7,8-Dihydroxyflavone protects human keratinocytes against oxidative stress-induced cell damage via the ERK and PI3K/Akt-mediated Nrf2/HO-1 signaling pathways

  • Authors:
    • Min Ju Ryu
    • Kyoung Ah Kang
    • Mei Jing Piao
    • Ki Cheon Kim
    • Jian Zheng
    • Cheng Wen Yao
    • Ji Won Cha
    • Ha Sook Chung
    • Sang Cheol Kim
    • Eunsun Jung
    • Deokhoon Park
    • Sungwook Chae
    • Jin Won Hyun
  • View Affiliations

  • Published online on: February 4, 2014     https://doi.org/10.3892/ijmm.2014.1643
  • Pages: 964-970
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

This study investigated the effect of 7,8-dihydroxyflavone (DHF) on the expression and activity of heme oxygenase-1 (HO-1), an enzyme with potent antioxidant properties, as well as the molecular mechanisms involved. DHF markedly upregulated HO-1 mRNA and protein expression in human keratinocytes (HaCaT cells), resulting in increased HO-1 activity. DHF also increased the protein level of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates HO-1 expression by binding to the antioxidant response element (ARE) within the HO-1 gene promoter, in a time‑dependent manner. Moreover, DHF decreased the expression of Kelch-like ECH-associated protein 1, a repressor of Nrf2 activity, and induced the translocation of Nrf2 from the cytosol into the nucleus, thereby allowing its association with the ARE site. DHF activated extracellular-regulated kinase (ERK) and protein kinase B (PKB, Akt) in keratinocytes, while the ERK and Akt inhibitors attenuated DHF-enhanced Nrf2 and HO-1 expression. DHF also protected the keratinocytes against hydrogen peroxide- and ultraviolet B-induced oxidative damage, while HO-1, ERK and Akt inhibitors markedly suppressed DHF-mediated cytoprotection. Taken together, the results suggested that DHF activates ERK- and Akt-Nrf2 signaling cascades in HaCaT cells, leading to the upregulation of HO-1 and cytoprotection against oxidative stress.

Introduction

Reactive oxygen species (ROS) [e.g., superoxide anions, hydroxyl radicals, and hydrogen peroxide (H2O2)] can harm cellular proteins, lipids and DNA (1,2). Long-term effects of oxidative damage to the skin include premature aging, cancer and inflammation (35).

Heme oxygenase-1 (HO-1, one of three heme oxygenase isoforms) is an extensively studied phase II enzyme that is essential for the rate-limiting step of heme catabolism. Heme catabolism leads to the formation of carbon monoxide, ferrous iron (Fe2+) and biliverdin, which is converted into bilirubin by biliverdin reductase (6). The end products of heme catabolism exert antioxidant effects by neutralizing intracellular ROS (7,8). Thus, HO-1 demonstrates potent cytoprotective properties against oxidative damage (911), and the induction of its expression increases cellular resistance to oxidative stress.

HO-1 expression is primarily regulated at the transcriptional level and is induced by the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) (12,13). Under basal conditions, Nrf2 is constitutively docked to Kelch-like ECH-associated protein 1 (Keap1) in the cytoplasm, culminating in the ubiquitination and proteosomal degradation of Nrf2 (14). However, Keap1 is oxidized under conditions of electrophilic stress, which blocks Nrf2 degradation and allows Nrf2 phosphorylation, nuclear translocation, and association with the antioxidant response element (ARE) within the HO-1 gene promoter region. The net result of ARE binding by Nrf2 is the activation of multiple antioxidant genes, including HO-1 (15).

Similar to Keap1, extracellular signal-regulated kinase (ERK) and protein kinase B (PKB, Akt) are involved in the transduction of various signals from the cell surface to the nucleus. Both of their signaling pathways are associated with the modulation of ARE-driven gene expression via Nrf2 activation (16,17).

The polyphenolic compound 7,8-dihydroxyflavone (DHF) is a member of the flavonoid family with antioxidant properties (1821).

In a previous study, we reported that DHF protected cells against oxidative stress-induced genotoxicity by scavenging intracellular ROS and enhancing Akt activity (22).

