Detoxifying effect of fermented black ginseng on H2O2-induced oxidative stress in HepG2 cells

  • Authors:
    • Min-Ji Bak
    • Woo-Sik Jeong
    • Kyu-Bong Kim
  • View Affiliations

  • Published online on: October 16, 2014     https://doi.org/10.3892/ijmm.2014.1972
  • Pages: 1516-1522
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Fermented black ginseng (FBG) is prepared by repeated steaming and drying processes with fresh ginseng followed by fermentation with Saccharomyces cerevisiae. It has recently been shown to have several bioactivities. FBG contains crude saponin (1,440 µg/ml), ginsenoside Rg2 (2.86 µg/ml), ginsenoside Rg3 (24.52 µg/ml), ginsenoside Rh1 (12.64 µg/ml), ginsenoside Rh2 (0.63 µg/ml) and ginsenoside Rf (1.32 µg/ml). The present study investigated the antioxidant defense properties of FBG against hydrogen peroxide (H2O2)-mediated oxidative stress in HepG2 human hepatocellular carcinoma cells. The increased production of reactive oxygen species (ROS) induced by H2O2 was attenuated in a dose-dependent manner when the cells were pre-treated with FBG (10-50 µg/ml). FBG induced both the expression and activity of antioxidant enzymes, such as superoxide dismutase, catalase and glutathione peroxidase in the H2O2-treated HepG2 cells. The inhibitory effects of FBG on the phosphorylation of upstream mitogen-activated protein kinases (MAPKs), such as c-Jun N-terminal kinase, extracellular signal-regulated kinase and p38 were also observed. Overall, our results demonstrate that FBG protects HepG2 cells from oxidative stress through the induction of antioxidant enzyme activity and the inhibition of MAPK pathways.

Introduction

Reactive oxygen species (ROS), including superoxide anion radical (O2•−), hydroxyl radical (•OH) and hydrogen peroxide (H2O2) have been implicated in the development of a variety of diseases, such as carcinogenesis, inflammation, aging and atherosclerosis. The accumulation of ROS induces lipid peroxidation, the inactivation of proteins and DNA damage in cells (1,2). Protective enzymatic and non-enzymatic antioxidant defense mechanisms reduce oxidative stress by scavenging ROS. To protect themselves against toxic free radicals and ROS, cells have developed a variety of antioxidant defenses (3). These include antioxidant enzymes, such as superoxide dismutase (SOD), which catalyzes the dismutation of superoxide anions to hydrogen peroxide; catalase (CAT), which converts H2O2 into molecular oxygen and water; glutathione peroxidase (GPx), which catalyzes the degradation of H2O2 and hydroperoxides. On the non-enzymatic level, certain vitamins and other antioxidant compounds scavenge free radicals and delay the oxidation of molecules (4,5). Phytochemicals provide further protection against oxidative damage from free radicals. A large number of studies have indicated that phytochemicals present in fruits, vegetables and herbs, exert their antioxidant effects against oxidative stress through the induction or activation of these endogenous antioxidant enzymes (69). In addition, increased ROS production not only directly damages cells by oxidizing macromolecules, such as DNA and lipids, but also indirectly by triggering mitogen-activated protein kinase (MAPK) signaling pathways (10). MAPKs, such as extracellular signal-regulated kinase (ERK), p38 and c-Jun N-terminal kinase (JNK) are involved in crucial signaling pathways in cell proliferation, differentiation and cell death in response to various signals produced by growth factors, hormones and cytokines, as well as genotoxic and oxidative stressors (11). Studies have demonstrated that MAPK signaling pathways can also be modulated by the production of ROS and antioxidant enzymatic activity and expression (12,13).

Ginseng (Panax ginseng C.A. Meyer) is considered as one of the most popular medicinal herbs, and has well known pharmacological activities which include anticancer (14), anti-aging (15), anti-diabetic (16), anti-stress (17) and neuroprotective effects (18). Black ginseng is a processed ginseng produced by a nine-time steaming at approximately 85°C and a nine-time drying process (repetitive steaming and drying) using fresh ginseng, at which point the ginseng becomes black in color (19). Fermented black ginseng (FBG) is processed further by incubating black ginseng with Saccharomyces cerevisiae for 24 h to produce more active ginsenosides. Black ginseng has been shown to enhance biological activities possibly due to the enrichment of the bioactive chemical constituents during the heat and drying processing stage (20). However, to the best of our knowledge, studies on the antioxidant properties and the underlying molecular mechanisms of FBG are limited. In the present study, we investigated the antioxidant defense properties of FBG at the enzymatic and cellular levels and its ability to inhibit ROS production; we demonstrate that FBG induces both the activity and the expression of antioxidant enzymes and modulates upstream protein kinases, including MAPKs.

