a gallic acid standard curve (50–1,500 µg/ml). The TPC is presented as µg of gallic acid equivalents per mg of extract.

2,2-diphenyl-1-picrylhydrazyl (DPPH) radical scavenging assay

The free-radical scavenging capacity (RSC) of the TPF and HT extracts was evaluated using the DPPH radical assay (40). Briefly, a 1 ml freshly prepared methanolic solution of DPPH radical (100 µM) was mixed with the tested extract solution at various concentrations (7.8–250 µg/ml TPF and 0.7–100 µg/ml HT). The mixture was vortexed and incubated at room temperature in the dark for 20 min, followed by absorbance measurement at 517 nm on a Hitachi U-1900 radio beam spectrophotometer (serial no. 2023–029; Hitachi, Ltd.). MeOH was used as a blank and DPPH alone in MeOH was used as the control.

The percentage RSC of the tested extracts was calculated using the following equation: RSC (%) = [(ODcontrol − ODsample)/ODcontrol] ×100, where ODcontrol and ODsample are the optical density (OD) values of the control and the test sample, respectively. Moreover, the IC50 value indicating the polyphenolic amount that caused 50% scavenging of the DPPH radical was calculated. In order to compare the radical scavenging efficiency of the extracts, the specific activity was determined. The specific activity was evaluated by dividing the IC50 value obtained from the DPPH and 2,2′-Azinobis-(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS•+) assays by the amount of polyphenols contained in each mg of the TPF and HT. All experiments were carried out in triplicate and on at least 3 separate occasions.

ABTS•+ radical scavenging assay

The ABTS•+ RSC of the extract was determined as previously described by Cano et al (41) with minor modifications. Briefly, 1 ml reactions were prepared in distilled water containing ABTS•+ (1 mM), H2O2 (30 µM) and horseradish peroxidase (6 µM) in 50 mM phosphate-buffered saline (PBS; pH 7.5). The solution was vortexed, followed by incubation for 45 min at room temperature in the dark. Subsequently, 10 µl of the tested extracts, at various concentrations, were added and the absorbance at 730 nm was read on a Hitachi U-1900 radio beam spectrophotometer (serial no. 2023–029; Hitachi, Ltd.). In each experiment, a blank lacking the peroxidase was used, while the ABTS•+ radical solution without the extract was used as the control. The RSC percentage and the specific activity values were determined as described above for the DPPH method. All experiments were carried out in triplicate and on at least 3 separate occasions.

Cell culture conditions

The C2C12 murine myoblasts were cultured in Dulbecco's modified Eagle's medium (DMEM), containing 10% (v/v) fetal bovine serum (FBS), 2 mM L-glutamine, 100 U/ml of penicillin and 100 U/ml of streptomycin (all from Gibco, Paisley, UK) in plastic disposable tissue culture flasks at 37°C in 5% CO2 (42). The C2C12 myoblasts were donated by from Professor Koutsilieris (National and Kapodistrian University of Athens, Athens, Greece).

EA.hy926 endothelial cells were cultured as described previously in tissue culture flasks at 37°C in 5% CO2 (34,42). The medium used was DMEM, containing 10% (v/v) FBS, 25 mM HEPES, 2 mM L-glutamine, 100 U/ml of penicillin and 100 U/ml of streptomycin (Gibco) (42). The EA.hy926 cells were donated by Professor Koukoulis (University of Thessaly, Larissa, Greece).

XTT assay

Cell viability was assessed using the XTT assay kit (Roche, Mannheim, Germany) as described previously (42). Briefly, the C2C12 and EA.hy926 cells were cultured in a 96-well plate in a density of 1×104 cells per well in DMEM. Following 24 h of incubation, various concentrations of the TPF and HT extracts in serum-free DMEM were administered for 24 h. Afterwards, 50 µl of XTT test solution were added to each well. Following 4 h of incubation, absorbance was measured at 450 nm and also at 630 nm as a reference wavelength in a BioTek ELx800 microplate reader (BioTek Instruments, Inc., Winooski, VT, USA). Serum-free DMEM was used as a negative control. In addition, the absorbance of the TPF and HT alone in serum-free DMEM and XTT test solution was measured at 450 nm. The absorbance values of the TPF and HT alone were subtracted from those derived from cell treatment with the tested compounds. Data were calculated as the percentage of viability using the following formula: Viab ility (%) = [(ODcontrol − ODsample)/ODcontrol] ×100, where ODcontrol and ODsample indicate the OD of the negative control and the tested compounds, respectively. All experiments were carried out in triplicate and on 3 separate occasions.