In the present study, we investigated the cytoprotective mechanisms of DHF against oxidative stress-induced cell damage in human keratinocytes with respect to its stimulatory effects on HO-1 expression and activity. We also investigated the molecular mechanisms involved in DHF-mediated cytoprotection and HO-1 induction, with a particular focus on ERK- and Akt-Nrf2 signaling cascades.

Materials and methods

Materials

[3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium] bromide (MTT) and zinc protoporphyrin (ZnPP) were purchased from Sigma Chemical Co. (St. Louis, MO, USA). Both U0126 and LY294002 were provided by Calbiochem (San Diego, CA, USA). Primary antibodies against HO-1, ERK, phospho ERK, Nrf2, and β-actin were purchased from Santa Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). The primary Akt and phospho Akt (Ser 473) antibodies were purchased from Cell Signaling Technology (Beverly, MA, USA). Rabbit polyclonal antibody specific for phospho Nrf2 was purchased from Epitomics, Inc. (Burlingame, CA, USA). Any other chemicals and reagents were of analytical grade.

Cell culture

Human keratinocytes (HaCaT cells) were obtained from Amore Pacific (Gyeonggi-do, Republic of Korea). Cells were cultured in Dulbecco’s modified Eagle’s medium containing 10% heat-inactivated fetal calf serum, streptomycin (100 μg/ml), and penicillin (100 U/ml). The cells were maintained at 37°C in an incubator with a humidified atmosphere of 5% CO2.

Reverse transcriptase-polymerase chain reaction (RT-PCR)

Cells (3×106 cells/ml) were seeded into 100-mm culture dish. Sixteen hours later, the cells were treated with DHF at concentrations of 0.5, 1, 5, 10, or 20 μg/ml. The cells were collected at the indicated times and washed twice with phosphate-buffered saline (PBS). Total RNA was isolated from the cells using TRIzol® reagent (Gibco-BRL, Grand Island, NY, USA). RT-PCR conditions for HO-1 and the housekeeping gene, glyceraldehyde 3-phosphate dehydrogenase (GAPDH), were as follows: 35 cycles of 94°C for 45 sec; 53°C for 45 sec; and 72°C for 60 sec. The primer pairs (Bionics, Seoul, Republic of Korea) used were: HO-1, forward: 5′-GAGAATGCTGAGTTCATG-3′ and reverse: 5′-ATGTTGAGCAGGAAGGC-3′; GAPDH, forward: 5′-GTGGGCCGCCCTAGGCACCAGG-3′ and reverse: 5′-GGAGGAAGAGGATGCGGCAGTG-3′. Amplified products were resolved on 1% agarose gels, stained with ethidium bromide, and images were captured under ultraviolet light.

HO-1 activity assay

HO-1 enzyme activity was measured as previously described (23). Briefly, 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. Supernatants were centrifuged at 9000 × g for 20 min, and then centrifuged at 15,000 × g for 60 min. The resulting pellet (cell lysate) was resuspended in 50 mM potassium phosphate buffer (pH 7.4), and the amount of protein in the cell lysate was determined.

A reaction mixture (200 μl) containing 500 μg/ml cell lysate, 0.2 mM hemin, 0.5 mg/ml rat liver cytosol as the source of biliverdin reductase, 0.2 mM MgCl2, 2 mM glucose-6-phosphate, 1 U/ml glucose-6-phosphate dehydrogenase, 1 mM NADPH, and 50 mM potassium phosphate buffer (pH 7.4) was incubated at 37°C for 1 h. The reaction was stopped with chloroform (0.5 ml). Following extraction, the chloroform layer was measured spectrophotometrically. Bilirubin formation from the hemin substrate was calculated according to the difference in absorption at 464 and 530 nm.