Materials and methods

Reagents

FBG was obtained from Ginseng-By-Pharm Co. (Wonju, Korea). The main composition of FBG was determined using the liquid chromatography-mass spectrometry (LC/MS) method (Joongbu University, Geumsan, Korea) and is shown in Table I. KCN (potassium cyanide, 60178), H2O2, sulfanilamide (S9251), N-(1-naphthyl)ethylenediamine dihydrochloride (P7626), nitroblue tetrazolium salt, xanthine, copper chloride, glutathione, xanthine oxidase from bovine milk (0.1–0.4 U/mg of protein), glutathione reductase from baker’s yeast (Saccharomyces cerevisiae; 100–300 U/mg of protein) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium, inner salt (MTT) were purchased from Sigma-Aldrich (St. Louis, MO, USA). 2′,7′-Dichlorohydrofluorescein diacetate (DCF-DA) was purchased from Molecular Probes, Inc. (Eugene, OR, USA). Antibodies against CAT (14097), phosphorylated ERK (9101), phosphorylated JNK (9251), phosphorylated p38 (9211) and horseradish peroxidase-conjugated anti-rabbit IgG (7074) were purchased from Cell Signaling Technology Inc. (Beverly, MA, USA). Antibodies against SOD (sc-18504), GPx (sc-30147), β-actin (sc-1616) and horseradish peroxidase-conjugated anti-goat IgG (sc-2350) were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). All other reagents were of the highest quality generally available.

Table I

Saponin and main ginsenosides found in fermented black ginseng.

Table I

Saponin and main ginsenosides found in fermented black ginseng.

GinsenosidesConcentration (μg/ml)
Crude saponin1,440
Ginsenoside Rg22.86
Ginsenoside Rg324.52
Ginsenoside Rh112.62
Ginsenoside Rh20.63
Ginsenoside Rf1.32
Cell culture

HepG2 human hepatocellular carcinoma cells were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). The cells were maintained in Dulbecco’s minimum essential medium with 10% fetal bovine serum (FBS), 100 U/ml penicillin, 100 μg/ml streptomycin (all from HyClone, Logan, UT, USA), 1% essential amino acids and 1% glutamax (both from Gibco, Grand Island, NY, USA) at 37°C in a humidifying incubator containing 5% CO2. For subculture, the HepG2 cells were harvested at 80–90% confluence.

Cell viability assay

MTT assay was used to measure cell viability. The HepG2 cells were seeded in 24-well plates at a rate of 1×105 cells/well. After 24 h of incubation, the cells were treated with distilled water (DW; control) or different doses of FBG for 24 h. Subsequently, 50 μl of 1 mg/ml MTT were added and the plates were incubated for 4 h. Following incubation at 37°C for 4 h, the MTT medium was removed by aspiration and 200 μl of dimethyl sulfoxide (DMSO) were added to each well. After reacting for 10 min at room temperature, formazan production was detected by the measurement of the optical density (OD) at 570 nm using a PowerWave XS microplate reader (BioTek Instruments, Winooski, VA, USA). The data are expressed as the percentage cell viability compared to the vehicle-treated control.

ROS formation assay

Intracellular ROS levels were determined by the DCF-DA assay. Briefly, the cells were seeded in 96-well dark plate at a rate of 1×104 cells/well and pre-incubated with 20 μM DCF-DA (dissolved in DMSO) for 1 h at 37°C in the dark. After washing out the excess probe using 1× ice-cold phosphate-buffered saline (PBS), the cells were treated with the vehicle (DW for control), vitamin C (positive control), or FBG in the presence or absence of 1 mM H2O2 for 12 h and then washed twice with 1× ice-cold PBS. Fluorescence was detected by excitation at 485 nm and emission at 535 nm using a fluorescence multi-detection reader (BioTek Instruments). Vitamin C (100 μg/ml) was used as positive control for current study. Vitamin C is well known as a strong antioxidant and is widely used as a positive control in antioxidant studies.