Assessment of GSH and ROS levels in cells by flow cytometry

The intracellular GSH and ROS levels were assessed using the fluorescent dyes, mercury orange and 2,7-dichlorofluorescein diacetate (DCF-DA), respectively (43). Mercury orange binds directly to GSH, while DCF-DA is deacetylated within cells by esterases and is further converted to fluorescent DCF by the oxidative action of ROS. A 400 µM stock solution of mercury orange was prepared in acetone and stored at 4°C, and a fresh 400 µM stock solution of DCF-DA was prepared in MeOH. To measure the GSH and ROS levels the cells were first trypsinized and centrifuged at 300 × g for 5 min at 4°C. The supernatant was discarded and the cell pellet was resuspended in PBS at 1×106 cells/ml and incubated in the presence of mercury orange (40 µM) or DCF-DA (10 µM) in the dark at 37°C for 30 min. The cells were then washed, resuspended in PBS, and subjected to flow cytometric analysis using a FACSCalibur flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA) with excitation and emission wavelengths at 488 and 530 nm for ROS and at 488 and 580 nm for GSH, respectively. In addition, forward-angle and right-angle light scattering showing the cell size and cell internal complexity, respectively, were measured. The cells were analyzed at a flow rate of 1,000 events/sec. Analyses were performed on 10,000 cells per sample, and the fluorescence intensities were measured on a logarithmic scale. Data were analyzed using BD Cell Quest software (BD Biosciences). Each experiment was repeated at least 6 times.

Statistical analysis

All results are expressed as the means ± standard error of the mean. A Spearman's correlation analysis for examining the results from the TPC, DPPH and ABTS•+ assays was performed. A value of P<0.05 was considered to indicate a statistically significant difference. In addition, one-way ANOVA was applied, followed by Dunnett's tests for multiple pair wise comparisons using SPSS software (SPSS, Inc., Chicago, IL, USA).

Results and Discussion

Preliminary analysis by our research group on different EVOOs demonstrated that EVOO originating from the olive tree variety grown in Mount Athos is rich in biophenols (data not shown). Thus, this EVOO was used for the isolation of the TPF extract, as well as that of pure HT. TPF extract was also characterized chemically by determining the amount of HT, T, OLEO and OLEA biophenols. Moreover, both TPF and HT were examined for their antioxidant activity in vitro and at a cellular level in endothelial cells and myoblasts.

The recovery of the TPF was performed by applying a recently developed CPE-based extraction process which included the use of the biphasic system n-hexane/EVOO/etOH/water-3/2/3/2 v/v/v/v and a multiple dual-mode CPE method for the treatment of OO (27,35). Compared to other purposed OO extraction methods (44,45), this green process allows the treatment of large volumes of OO in a continuous extraction procedure and with extremely low solvent consumption. Thus, 6 extraction-recovery cycles were performed by pumping per each cycle 2.5 liters of feed oil phase (at 60 ml/min in ascending mode) and replacing the concentrated in biophenols stationary phase with 300 ml of fresh aqueous extraction phase (see experimental part). It is important to note that the treatment by this process of 6 liters of OO (corresponding to 5,478 g) resulted the recovery of 6.35 g of TPF in only 5 h. However, due to the high complexity of TPF the first step of the separation process included a step-gradient CPC fractionation with a series of four biphasic systems consisting of the solvents n-hexane, ethyl acetate, etOH and water in proportions 4/1/2/3 (S1), 3/2/2/3 (S2), 2/3/2/3 (S3) and 1/4/2/3 v/v/v/v (S4) (27,35). Even if this method afforded approximately HT in 87% purity, an additional purification step was required and therefore size exclusion column chromatography was used. The combination of these two chromatographic techniques has the advantage of using different criteria of selectivity between two purification steps and has been successfully applied for the recovery of pure compounds from complex natural extracts (46). The result of this two-step purification procedure was the effective recovery of high purity (>97%) HT. The purity of the recovered compounds was firstly examined by proton nuclear magnetic resonance (1H-NMR) and was then evaluated by HPLC-DAD analysis.

In addition, since the compounds under investigation behave differently in the presence or absence of acid two different elution systems have been used. Specifically, for the analysis of the TPF constituents, such as the phenyl alcohols (HT and T), the mobile phase of the used HPLC method (method A, see Materials and methods) contained acidified solvent (water acidified with 0.2% acetic acid) in order to increase the quality of the analysis. On the other hand, OLEA and OLEO are labile components and they are not stable in acidic conditions. Thus, the mobile phase of the HPLC method (method B, see Materials and methods) used for the analysis of those compounds was composed of water (solvent A) and acetonitrile (solvent B) without acidification. The profiling of TPF as resulting from HPLC-DAD chromatograms is depicted in Fig. 1. The results of the quantitative analysis are expressed in mg/g TPF. Specifically, TPF extract contained 19.4 mg/g HT, 24.4 mg/g T, 56.3 mg/g OLEA and 135.5 mg/ml OLEO.