Western blot analysis

Cells (3×106 cells/ml) were seeded into a 100-mm culture dish. Sixteen hours later, the cells were treated with DHF (5 μg/ml), collected at the indicated times and washed twice with PBS. The collected cells were lysed on ice for 30 min in 100 μl lysis buffer [120 mM NaCl, 40 mM Tris (pH 8.0), and 0.1% NP 40] and centrifuged at 13,000 × g for 15 min. Supernatants were collected from the lysates, and protein concentrations of the supernatants were determined. Aliquots of the lysates (40 μg protein) were boiled for 5 min and electrophoresed on 10% sodium dodecyl sulfate polyacrylamide gels. Electrophoresed proteins were transferred onto nitrocellulose membranes (Bio-Rad, Hercules, CA, USA), which were incubated with primary antibodies. Membranes were then incubated with horseradish peroxidase-conjugated secondary antibodies (Pierce, Rockland, IL, USA) and images were captured with LAS-3000 (Fujifilm Corp., Tokyo, Japan). Protein bands were detected using an enhanced chemiluminescence Western blot detection kit (Amersham, Little Chalfont, UK).

Preparation of nuclear extracts

The cells were collected at the indicated times and lysed on ice in 1 ml lysis buffer [10 mM Tris-HCl (pH 7.9), 10 mM NaCl, 3 mM MgCl2, and 1% NP-40] for 4 min. Following centrifugation at 3,000 × g for 10 min, the pellets were resuspended in 50 μl extraction buffer [20 mM HEPES (pH 7.9), 20% glycerol, 1.5 mM MgCl2, 300 mM NaCl, 0.2 mM EDTA, 1 mM DTT, and 1 mM PMSF], incubated on ice for 30 min, and centrifuged at 13,000 × g for 5 min. Supernatants were collected and stored at −70°C after measurement of the protein concentration.

Immunocytochemistry

Cells were plated on coverslips, fixed with 4% paraformaldehyde for 30 min, and permeabilized with PBS containing 0.1% Triton X-100 for 2.5 min. The cells were then treated with blocking medium (PBS containing 3% bovine serum albumin) for 1 h and incubated with a primary anti-Nrf2 antibody diluted in blocking medium for 2 h. The immunoreacted primary antibody was detected by incubating cells with a fluorescein isothiocyanate (FITC)-conjugated secondary antibody (1:500 dilution) (Jackson ImmunoResearch Laboratories, West Grove, PA, USA) for 1 h. After washing with PBS, stained cells were mounted onto microscope slides in mounting medium containing DAPI to label nuclei (Vector, Burlingame, CA, USA). Images were collected using a Zeiss confocal microscope and Zeiss LSM 510 software.

Chromatin immunoprecipitation (ChIP)

The ChIP assay was performed with a Simple ChIP™ enzymatic chromatin IP kit (Cell Signaling Technology, Danvers, MA, USA) according to the manufacturer’s instructions, with slight modifications. Briefly, cellular proteins were crosslinked by the addition of 1% formaldehyde. Prepared chromatin was digested using a nuclease for 12 min at 37°C. A primary anti-Nrf2 antibody and normal rabbit IgG were added to the chromatin digest, and the mixture was incubated with constant rotation overnight at 4°C. To capture the immunoprecipitated complexes, ChIP-grade protein G magnetic beads were added. The beads were washed, and the immunoprecipitate was eluted with ChIP elution buffer. Crosslinking was reversed by incubating the eluent at 65°C for 30 min, followed by the addition of proteinase K and incubation at 65°C for 2 h. The immunoprecipitated DNA fragments were purified on spin columns. DNA recovered from the immunoprecipitated complexes was subjected to 35 cycles of PCR. The primers for the HO-1 gene promoter were: forward: 5′-CCAGAAAGTGGGCATCAGCT-3′ and reverse: 5′-GTCACATTTATGCTCGGCGG-3′. PCR products were resolved on 1% agarose gels, and DNA bands were visualized using ethidium bromide staining and images were captured under ultraviolet light.

Cell viability assay

The effect of DHF on cell viability was determined using the MTT assay, which is based on the reduction of a tetrazolium salt by a mitochondrial dehydrogenase in viable cells (24). Cells were seeded in a 96-well plate at a density of 3×105 cells/ml and treated for 2 h with 5 μg/ml DHF, 10 μM ZnPP (an inhibitor of HO-1), 10 nM U0126 (an inhibitor of ERK kinase), and 5 μM LY294002 (an inhibitor of PI3K), followed by 1 mM H2O2 or 30 mJ/cm2 UVB for 1 h. After incubation for 24 h at 37°C, 50 μl MTT stock solution (2 mg/ml) was added to each well to achieve a total reaction volume of 250 μl. After incubation for 2.5 h, the supernatants were aspirated. The formazan crystals in each well were dissolved in 150 μl dimethyl sulfoxide, and the absorbance at 540 nm was read on a scanning multi-well spectrophotometer.