Assays for antioxidant enzymes

The cells were seeded in 6-well plates at a rate of 2×105 cells/well. After 24 h of incubation, the cells were treated with DW (control), vitamin C (positive control), or different doses of FBG for 24 h. For the CAT and GPx enzyme assays, the cells were homogenized with 1 ml of 50 mM potassium phosphate buffer (pH 7.0), and then centrifuged at 12,000 rpm for 20 min at 4°C. For the manganese-superoxide dismutase (Mn-SOD) assay, the cells were homogenized with 1 ml of 65 mM phosphate buffer (pH 7.8), and then centrifuged at 12,000 rpm for 20 min at 4°C. The cell lysis supernatant was analyzed to determine CAT and GPx activity, while the cell pellet was used to detect Mn-SOD activity. The protein concentration was determined using the BCA protein assay (Pierce Biotechnology, Rockford, IL, USA) according to the manufacturer’s instructions. The results are expressed as the enzyme activity per milligram protein compared with the corresponding control cultures.

Mn-SOD activity

The activity of Mn-SOD was measured according to the method described in the study by Oyanagui (21). The remaining pellet (i.e., the mitochondrial fraction) was dissolved in 0.1% Triton X-100 and used for the determination of Mn-SOD activity. For the Mn-SOD activity assay, 60 μl of 4 mM of KCN solution were added to the assay mixture to inhibit copper- and zinc-containing superoxide dismutase (Cu/Zn-SOD). The samples were pre-incubated with 15 μl of 75 mM Na-xanthine and 15 μl of 10 mM hydroxylamine hydrochloride at 37°C for 10 min. Subsequently, 0.1 units of xanthine oxidase were added and the samples were incubated at 37°C for an additional 20 min. The reaction was terminated by the addition of 1% sulphanilamide and 0.02% ethylenediamine dihydrochloride. After standing at room temperature for 20 min, the absorbance of the final mixture was measured at 450 nm. Enzyme activity was expressed as units per milligram protein.

CAT activity

The activity of CAT was measured as previously described (22). The reaction mixture contained 12 μl of 3% (vol/vol) H2O2 and 20 μg of cell lysates in 50 mM potassium phosphate buffer (pH 7.0) at a final volume of 1.0 ml. The samples were incubated for 5 min at 37°C and the absorbance of the samples was monitored for 5 min at 240 nm. The change in absorbance is proportional to the breakdown of H2O2.

GPx activity

The activity of GPx was measured by as previously described (23). A total of 20 μg of supernatant, containing cytosolic fraction, was incubated with 1 mM EDTA, 1 mM sodium azide (NaN3), 5 mM GSH, 1 mM NADPH and 1 unit glutathione reductase at room temperature for 5 min. The reaction was initiated by the addition of 25 μl of 2.5 mM H2O2. GPx activity was measured as the rate of NADPH oxidation at 340 nm.

Western blot analysis

The cells were pre-treated with the vehicle (DW for control), vitamin C (positive control), or FBG (10, 25 or 50 μg/ml) for 1 h and then challenged with H2O2 for another 1 h. Subsequently, the cells were washed 3 times with ice-cold PBS (pH 7.4) and harvested with 200 μl of whole cell lysis buffer (pH 7.4) containing 10 mM Tris-HCl, 50 mM sodium chloride, 30 mM sodium pyrophosphate, 50 mM sodium fluoride, 100 μM sodium orthovanadate, 2 mM iodoacetic acid, 5 mM ZnCl2, 1 mM phenylmethylsulfonyl fluoride and 0.5% Triton X-100. The cell lysates were vigorously vortexed, homogenized in a sonicator for 10 sec and left on ice for 1 h. The homogenates were centrifuged at 13,000 × g for 10 min at 4°C. The supernatants were collected and equal amounts of total protein, as determined by BCA protein assay (Pierce Biotechnology), were mixed with 2× loading buffer and heated at 95°C for 5 min. An equal amount (30 μg) of protein from each cell lysate was separated by 12% SDS-polyacrylamide gel electrophoresis and transferred onto polyvinylidene difluoride (PVDF) membranes (Millipore, Billerica, MA, USA). The membranes were blocked with 5% non-fat dry milk in 1× PBST buffer (0.1% Tween-20 in PBS) for 1 h at room temperature and then incubated overnight with the appropriate primary antibody. Following hybridization with the primary antibody, the membranes were washed 3 times with PBST, and then incubated with anti-rabbit and anti-goat antibodies with horseradish peroxidase for 1 h at room temperature and washed with PBST 3 times. Final detection was performed with enhanced chemiluminescence (ECL™) western blotting reagents (Santa Cruz Biotechnology, Inc.).