As indicated by the quantitative analysis, the TPF was rich in biophenols. Specifically, the TPC as measured by Folin-Ciocalteau assay was 484 µg of biophenols per mg of TPF extract. According to a comparative study in which 221 EVOOs were extracted from 4 olive mono-cultivars (Koroneiki, Tsounati, Adramitini, and Throubolia) originating from 4 divisions of Greece (47), the range of TPC was between 30–351 mg/kg EVOOs. Of note, according to a health claim on OO polyphenols approved by the European Food Safety Authority (EFSA; Commission Regulation EU 432/2012), OOs are considered to protect from oxidative stress-induced lipid peroxidation in blood, when they contain about 5 mg of HT and its derivatives (e.g., oleuropein complex and T) per 20 g of OO. Based on the content of TPF in biophenols, OO in the present study, contain 4.15 mg of HT and its derivatives per 20 g.

The chemical analysis of TPF indicated that the T levels were high (28.28 mg/kg TPF) and close to those of HT (22.48 mg/kg TPF), according to a comparative study, where the T levels fluctuated between 1.97–16.2 mg/kg OO (47). Additionally, OLEO exhibited the highest concentration which was >2-fold higher than that of OLEA.

Following the determination of the biophenolic content, the antioxidant activities of TPF and HT were evaluated by DPPH and ABTS•+ assays (Fig. 2). In order to examine the antioxidant potency of the biophenols contained in the tested compounds, the specific activity was determined. Specific activity is a unit that compares extracts with varying composition. Since HT was a pure polyphenol, for the calculation of the specific activity, its polyphenolic content was considered as 100%. According to DPPH assay, HT exhibited a significantly (P<0.05) higher antioxidant activity compared with TPF. by contrast, in ABTS•+ assay, the antioxidant activity of HT was significantly (P<0.05) lower than that of TPF. These differences between the DPPH and ABTS•+ assays may be ascribed to the different solvents used in each assay (48). In particular, in DPPH assay the solvent is MeOH while in ABTS•+ assay the solvent is water (49). Namely, the bioactive compounds of TPF may be more polar and so they were dissolved more in water and less in MeOH than HT.

The antioxidant activity of the tested compounds was also examined in C2C12 myoblasts and EA.hy926 endothelial cells. In order to use non-cytotoxic concentrations of the tested compounds for these experiments, their cytotoxicity was assessed by XTT assay. The results from XTT assay revealed that TPF exhibited cytotoxicity at concentrations >24.0 µg/ml in both the C2C12 and EA.hy926 cells (Fig. 3A and B). HT exerted cytotoxic effects at concentrations >30.0 µg/ml in the C2C12 cells and >2.5 µg/ml in the EA.hy926 cells (Fig. 3C and D). Thus, the range of concentrations used for assessing the antioxidant activity of the tested compounds were 2.5–10.0 µg/ml for TPF in both the C2C12 and EA.hy926 cells, and 10–25 and 0.25–1.0 µg/ml for HT in the C2C12 and EA.hy926 cells, respectively.

The results from flow cytometric analysis revealed that treatment of the C2C12 cells with TPF significantly increased the GSH levels by 26, 28, 37 and 23% at 2.5, 5, 7.5 and 10 µg/ml, respectively, compared with control (Fig. 4A). Treatment of the C2C12 cells with HT also significantly increased the GSH levels by 24, 23, 54 and 38% at 10, 15, 20 and 25 µg/ml, respectively (Fig. 4B). Moreover, treatment of the EA.hy926 cells with TPF and HT significantly increased the GSH levels compared to the controls (Fig. 4C and D). Thus, the effects of TPF and HT on the GSH levels seemed to be cell type-independent. Particularly, TPF increased the GSH levels in the EA.hy926 cells by 23, 22, 38 and 20% at 2.5, 5, 7.5 and 10 µg/ml, respectively, while HT by 34, 40, 15 and 22% at 0.25, 0.5, 0.75 and 1.0 µg/ml respectively, compared to controls (Fig. 4C and D). The HT-induced increase in GSH levels suggested that this polyphenol was one of the bioactive compounds in TPF accounting for its effects on the GSH levels.

HT has been shown to increase GSH synthesis in human retinal pigment epithelial cells by the activation of the nuclear factor (erythroid-derived-2)-like 2 (Nrf2), a transcription factor and a key regulator of the antioxidant defense system (Fig. 6) (5053). Nrf2 induces the expression of gamma-glutamylcysteine ligase (GCL), which has the ability to regulate the synthesis of GSH (54). Moreover, HT seems to activate the NK-p62/SQSTM1 pathway, required for Nrf2-induced GSH synthesis (42,50). Manzanares et al (55) indicated that after a high EVOO diet, the activation of Nrf2 was observed in rat livers and mammary glands.