Statistical analysis

Values are given as the mean ± standard error of the mean (SEM). The results were subjected to an analysis of variance (ANOVA) followed by the Tukey’s test to analyze differences between conditions. P<0.05 was considered to indicate statistical significance.

Results

DHF induces HO-1 mRNA levels and activity

DHF at concentrations of 0.5, 1 and 5 mg/ml dose-dependently increased HO-1 mRNA levels in HaCaT cells, while DHF at concentrations of 10 and 20 mg/ml slightly decreased mRNA levels relative to the level induced by 5 mg/ml DHF (Fig. 1A). Furthermore, DHF (0.5, 1, and 5 mg/ml) dose-dependently increased HO-1 activity, with a clear effect at 5 mg/ml (Fig. 1B). When cell viability was assessed by the MTT assay, DHF did not show any cytotoxicity at 0.5, 1, or 5 mg/ml (data not shown). From these results, 5 mg/ml DHF was selected as the optimal concentration for subsequent experiments. DHF (5 mg/ml) enhanced HO-1 mRNA and protein expression in a time-dependent manner (Fig. 1C and D). The time course for the elevated HO-1 expression was closely correlated with that for the increased HO-1 activity (Fig. 1E).

DHF induces Nrf2 expression, nuclear translocation and binding to ARE

Nrf2 is a redox-sensitive basic-leucine zipper transcription factor. Its activity is blocked by Keap1, a negative regulator of Nrf2 that forms a complex with the transcription factor in the cytoplasm and prevents its phosphorylation and translocation into the nucleus (14). Protein levels of total nuclear and phosphorylated Nrf2 (phospho Nrf2) were increased in a time-dependent manner by DHF treatment of HaCaT cells (Fig. 2A), whereas Keap1 expression was decreased (Fig. 2B). DHF markedly increased the translocation of Nrf2 from the cytosol into the nucleus (Fig. 2C). Moreover, Nrf2 binding to the ARE sequence in the HO-1 promoter region was markedly higher in DHF-treated cells than in untreated cells, as assessed by the ChIP assay (Fig. 2D).

DHF activates Nrf2 and increases HO-1 expression via phosphorylation of ERK and Akt

To investigate the upstream signaling pathways involved in DHF-enhanced Nrf2 activation and HO-1 expression, we examined the phosphorylation of ERK and Akt, which are important signaling enzymes that are involved in cellular protection against oxidative stress (16,25). DHF treatment of keratinocytes increased the phosphorylation of ERK and Akt in a time-dependent manner (Fig. 3A). Moreover, selective inhibitors of ERK (U0126) and Akt (LY294002, which blocks PI3K upstream of Akt) reduced the levels of DHF-induced phospho-Nrf2 (Fig. 3B) and HO-1 (Fig. 3C). These results suggest that DHF increased the levels of phospho-Nrf2 and HO-1 via ERK- and PI3K/Akt-dependent pathways.

Involvement of HO-1 and ERK and Akt pathways in DHF-induced cytoprotection against oxidative stress

To determine whether DHF-enhanced HO-1 activity confers cytoprotection to keratinocytes against oxidative stress, HaCaT cells were pre-treated with the HO-1 inhibitor ZnPP and then exposed to H2O2 or UVB radiation. Cell viability was significantly reduced by H2O2 or UVB exposure, but not by pre-treatment with DHF or ZnPP alone (Fig. 4A and B). Pre-treatment with DHF prevented H2O2 and UVB-induced cytotoxicity. ZnPP attenuated the protective effect of DHF against H2O2 and UVB-induced cellular damage, as assessed by reduced cell viability in the MTT assay (Fig. 4A and B). Therefore, the cytoprotective effect of DHF is likely to be mediated through HO-1 induction.