Statistical analysis

The data are expressed as the means ± standard deviation (SD) values using Microsoft Excel. The values were compared with those of the control using analysis of variance, followed by Bonferroni’s test (GraphPad Prism software version 5.01; GraphPad Software, San Diego, CA, USA). The significance level was defined at P-values <0.05.

Results

Effects of FBG on cell viability

The cytotoxic effects of FBG on the HepG2 cells were evaluated by MTT assay. The percentage of viable cells was determined by MTT assay and compared to that of the control cells. The cells were treated with FBG at the concentration range of 0–200 μg/ml for 24 h. Treatment with 10–200 μg/ml FBG inhibited cell viability in a dose-dependent manner (Fig. 1). The survival rate of the cells treated with 50 μg/ml of FBG was approximately 70% compared to that of the control cells. Based on the cell viability data, subsequent experiments were performed using concentrations of FBG below 50 μg/ml of FBG.

Effects of FBG on ROS production

We examined whether FBG exerts inhibitory effects on the production of ROS using DCF-DA in H2O2-treated HepG2 cells. When the cells were treated with 1 mM H2O2, a >2.1-fold increase in the generation of ROS compared to the vehicle-treated controls was observed. Pre-treatment with FBG decreased the H2O2-mediated production of ROS in a dose-dependent manner and vitamin C (100 μg/ml) also markedly decreased ROS formation in the presence and/or absence of H2O2 in the HepG2 cells (Fig. 2). The dose-dependent effects of FBG were observed even in the absence of H2O2. These results suggest that FBG acts as an antioxidant which can directly scavenge excessive ROS generation in cells.

Effects of FBG on the activity of antioxidant enzymes

In order to investigate whether the antioxidant activity of FBG is mediated by its ability to increase the activity of cellular antioxidant enzymes, we measured the activity of antioxidant enzymes, including CAT, GPx and Mn-SOD in the H2O2-treated HepG2 cells. When the cells were treated with H2O2 (1 mM) alone, the activity of CAT, GPx and SOD-2 significantly decreased below the basal level of the vehicle-treated controls (Fig. 3). However, treatment with FBG increased the activity of these enzymes; the increased activity levels of CAT and GPx following treatment with 50 μg/ml FBG were much higher than the basal control (DW) levels and were even higher than the levels observed following treatment with vitamin C (positive control; 100 μg/ml), a well known antioxidant.

Effects of FBG on the expression of antioxidant enzymes

In addition to its stimulatory effects on enzyme activity, we determined the effects of FBG on the protein expression of these antioxidant enzymes in HepG2 cells. When the cells were treated with H2O2 alone, CAT, GPx and Mn-SOD protein expression levels were diminished compared with the vehicle-treated controls. However, treatment with FBG restored and upregulated the protein expression of these enzymes in a dose-dependent manner and vitamin C significantly induced the protein level of antioxidant enzymes compared to the H2O2-treated group (Fig. 4).

Effects of FBG on the phosphorylation of MAPKs

In order to determine whether antioxidant enzyme expression induced by FBG was associated with the MAPK pathway, we examined the phosphorylation levels of MAPK subfamilies, such as ERK, JNK and p38. The results revealed that H2O2 stimulated the phosphorylation of all MAPKs (Fig. 5). The H2O2-stimulated phosphorylation of MAPKs was decreased following treatment with FBG in a dose-dependent manner and vitamin C inhibited the phosphorylation levels of MAPKs in the presence of H2O2. This confirmed that protective effects of vitamin C against H2O2 by the induction of antioxidant enzymes through the inhibition of MAPK phosphorylation.