Moreover, OO biophenols, from olive mill waste water have been shown to increase GSH concentrations in human blood (48,56). This increase in GSH levels may be due to the upregulation of GCL and GSH synthetase, mediated by the antioxidant response element (ARE; a target of Nrf2) (Fig. 6) (56). Covas et al indicated that an EVOO rich in biophenols (366 mg/kg) beneficially modulated the balance between GSH and oxidized glutathione (GSSG). Thus, EVOO biophenols exert their antioxidant effects as a part of the induction of Nrf2 (57).

It is also worth mentioning that at the highest concentration, both TPF and HT exhibited less potency for increasing the GSH levels than at lower concentrations. This observed decrease in the potency of TPF and HT at the highest concentrations may be explained by a potential pro-oxidant activity of polyphenols after reaching a 'crucial' concentration (43,5861). A potential pro-oxidant activity was also supported by the cytotoxicity exerted by TPF and HT after these high concentrations.

Oleuropein and HT have been reported to exert pro-oxidant effects, due to their iron- and copper-reducing activities. These reduced metals, in turn, can catalyze the production of OH radicals by the Fenton reaction. The ability of dietary polyphenols to act as antioxidants/prooxidants under in vitro and in vivo systems is dependent on a number of factors such their concentration and structure (62).

The observed TPF- and HT-induced increase in GSH levels in C2C12 myoblasts and EA.hy926 endothelial cells is of importance. GSH is a conserved molecule among many species, reflecting its crucial biological role. In particular, it has been established that the thiol moiety of GSH is important for the direct scavenging of radical species (63). Thus, a decrease at GSH levels contributes to oxidative stress associated with aging and many pathological conditions, such as neurodegenerative diseases, inflammation, and infections (63).

Unlike GSH, the ROS levels were not significantly affected by the two test compounds compared to the controls in neither cell line (Fig. 5). It should be mentioned that the measured ROS levels corresponded to the naturally occurring levels in these cells; that is, there was not any treatment of cells with oxidizing agents before the addition of TPF and HT. Previous studies have also reported that changes in oxidative stress levels or antioxidant mechanisms are not always accompanied by changes in ROS levels (42,43,64).

In conclusion, EVOO from a Greek endemic olive tree variety, Olea europea, was used for the isolation of TPF, as well as that of pure HT. For the extraction process, it was used a recently developed green CPE-based method that allows the treatment of large volumes of OO in a continuous extraction procedure and with extremely low solvent consumption. For the isolation of pure HT, a combination of two chromatographic techniques was used, which has the advantage of using different criteria of selectivity between two purification steps and thus it is effective for the recovery of pure compounds from complex natural extracts. As indicated from the quantitative analysis, the TPF extract was rich in biophenols, such as T, HT, OLEO and OLEA. Finally, TPF and HT exhibited in vitro potent free radical scavenging activity, while they increased (at low concentrations), the levels of GSH, one of the most important antioxidant molecules, in both endothelial cells and myoblasts.

Abbreviations:

ROS

reactive oxygen species

OOs

olive oils

H2O2

hydrogen peroxide

FA

fatty acid

MUFA/SFA

monounsaturated/saturated fatty acid ratio

GSH

glutathione

HT

hydroxytyrosol

EVOO

extra virgin olive oil

TPF

total polyphenolic fraction

T

tyrosol

OLEO

oleocanthal

OLEA

oleacein

DMEM

Dulbecco's modified Eagle's medium

FBS

fetal bovine serum

TLC

thin layer chromatography

CPC

centrifugal partition chromatography

etOH

ethanol

MeOH

methanol

ISTD

internal standard

RSC

radical scavenging capacity

1H-NMR

proton nuclear magnetic resonance

OD

optical density

Nrf2

nuclear factor (erythroid-derived-2)-like 2

Acknowledgments

The present study was funded in part by a grant (Demetrios Kouretas) from the 'Tsantali Wineries'.