We also examined whether DHF can mitigate oxidative damage to cells through the activation of ERK and Akt. U0126 and LY294002 reduced the protective effect of DHF against H2O2 and UVB exposure (Fig. 4C and D), suggesting that ERK and Akt signaling pathways are also involved in DHF-mediated cytoprotection.

Discussion

The epidermis and its associated cells comprise a critical defense mechanism against oxidative damage. Keratinocytes in particular are constantly exposed to environmental pro-oxidants. Such exposure leads to harmful effects on the skin (e.g., premature aging and cancer) in the absence of appropriate protective cell responses.

The current study has demonstrated that DHF, a naturally occurring antioxidant, activated HO-1 expression by targeting the ARE sequence within the HO-1 gene promoter region. The DHF-mediated activation of HO-1 was regulated through the stimulation of Nrf2 in an ERK and PI3K/Akt-dependent manner. Findings of previous studies have demonstrated the involvement of ERK and PI3K/Akt in the activation of HO-1 expression and Nrf2-mediated signal transduction (2527). For example, ERK phosphorylates Nrf2, triggering dissociation of the Keap1-Nrf2 complex and translocation of Nrf2 into the nucleus, where it forms a heterodimer with a small Maf protein (16,2830). In the present study, DHF treatment increased the levels of phospho-ERK and -PI3K/Akt. In addition, specific inhibitors of the ERK and PI3K/Akt pathways suppressed the nuclear translocation of Nrf2, suggesting that Nrf2 is a direct downstream target of ERK and PI3K/Akt. Thus, ERK and PI3K/Akt may be crucial components of the cellular signaling network responsible for the activation of Nrf2 and the transcriptional regulation of HO-1 gene expression in keratinocytes.

Similar to DHF, several compounds isolated from natural products induce HO-1 expression via activation of Nrf2 downstream of ERK and/or PI3K/Akt. For instance, eckol (found in brown algae) and (-)-epigallocatechin gallate (found in green tea) increase HO-1 levels by activating ERK, PI3K/Akt and Nrf2 (31,32). In addition, baicalein (originally isolated from the root of Scutellaria baicalensis) prevents oxidative damage in PC12 cells by stimulating the PI3K/Akt/Nrf2 pathway in order to upregulate HO-1 expression (33). Furthermore, DHP exhibited the most potent protective effects as compared with other flavonoids in the neuroprotective area (34).

Under normal physiological conditions, Nrf2 is sequestered in the cytoplasm by Keap1. Following the oxidation of specific cysteine residues within Keap1 by electrophilic agents or ROS, Nrf2 dissociates from its cytoplasmic docking protein and translocates into the nucleus. There, it binds to the promoter regions of several phase II detoxifying enzymes or antioxidant enzymes including HO-1, thereby activating their transcription (35).However, because the half-life of Nrf2 is very short (<10 min), these mechanisms necessitate the stabilization of the Nrf2 protein (36,37).

In our system, DHF treatment decreased Keap1 expression and increased the level and nuclear translocation of Nrf2. These results suggest that DHF may retard Keap1-mediated Nrf2 degradation. Similarly, baicalein increases Keap1 degradation by the proteasome system and prevents the Keap1-mediated inactivation of Nrf2 (33). We also showed that ERK and PI3K inhibitors decreased the DHF-induced accumulation of phospho-Nrf2 through the inhibition of ERK and Akt phosphorylation. This suggests that ERK and PI3K/Akt phosphorylate Nrf2 and facilitate its release from the Keap1-Nrf2 complex, allowing activated Nrf2 to translocate into the nucleus.

In summary, the present results suggest that DHF protects HaCaT cells against oxidative stress-induced cell damage. DHF exerts its protective functions by i) activating ERK and PI3K/Akt, ii) facilitating the translocation of Nrf2 into the nucleus and its subsequent binding to ARE and iii) upregulating HO-1 expression.

Acknowledgements

This study was supported by a grant from the Korean Ministry of Knowledge and Economy (R0000445).