Discussion

The induction of antioxidant enzyme activity may be considered as a frontline defense strategy to protect human health against various oxidative stress-related diseases. Accordingly, numerous bioactive plant materials have been investigated for their antioxidant potential (4,68). In the present study, to the best of our knowledge, we demonstrate for the first time that FBG protects HepG2 cells against H2O2-induced oxidative stress through the regulation of ROS production and antioxidant enzymes, and signaling pathways including MAPKs.

ROS are known to play a central role in mediating various metabolic disorders related to several diseases. Thus, inhibiting ROS production and enhancing the scavenging ability of antioxidants may prove to be a useful strategy in the treatment of diseases related to oxidative stress (24). ROS-induced oxidative DNA damage has been implicated in mutagenesis and carcinogenesis and has attracted extensive attention in recent years. In addition, H2O2 is a major component of ROS produced intracellularly during a number of physiological and pathological processes, and causes oxidative damage (25,26). For this reason, H2O2 has often been used as an experimental model to investigate the mechanisms of cell injury induced by oxidative stress (2729). In the present study, when HepG2 cells pre-treated with FBG were challenged with H2O2, ROS formation decreased. A previous study demonstrated that Korean red ginseng extract exerted antioxidant and chemopreventive effects by decreasing ROS production in HepG2 cells treated with arachidonic acid and iron (30). These results suggest that ginsengs may have an antioxidant capacity by directly scavenging radicals. Maintaining the balance between free radicals (and/or ROS) and antioxidants is an essential part of biological homeostasis (31).

Antioxidant enzymes, including SOD, CAT and GPx are regarded as the firstline of the antioxidant defense system against ROS generation during oxidative stress. Recently, white ginseng has been reported to prevent oxidative stress by enhancing the intracellular activity of antioxidant enzymes and decreasing ROS formation (32). In addition, red ginseng has been reported to exhibit a variety of antioxidant and hepatoprotective effects on ethanol-induced oxidative injury in rat liver and TIB-73 cells (33,34). Jun and Chang (35), reported that red ginseng extract increased SOD, CAT and GPx activity after ICR male mice were γ-irradiated. Furthermore, a ginseng extract has been shown to induce hepatic SOD, CAT and GPx activity in Sprague-Dawley rats (36). Our results are in accordance with those of these studies, suggesting that FBG may prove useful against oxidate stress by reducing ROS levels and increasing antioxidant enzymes activity and expression. According to previous studies, a variety of ginsengs has shown antioxidant properties in maintaining cellular function against free radicals in vivo, as well as in vitro (4,7,37). In addition, the activity of intracellular antioxidant enzymes, such as SOD, CAT and GPx plays an important role in protecting cells against oxidative stress. Since the changes occurring in the activities of these enzymes can be considered a biomarker of antioxidant response under conditions of oxidative stress, the increased activity of these enzymes in FBG-treated HepG2 cells strongly suggests that FBG has antioxidant properties that function by simulating the activity of antioxidant enzymes in addition to directly scavenging ROS/free radicals. Lee et al (38), revealed that black ginseng has a protective effect on ethanol-induced teratogenesis through the augmentation of antioxidant activity in embryos.

Heat processing has been reported to increase the free radical scavenging activity of ginseng and stimulate the protective effects of ginseng against oxidative damage caused by oxidative stress (20,39). Black ginseng is known to contain different ginsenosides (Rg3, Rg4, Rg6, Rk3, Rs3 and Rs4) which are not present in white ginseng (40), and exhibits more potent pharmacological activities than white ginseng and red ginseng (41,42). Black ginseng is prepared by steaming at 85°C for 8 h and then drying until the water content decreases below 20%. This steaming and drying process is repeated 9 times. This process makes white ginseng black, and from this point on it is known as ‘black ginseng’. For the preparation of FBG, black ginseng is grinded and extracted with distilled water at 80°C for 72 h. Subsequently, this water extract is fermented with Saccharomyces cerevisiae at 35°C for 24 h.