References

1 

Schieber M and Chandel NS: ROS function in redox signaling and oxidative stress. Curr Biol. 24:R453–R462. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Pham-Huy LA, He H and Pham-Huy C: Free radicals, antioxidants in disease and health. Int J Biomed Sci. 4:89–96. 2008.PubMed/NCBI

3 

Landete JM: Dietary intake of natural antioxidants: Vitamins and polyphenols. Crit Rev Food Sci Nutr. 53:706–721. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Fang YZ, Yang S and Wu G: Free radicals, antioxidants, and nutrition. Nutrition. 18:872–879. 2002. View Article : Google Scholar : PubMed/NCBI

5 

Landete JM: Updated knowledge about polyphenols: functions, bioavailability, metabolism, and health. Crit Rev Food Sci Nutr. 52:936–948. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Li HY, Yang M, Li Z and Meng Z: Curcumin inhibits angio-tensin II-induced inflammation and proliferation of rat vascular smooth muscle cells by elevating PPAR-γ activity and reducing oxidative stress. Int J Mol Med. 39:1307–1316. 2017.PubMed/NCBI

7 

Kerasioti E, Stagos D, Georgatzi V, Bregou E, Priftis A, Kafantaris I and Kouretas D: Antioxidant effects of sheep whey protein on endothelial cells. Oxid Med Cell Longev. 2016:65857372016. View Article : Google Scholar : PubMed/NCBI

8 

Deanfield JE, Halcox JP and Rabelink TJ: Endothelial function and dysfunction: Testing and clinical relevance. Circulation. 115:1285–1295. 2007.PubMed/NCBI

9 

Woywodt A, Bahlmann FH, De Groot K, Haller H and Haubitz M: Circulating endothelial cells: Life, death, detachment and repair of the endothelial cell layer. Nephrol Dial Transplant. 17:1728–1730. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Kokura S, Wolf RE, Yoshikawa T, Granger DN and Aw TY: Molecular mechanisms of neutrophil-endothelial cell adhesion induced by redox imbalance. Circ Res. 84:516–524. 1999. View Article : Google Scholar : PubMed/NCBI

11 

Zou Y, Yoon S, Jung KJ, Kim CH, Son TG, Kim MS, Kim YJ, Lee J, Yu BP and Chung HY: Upregulation of aortic adhesion molecules during aging. J Gerontol A Biol Sci Med Sci. 61:232–244. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Spanou C, Veskoukis AS, Kerasioti T, Kontou M, Angelis A, Aligiannis N, Skaltsounis AL and Kouretas D: Flavonoid glycosides isolated from unique legume plant extracts as novel inhibitors of xanthine oxidase. PLoS One. 7:e322142012. View Article : Google Scholar : PubMed/NCBI

13 

Goutzourelas N, Stagos D, Spanidis Y, Liosi M, Apostolou A, Priftis A, Haroutounian S, Spandidos DA, Tsatsakis AM and Kouretas D: Polyphenolic composition of grape stem extracts affects antioxidant activity in endothelial and muscle cells. Mol Med Rep. 12:5846–5856. 2015.PubMed/NCBI

14 

Reid MB: Free radicals and muscle fatigue: Of ROS, canaries, and the IOC. Free Radic Biol Med. 44:169–179. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Rafehi H, Smith AJ, Balcerczyk A, Ziemann M, Ooi J, Loveridge SJ, Baker EK, El-Osta A and Karagiannis TC: Investigation into the biological properties of the olive poly-phenol, hydroxytyrosol: Mechanistic insights by genome-wide mRNA-Seq analysis. Genes Nutr. 7:343–355. 2012. View Article : Google Scholar

16 

Montaño A, Hernández M, Garrido I, Llerena JL and Espinosa F: Fatty acid and phenolic compound concentrations in eight different monovarietal virgin olive oils from extremadura and the relationship with oxidative stability. Int J Mol Sci. 17:172016. View Article : Google Scholar

17 

Local Food-Nutraceuticals Consortium: Understanding local Mediterranean diets: a multidisciplinary pharmacological and ethnobotanical approach. Pharmacol Res. 52:353–366. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Trichopoulou A, Costacou T, Bamia C and Trichopoulos D: Adherence to a Mediterranean diet and survival in a Greek population. N Engl J Med. 348:2599–2608. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Trichopoulou A, Lagiou P, Kuper H and Trichopoulos D: Cancer and Mediterranean dietary traditions. Cancer Epidemiol Biomarkers Prev. 9:869–873. 2000.PubMed/NCBI

20 

Brinkman MT, Buntinx F, Kellen E, Van Dongen MC, Dagnelie PC, Muls E and Zeegers MP: Consumption of animal products, olive oil and dietary fat and results from the Belgian case-control study on bladder cancer risk. Eur J Cancer. 47:436–442. 2011. View Article : Google Scholar

21 

Oliveras-Ferraros C, Fernández-Arroyo S, Vazquez-Martin A, Lozano-Sánchez J, Cufí S, Joven J, Micol V, Fernández-Gutiérrez A, Segura-Carretero A and Menendez JA: Crude phenolic extracts from extra virgin olive oil circumvent de novo breast cancer resistance to HER1/HER2-targeting drugs by inducing GADD45-sensed cellular stress, G2/M arrest and hyperacetylation of Histone H3. Int J Oncol. 38:1533–1547. 2011.PubMed/NCBI