References

1 

Ismail NS, Pravda EA, Li D, Shih SC and Dallabrida SM: Angiopoietin-1 reduces H(2)O(2)-induced increases in reactive oxygen species and oxidative damage to skin cells. J Invest Dermatol. 130:1307–1317. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Okayama Y: Oxidative stress in allergic and inflammatory skin diseases. Curr Drug Targets Inflamm Allergy. 4:517–519. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Black HS: Potential involvement of free radical reactions in ultraviolet light-mediated cutaneous damage. Photochem Photobiol. 46:213–221. 1987. View Article : Google Scholar : PubMed/NCBI

4 

Cerutti PA: Prooxidant states and tumor promotion. Science. 227:375–381. 1985. View Article : Google Scholar : PubMed/NCBI

5 

Harman D: Free radical theory of aging: an update: increasing the functional life span. Ann NY Acad Sci. 1067:10–21. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Ryter SW, Alam J and Choi AM: Hemeoxygenase-1/carbon monoxide: from basic science to therapeutic applications. Physiol Rev. 86:583–650. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Tenhunen R, Marver HS and Schmid R: The enzymatic conversion of heme to bilirubin by microsomal heme oxygenase. Proc Natl Acad Sci USA. 61:748–755. 1968. View Article : Google Scholar : PubMed/NCBI

8 

Maines MD: Heme oxygenase: function, multiplicity, regulatory mechanisms, and clinical applications. FASEB J. 2:2557–2568. 1988.PubMed/NCBI

9 

Ryter SW and Choi AM: Heme oxygenase-1: molecular mechanisms of gene expression in oxygen-related stress. Antioxid Redox Signal. 4:625–632. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Otterbein LE, Soares MP, Yamashita K and Bach FH: Heme oxygenase-1: unleashing the protective properties of heme. Trends Immunol. 24:449–455. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Li MH, Cha YN and Surh YJ: Peroxynitrite induces HO-1 expression via PI3K/Akt-dependent activation of NF-E2-related factor 2 in PC12 cells. Free Radic Biol Med. 41:1079–1091. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Khor TO, Huang MT, Kwon KH, Chan JY, Reddy BS and Kong AN: Nrf2-deficient mice have an increased susceptibility to dextran sulfate sodium-induced colitis. Cancer Res. 66:11580–11584. 2006. View Article : Google Scholar : PubMed/NCBI

13 

Alam J and Cook JL: Transcriptional regulation of the heme oxygenase-1 gene via the stress response element pathway. Curr Pharm Des. 9:2499–2511. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Itoh K, Wakabayashi N, Katoh Y, Ishii T, Igarashi K, Engel JD and Yamamoto M: Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 13:76–86. 1999. View Article : Google Scholar : PubMed/NCBI

15 

Yu S, Khor TO, Cheung KL, Li W, Wu TY, Huang Y, Foster BA, Kan YW and Kong AN: Nrf2 expression is regulated by epigenetic mechanisms in prostate cancer of TRAMP mice. PLoS One. 5:e85792010. View Article : Google Scholar : PubMed/NCBI

16 

Li MH, Jang JH, Na HK, Cha YN and Surh YJ: Carbon monoxide produced by heme oxygenase-1 in response to nitrosative stress induces expression of glutamate-cysteine ligase in PC12 cells via activation of phosphatidylinositol 3-kinase and Nrf2 signaling. J Biol Chem. 282:28577–28586. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Yu R, Chen C, Mo YY, Hebbar V, Owuor ED, Tan TH and Kong AN: Activation of mitogen-activated protein kinase pathways induces antioxidant response element-mediated gene expression via a Nrf2-dependent mechanism. J Biol Chem. 275:39907–39913. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Silva MM, Santos MR, Caroco G, Rocha R, Justino G and Mira L: Structure-antioxidant activity relationships of flavonoids: a re-examination. Free Radic Res. 36:1219–1227. 2002. View Article : Google Scholar : PubMed/NCBI

19 

Melidou M, Riganakos K and Galaris D: Protection against nuclear DNA damage offered by flavonoids in cells exposed to hydrogen peroxide: the role of iron chelation. Free Radic Biol Med. 39:1591–1600. 2005. View Article : Google Scholar : PubMed/NCBI