The activity and expression of antioxidant enzymes may be modulated by upstream protein MAPKs, such as JNK, ERK and p38. The phosphorylation of these proteins has been shown to be mediated through H2O2-induced oxidative stress (11). Although FBG inhibits the phosphorylation of MAPK, as observed in this study, its effects on the downstream targets, such as antioxidant enzymes may be selective, at least in the current model system. Dong et al (30), indicated that red ginseng extract attenuates oxidative stress by reducing ROS formation through the LKB1-AMPK pathway in the HepG2 cell line, but not through the MAPK signaling pathway. It has been reported that the effects of dietary compounds on antioxidant enzyme expression and activity are mediated by the modification of several different signal transduction pathways (43). In addition, different agents can play one or more roles at different targets, and the cellular events may depend on the types and concentrations of the agents, as well as on the cell or tissue types. For example, Fan et al (44), revealed that ginseng pectin exerts protective effects against H2O2 through the ERK1/2 and Akt pathway in U87 neuronal cells. In addition, red ginseng and its primary ginsenosides inhibit ethanol-induced oxidative injury by reducing ROS production and lipid peroxidation through MAPK pathways in TIB-73 mouse hepatocytes (34). To the best of our knowledge, in this study, we report for the first time the modulatory role of FBG on upstream MAPKs. However, further studies are required using animals in order to determine the optimal dose, duration and method of administration.

In conclusion, FBG has the ability to protect cells against oxidative damage by scavenging ROS and inducing both the activity and the expression of cellular antioxidant enzymes possibly through the inhibition of MAPK signaling pathways. Therefore, FBG may be a potential natural agent for cellular defense, at least in liver cells. However, further in vivo studies using FBG are warranted.

Acknowledgements

This study was supported by the research fund of Dankook University in 2012.

References

1 

Ray PD, Huang BW and Tsuji Y: Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 24:981–990. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Valko M, Leibfritz D, Moncol J, Cronin MT, Mazur M and Telser J: Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol. 39:44–84. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Matés JM: Effects of antioxidant enzymes in the molecular control of reactive oxygen species toxicology. Toxicology. 153:83–104. 2000.PubMed/NCBI

4 

Bak MJ, Jun M and Jeong WS: Antioxidant and hepatoprotective effects of the red ginseng essential oil in H2O2-treated HepG2 cells and CCl4-treated mice. Int J Mol Sci. 13:2314–2330. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Schnabel D, Salas-Vidal E, Narváez V, et al: Expression and regulation of antioxidant enzymes in the developing limb support an function of ROS in interdigital cell death. Dev Biol. 291:291–299. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Naik SR and Panda VS: Antioxidant and hepatoprotective effects of Ginkgo biloba phytosomes in carbon tetrachloride-induced liver injury in rodents. Liver Int. 27:393–399. 2007.

7 

Jung CH, Seog HM, Choi IW, Choi HD and Cho HY: Effects of wild ginseng (Panax ginseng C.A. Meyer) leaves on lipid peroxidation levels and antixodant enzyme activities in streptozotocin diabetic rats. J Ethnopharmacol. 98:245–250. 2005.

8 

Bak MJ, Jeong JH, Kang HS, Jin KS, Jun M and Jeong WS: Stimulation of activity and expression of antioxidant enzymes by solvent fractions and isolated compound from Cedrela sinensis leaves in HepG2 cells. J Med Food. 14:405–412. 2011. View Article : Google Scholar : PubMed/NCBI

9 

Luna-Vázquez FJ, Ibarra-Alvarado C, Rojas-Molina A, et al: Nutraceutical value of black cherry Prunus serotina Ehrh. fruits: antioxidant and antihypertensive properties. Molecules. 18:14597–14612. 2013.

10 

Son Y, Cheong YK, Kim NH, Chung HT, Kang DG and Pae HO: Mitogen-activated protein kinases and reactive oxygen species: How can ROS activate MAPK pathways? J Signal Transduct. 2011:7926392011.PubMed/NCBI

11 

Chen C and Kong AN: Dietary cancer-chemopreventive compounds: from signaling and gene expression to pharmacological effects. Trends Pharmacol Sci. 26:318–326. 2005. View Article : Google Scholar : PubMed/NCBI

12 

Yu R, Jiao JJ, Duh JL, Gudehithlu K, Tan TH and Kong AN: Activation of mitogen-activated protein kinases by green tea polyphenols: potential signaling pathways in the regulation of antioxidant-responsive element-mediated phase II enzyme gene expression. Carcinogenesis. 18:451–456. 1997. View Article : Google Scholar

13 

Shin MH, Rhie GE, Kim YK, et al: H2O2 accumulation by catalase reduction changes MAP kinase signaling in aged human skin in vivo. J Invest Dermatol. 125:221–229. 2005.