22 

Tunca B, Tezcan G, Cecener G, Egeli U, Ak S, Malyer H, Tumen G and Bilir A: Olea europaea leaf extract alters microRNA expression in human glioblastoma cells. J Cancer Res Clin Oncol. 138:1831–1844. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Bouallagui Z, Han J, Isoda H and Sayadi S: Hydroxytyrosol rich extract from olive leaves modulates cell cycle progression in MCF-7 human breast cancer cells. Food Chem Toxicol. 49:179–184. 2011. View Article : Google Scholar

24 

Cárdeno A, Sánchez-Hidalgo M, Cortes-Delgado A and Alarcón de la Lastra C: Mechanisms involved in the antiproliferative and proapoptotic effects of unsaponifiable fraction of extra virgin olive oil on HT-29 cancer cells. Nutr Cancer. 65:908–918. 2013. View Article : Google Scholar : PubMed/NCBI

25 

Acquaviva R, Di Giacomo C, Sorrenti V, Galvano F, Santangelo R, Cardile V, Gangia S, D'Orazio N, Abraham NG and Vanella L: Antiproliferative effect of oleuropein in prostate cell lines. Int J Oncol. 41:31–38. 2012.PubMed/NCBI

26 

Coccia A, Bastianelli D, Mosca L, Monticolo R, Panuccio I, Carbone A, Calogero A and Lendaro E: Extra virgin olive oil phenols suppress migration and invasion of T24 human bladder cancer cells through modulation of matrix metalloproteinase-2. Nutr Cancer. 66:946–954. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Coccia A, Mosca L, Puca R, Mangino G, Rossi A and Lendaro E: Extra-virgin olive oil phenols block cell cycle progression and modulate chemotherapeutic toxicity in bladder cancer cells. Oncol Rep. 36:3095–3104. 2016.PubMed/NCBI

28 

Miró-Casas E, Covas MI, Fitó M, Farré-Albadalejo M, Marrugat J and de la Torre R: Tyrosol and hydroxytyrosol are absorbed from moderate and sustained doses of virgin olive oil in humans. Eur J Clin Nutr. 57:186–190. 2003. View Article : Google Scholar : PubMed/NCBI

29 

Keys A, Menotti A, Karvonen MJ, Aravanis C, Blackburn H, Buzina R, Djordjevic BS, Dontas AS, Fidanza F, Keys MH, et al: The diet and 15-year death rate in the seven countries study. Am J Epidemiol. 124:903–915. 1986. View Article : Google Scholar : PubMed/NCBI

30 

Obied HK, Prenzler PD, Omar SH, Ismael R, Servili M, Esposto S, Taticchi A, Selvaggini R and Urbani S: Pharmacology of Olive Biophenols. Advances in Molecular Toxicology. Fishbein JC and Heilman JM: 6. Elsevier; Amsterdam: pp. 195–242. 2012, View Article : Google Scholar

31 

Lewandowska U, Szewczyk K, Hrabec E, Janecka A and Gorlach S: Overview of metabolism and bioavailability enhancement of polyphenols. J Agric Food Chem. 61:12183–12199. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Schaffer S, Podstawa M, Visioli F, Bogani P, Müller WE and Eckert GP: Hydroxytyrosol-rich olive mill wastewater extract protects brain cells in vitro and ex vivo. J Agric Food Chem. 55:5043–5049. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Rietjens SJ, Bast A and Haenen GR: New insights into controversies on the antioxidant potential of the olive oil antioxidant hydroxytyrosol. J Agric Food Chem. 55:7609–7614. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Rodríguez-Morató J, Boronat A, Kotronoulas A, Pujadas M, Pastor A, Olesti E, Pérez-Mañá C, Khymenets O, Fitó M, Farré M, et al: Metabolic disposition and biological significance of simple phenols of dietary origin: Hydroxytyrosol and tyrosol. Drug Metab Rev. 48:218–236. 2016. View Article : Google Scholar : PubMed/NCBI

35 

Angelis A, Hamzaoui M, Aligiannis N, Nikou T, Michailidis D, Gerolimatos P, Termentzi A, Hubert J, Halabalaki M, Renault JH, et al: An integrated process for the recovery of high added-value compounds from olive oil using solid support free liquid-liquid extraction and chromatography techniques. J Chromatogr A. 1491:126–136. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Mastralexi A, Nenadis N and Tsimidou MZ: Addressing analytical requirements to support health claims on 'olive oil polyphenols' (EC Regulation 432/2012). J Agric Food Chem. 62:2459–2461. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Impellizzeri J and Lin J: A simple high-performance liquid chromatography method for the determination of throat-burning oleocanthal with probated antiinflammatory activity in extra virgin olive oils. J Agric Food Chem. 54:3204–3208. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Vougogiannopoulou K, Lemus C, Halabalaki M, Pergola C, Werz O, Smith AB III, Michel S, Skaltsounis L and Deguin B: One-step semisynthesis of oleacein and the determination as a 5-lipoxygenase inhibitor. J Nat Prod. 77:441–445. 2014. View Article : Google Scholar : PubMed/NCBI

39 

Singleton VL, Orthofer R and Lamuela-Raventos RM: Analysis of total phenols and other oxidation substrates and antioxidants by means of Folin-Ciocalteu reagent. Methods Enzymol. 299:152–178. 1998. View Article : Google Scholar

40 

Brand-Williams W, Cuvelier ME and Berset C: Use of a free radical method to evaluate antioxidant activity. Lebensm Wiss Technol. 28:25–30. 1995. View Article : Google Scholar

41 

Cano A, Hernandez-Ruiz J, García-Cánovas F, Acosta M and Arnao MB: An end-point method for estimation of the total antioxidant activity in plant material. Phytochem Anal. 9:196–202. 1998. View Article : Google Scholar

42 

Goutzourelas N, Stagos D, Demertzis N, Mavridou P, Karterolioti H, Georgadakis S, Kerasioti E, Aligiannis N, Skaltsounis L, Statiri A, et al: Effects of polyphenolic grape extract on the oxidative status of muscle and endothelial cells. Hum Exp Toxicol. 33:1099–1112. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Priftis A, Stagos D, Konstantinopoulos K, Tsitsimpikou C, Spandidos DA, Tsatsakis AM, Tzatzarakis MN and Kouretas D: Comparison of antioxidant activity between green and roasted coffee beans using molecular methods. Mol Med Rep. 12:7293–7302. 2015.PubMed/NCBI

44 

Vulcano I, Halabalaki M, Skaltsounis L and Ganzera M: Quantitative analysis of pungent and anti-inflammatory phenolic compounds in olive oil by capillary electrophoresis. Food Chem. 169:381–386. 2015. View Article : Google Scholar

45 

Keiler AM, Zierau O, Bernhardt R, Scharnweber D, Lemonakis N, Termetzi A, Skaltsounis L, Vollmer G and Halabalaki M: Impact of a functionalized olive oil extract on the uterus and the bone in a model of postmenopausal osteoporosis. Eur J Nutr. 53:1073–1081. 2014. View Article : Google Scholar

46 

Angelis A, Hubert J, Aligiannis N, Michalea R, Abedini A, Nuzillard JM, Gangloff SC, Skaltsounis AL and Renault JH: Bio-guided isolation of methanol-soluble metabolites of common spruce (Picea abies) bark by-products and investigation of their dermo-cosmetic properties. Molecules. 21:212016. View Article : Google Scholar

47 

Agiomyrgianaki A, Petrakis PV and Dais P: Influence of harvest year, cultivar and geographical origin on Greek extra virgin olive oils composition: A study by NMR spectroscopy and biometric analysis. Food Chem. 135:2561–2568. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Pliszka B, Huszcza-Ciołkowska G and Wierzbicka E: Effects of solvents and extraction methods on the content and antiradical activity of polyphenols from fruits Actinidia arguta, Crataegus monogyna, Gaultheria procumbens and Schisandra chinensis. Acta Sci Pol Technol Aliment. 15:57–63. 2016. View Article : Google Scholar

49 

Schlesier K, Harwat M, Böhm V and Bitsch R: Assessment of antioxidant activity by using different in vitro methods. Free Radic Res. 36:177–187. 2002. View Article : Google Scholar : PubMed/NCBI

50 

Zou X, Feng Z, Li Y, Wang Y, Wertz K, Weber P, Fu Y and Liu J: Stimulation of GSH synthesis to prevent oxidative stress-induced apoptosis by hydroxytyrosol in human retinal pigment epithelial cells: Activation of Nrf2 and JNK-62/SQSTM1 pathways. J Nutr Biochem. 23:994–1006. 2012. View Article : Google Scholar

51 

Moi P, Chan K, Asunis I, Cao A and Kan YW: Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region. Proc Natl Acad Sci USA. 91:9926–9930. 1994. View Article : Google Scholar : PubMed/NCBI

52 

Li W and Kong AN: Molecular mechanisms of Nrf2-mediated antioxidant response. Mol Carcinog. 48:91–104. 2009. View Article : Google Scholar :