14 

Kang JH, Song KH, Woo JK, et al: Ginsenoside Rp1 from Panax ginseng exhibits anti-cancer activity by down-regulation of the IGF-1R/Akt pathway in breast cancer cells. Plant Foods Hum Nutr. 66:298–305. 2011.

15 

Lee HS, Kim MR, Park Y, et al: Fermenting red ginseng enhances its safety and efficacy as a novel skin care anti-aging ingredient: in vitro and animal study. J Med Food. 15:1015–1023. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Yuan HD, Quan HY, Jung MS, et al: Anti-diabetic effect of pectinase-processed ginseng radix (GINST) in high rat diet-fed ICR mice. J Ginseng Res. 35:308–314. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Kim Y, Choi EH, Doo M, et al: Anti-stress effects of ginseng via down-regulation of tyrosine hydroxylase (TH) and dopamine β-hydroxylase (DBH) gene expression in immobilization-stressed rats and PC12 cells. Nutr Res Pract. 4:270–275. 2010.PubMed/NCBI

18 

Demir I, Kiymaz N, Gudu BO, et al: Study of the neuroprotective effect of ginseng on superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels in experimental diffuse head trauma. Acta Neurochir (Wien). 155:913–922. 2013. View Article : Google Scholar

19 

Lee MR, Yun BS, In OH and Sung CK: Comparative study of korean white, red, and black ginseng extract on cholinesterase inhibitory activity and cholinergic function. J Ginseng Res. 35:421–428. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Kang KS, Yamabe N, Kim HY, Okamoto T, Sei Y and Yokozawa T: Increase in the free radical scavenging activities of American ginseng by heat processing and its safety evaluation. J Ethnopharmacol. 113:225–232. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Oyanagui Y: Reevaluation of assay methods and establishment of kit for superoxide dismutase activity. Anal Biochem. 142:290–296. 1984. View Article : Google Scholar : PubMed/NCBI

22 

Carrillo MC, Kanai S, Nokubo M and Kitani K: (−) deprenyl induces activities of both superoxide dismutase and catalase but not of glutathione peroxidase in the striatum of young male rats. Life Sci. 48:517–521. 1991.

23 

Bogdanska JJ, Korneti P and Todorova B: Erythrocyte superoxide dismutase, glutathione peroxidase and catalase activities in healthy male subjects in Republic of Macedonia. Bratisl Lek Listy. 104:108–114. 2003.PubMed/NCBI

24 

Kirkinezos IG and Moraes CT: Reactive oxygen species and mitochondrial diseases. Semin Cell Dev Biol. 12:449–457. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Ziech D, Franco R, Pappa A and Panayiotidis MI: Reactive oxygen species (ROS)-induced genetic and epigenetic alterations in human carcinogenesis. Mutat Res. 711:167–173. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Panayiotidis M: Reactive oxygen species (ROS) in multistage carcinogenesis. Cancer Lett. 266:3–5. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Campos J, Schmeda-Hirschmann G, Leiva E, et al: Lemon grass (Cymbopogon citratus (D.C) Stapf) polyphenols protect human umbilical vein endothelial cell (HUVECs) from oxidative damage induced by high glucose, hydrogen peroxide and oxidised low-density lipoprotein. Food Chem. 151:175–181. 2014.

28 

Suchaoin W and Chanvorachote P: Caveolin-1 attenuates hydrogen peroxide-induced oxidative damage to lung carcinoma cells. Anticancer Res. 32:483–490. 2012.PubMed/NCBI

29 

Meehan WJ, Spencer JP, Rannels DE, Welch DR, Knobbe ET and Ostrander GK: Hydrogen peroxide induces oxidative DNA damage in rat type II pulmonary epithelial cells. Environ Mol Mutagen. 33:273–278. 1999. View Article : Google Scholar : PubMed/NCBI

30 

Dong GZ, Jang EJ, Kang SH, et al: Red ginseng abrogates oxidative stress via mitochondria protection mediated by LKB1-AMPK pathway. BMC Complement Altern Med. 13:642013. View Article : Google Scholar : PubMed/NCBI

31 

Jaruga P and Oliński R: Activity of antioxidant enzymes in cancer diseases. Postepy Hig Med Dosw. 48:443–455. 1994.(In Polish).