53 

Nguyen T, Nioi P and Pickett CB: The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J Biol Chem. 284:13291–13295. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Kobayashi M and Yamamoto M: Nrf2-Keap1 regulation of cellular defense mechanisms against electrophiles and reactive oxygen species. Adv Enzyme Regul. 46:113–140. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Manzanares MA, Solanas M, Moral R, Escrich R, Vela E, Costa I and Escrich E: Dietary extra-virgin olive oil and corn oil differentially modulate the mRNA expression of xeno-biotic-metabolizing enzymes in the liver and in the mammary gland in a rat chemically induced breast cancer model. Eur J Cancer Prev. 24:215–222. 2015. View Article : Google Scholar

56 

Visioli F, Wolfram R, Richard D, Abdullah MI and Crea R: Olive phenolics increase glutathione levels in healthy volunteers. J Agric Food Chem. 57:1793–1796. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Covas MI, Nyyssönen K, Poulsen HE, Kaikkonen J, Zunft HJ, Kiesewetter H, Gaddi A, de la Torre R, Mursu J, Bäumler H, et al EUROLIVE Study Group: The effect of polyphenols in olive oil on heart disease risk factors: A randomized trial. Ann Intern Med. 145:333–341. 2006. View Article : Google Scholar : PubMed/NCBI

58 

Procházková D, Boušová I and Wilhelmová N: Antioxidant and prooxidant properties of flavonoids. Fitoterapia. 82:513–523. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Lambert JD and Elias RJ: The antioxidant and pro-oxidant activities of green tea polyphenols: A role in cancer prevention. Arch Biochem Biophys. 501:65–72. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Fukumoto LR and Mazza G: Assessing antioxidant and prooxidant activities of phenolic compounds. J Agric Food Chem. 48:3597–3604. 2000. View Article : Google Scholar : PubMed/NCBI

61 

Sakihama Y, Cohen MF, Grace SC and Yamasaki H: Plant phenolic antioxidant and prooxidant activities: Phenolics-induced oxidative damage mediated by metals in plants. Toxicology. 177:67–80. 2002. View Article : Google Scholar : PubMed/NCBI

62 

Maurya DK and Devasagayam TP: Antioxidant and prooxidant nature of hydroxycinnamic acid derivatives ferulic and caffeic acids. Food Chem Toxicol. 48:3369–3373. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Aquilano K, Baldelli S and Ciriolo MR: Glutathione: New roles in redox signaling for an old antioxidant. Front Pharmacol. 5:1962014. View Article : Google Scholar : PubMed/NCBI

64 

Kerasioti E, Stagos D, Priftis A, Aivazidis S, Tsatsakis AM, Hayes AW and Kouretas D: Antioxidant effects of whey protein on muscle C2C12 cells. Food Chem. 155:271–278. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2017
Volume 40 Issue 3

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
Kouka P, Priftis A, Stagos D, Angelis A, Stathopoulos P, Xinos N, Skaltsounis AL, Mamoulakis C, Tsatsakis AM, Spandidos DA, Spandidos DA, et al: Assessment of the antioxidant activity of an olive oil total polyphenolic fraction and hydroxytyrosol from a Greek Olea europea variety in endothelial cells and myoblasts. Int J Mol Med 40: 703-712, 2017
APA
Kouka, P., Priftis, A., Stagos, D., Angelis, A., Stathopoulos, P., Xinos, N. ... Kouretas, D. (2017). Assessment of the antioxidant activity of an olive oil total polyphenolic fraction and hydroxytyrosol from a Greek Olea europea variety in endothelial cells and myoblasts. International Journal of Molecular Medicine, 40, 703-712. https://doi.org/10.3892/ijmm.2017.3078
MLA
Kouka, P., Priftis, A., Stagos, D., Angelis, A., Stathopoulos, P., Xinos, N., Skaltsounis, A., Mamoulakis, C., Tsatsakis, A. M., Spandidos, D. A., Kouretas, D."Assessment of the antioxidant activity of an olive oil total polyphenolic fraction and hydroxytyrosol from a Greek Olea europea variety in endothelial cells and myoblasts". International Journal of Molecular Medicine 40.3 (2017): 703-712.
Chicago
Kouka, P., Priftis, A., Stagos, D., Angelis, A., Stathopoulos, P., Xinos, N., Skaltsounis, A., Mamoulakis, C., Tsatsakis, A. M., Spandidos, D. A., Kouretas, D."Assessment of the antioxidant activity of an olive oil total polyphenolic fraction and hydroxytyrosol from a Greek Olea europea variety in endothelial cells and myoblasts". International Journal of Molecular Medicine 40, no. 3 (2017): 703-712. https://doi.org/10.3892/ijmm.2017.3078