32 

Sohn SH, Kim SK, Kim YO, et al: A comparison of antioxidant activity of Korean White and Red Ginsengs on H2O2-induced oxidative stress in HepG2 hepatoma cells. J Ginseng Res. 37:442–450. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Seo SJ, Cho JY, Jeong YH and Choi YS: Effect of Korean red ginseng extract on liver damage induced by short-term and long-term ethanol treatment in rats. J Ginseng Res. 37:194–200. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Park HM, Kim SJ, Mun AR, et al: Korean red ginseng and its primary ginsenosides inhibit ethanol-induced oxidative injury by suppression of the MAPK pathway in TIB-73 cells. J Ethnopharmacol. 141:1071–1076. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Jun C and Chang CC: The effect of red ginseng extracts on the superoxide dismutase, peroxidase and catalase activities in the liver of gamma ray irradiated mice. Korean J Ginseng Sci. 17:29–34. 1993.

36 

Lee DW, Shon HO, Lim HB and Lee YG: Antioxidation of Panax ginseng. Korean J Ginseng Sci. 19:31–38. 1995.

37 

Kim GN, Lee JS, Song JH, Oh CH, Kwon YI and Jang HD: Heat processing decreases Amadori products and increases total phenolic content and antioxidant activity of Korean red ginseng. J Med Food. 13:1478–1484. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Lee SR, Kim MR, Yon JM, et al: Black ginseng inhibits ethanol-induced teratogenesis in cultured mouse embryos through its effects on antioxidant activity. Toxicol in Vitro. 23:47–52. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Kang KS, Kim HY, Pyo JS and Yokozawa T: Increase in the free radical scavenging activity of ginseng by heat-processing. Biol Pharm Bull. 29:750–754. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Sun BS, Gu LJ, Fang ZM, et al: Simultaneous quantification of 19 ginsenosides in black ginseng developed from Panax ginseng by HPLC-ELSD. J Pharm Biomed Anal. 50:15–22. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Lee MR, Kim BC, Kim R, et al: Anti-obesity effects of black gineng extract in high fat diet-fed mice. J Ginseng Res. 37:308–349. 2013. View Article : Google Scholar : PubMed/NCBI

42 

Park HJ, Shim HS, Kim KS and Shim I: The protective effect of black ginseng against transient focal ischemia-induced neuronal damage in rats. Korean J Physiol Pharmacol. 15:333–338. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Ramiro-Puig E, Urpí-Sardà M, Pérez-Cano FJ, et al: Cocoa-enriched diet enhances antioxidant enzyme activity and modulates lymphocyte composition in thymus from young rats. J Agric Food Chem. 55:6431–6438. 2007. View Article : Google Scholar : PubMed/NCBI

44 

Fan Y, Sun C, Gao X, et al: Neuroprotetive effects of ginseng pectin through the activation of ERK/MAPK and Akt survival signaling pathways. Mol Med Rep. 5:1185–1190. 2012.PubMed/NCBI

Related Articles

Journal Cover

December-2014
Volume 34 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Bak M, Jeong W and Kim K: Detoxifying effect of fermented black ginseng on H2O2-induced oxidative stress in HepG2 cells. Int J Mol Med 34: 1516-1522, 2014
APA
Bak, M., Jeong, W., & Kim, K. (2014). Detoxifying effect of fermented black ginseng on H2O2-induced oxidative stress in HepG2 cells. International Journal of Molecular Medicine, 34, 1516-1522. https://doi.org/10.3892/ijmm.2014.1972
MLA
Bak, M., Jeong, W., Kim, K."Detoxifying effect of fermented black ginseng on H2O2-induced oxidative stress in HepG2 cells". International Journal of Molecular Medicine 34.6 (2014): 1516-1522.
Chicago
Bak, M., Jeong, W., Kim, K."Detoxifying effect of fermented black ginseng on H2O2-induced oxidative stress in HepG2 cells". International Journal of Molecular Medicine 34, no. 6 (2014): 1516-1522. https://doi.org/10.3892/ijmm.2014.1972