20 

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

21 

Moridani MY, Pourahmad J, Bui H, Siraki A and O’Brien PJ: Dietary flavonoid iron complexes as cytoprotective superoxide radical scavengers. Free Radic Biol Med. 34:243–253. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Zhang R, Kang KA, Piao MJ, Ko DO, Wang ZH, Chang WY, You HJ, Lee IK, Kim BJ, Kang SS and Hyun JW: Preventive effect of 7,8-dihydroxyflavone against oxidative stress-induced genotoxicity. Biol Pharm Bull. 32:166–171. 2009. View Article : Google Scholar : PubMed/NCBI

23 

Kutty RK and Maines MO: Oxidation of heme c derivatives by purified heme oxygenase. Evidence for the presence of one molecular species of heme oxygenase in the rat liver. J Biol Chem. 257:9944–9952. 1982.PubMed/NCBI

24 

Carmichael J, DeGraff WG, Gazdar AF, Minna JD and Mtchell JB: Evaluation of a tetrazolium-based semiautomated colorimetric assay: assessment of chemosensitivity testing. Cancer Res. 47:936–942. 1987.PubMed/NCBI

25 

Cullinan SB and Diehl JA: PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress. J Biol Chem. 279:20108–20117. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Bloom DA and Jaiswal AK: Phosphorylation of Nrf2 at Ser40 by protein kinase C in response to antioxidants leads to the release of Nrf2 from INrf2, but is not required for Nrf2 stabilization/accumulation in the nucleus and transcriptional activation of antioxidant response element-mediated NAD(P)H: quinone oxidoreductase-1 gene expression. J Biol Chem. 278:44675–44682. 2003.

27 

Hwang YP and Jeong HG: Ginsenoside Rb1 protects against 6-hydroxydopamine-induced oxidative stress by increasing heme oxygenase-1 expression through an estrogen receptor-related PI3K/Akt/Nrf2-dependent pathway in human dopaminergic cell. Toxicol Appl Pharmacol. 242:18–28. 2010. View Article : Google Scholar

28 

Chan K, Han XD and Kan YW: An important function of Nrf2 in combating oxidative stress: detoxification of acetaminophen. Proc Natl Acad Sci USA. 98:4611–4616. 2001. View Article : Google Scholar : PubMed/NCBI

29 

Kim YC, Masutani H, Yamaguchi Y, Itoh K, Yamamoto M and Yodoi J: Hemin-induced activation of the thioredoxin gene by Nrf2. A differential regulation of the antioxidant responsive element by a switch of its binding factors. J Biol Chem. 276:18399–18406. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Kwak MK, Itoh K, Yamamoto M and Kensler TW: Enhanced expression of the transcription factor Nrf2 by cancer chemopreventive agents: role of antioxidant response element-like sequences in the nrf2 promoter. Mol Cell Biol. 22:2883–2892. 2002. View Article : Google Scholar : PubMed/NCBI

31 

Na HK, Kim EH, Jung JH, Lee HH, Hyun JW and Surh YJ: (-)-Epigallocatechin gallate induces Nrf2-mediated antioxidant enzyme expression via activation of PI3K and ERK in human mammary epithelial cells. Arch Biochem Biophys. 476:171–177. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Kim KC, Kang KA, Zhang R, Piao MJ, Kim GY, Kang MY, Lee SJ, Lee NH, Surh YJ and Hyun JW: Up-regulation of Nrf2-mediated heme oxygenase-1 expression by eckol, a phlorotannin compound, through activation of Erk and PI3K/Akt. Int J Biochem Cell Biol. 42:297–305. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Zhang Z, Cui W, Li G, Yuan S, Xu D, Hoi MP, Lin Z, Dou J, Han Y and Lee SM: Baicalein protects against 6-OHDA-induced neurotoxicity through activation of Keap1/Nrf2/HO-1 and involving PKCα and PI3K/AKT signaling pathways. J Agric Food Chem. 60:8171–8182. 2012.PubMed/NCBI

34 

Jang SW, Liu X, Yepes M, Shepherd KR, Miller GW, Liu Y, Wilson WD, Xiao G, Blanchi B, Sun YE and Ye K: A selective TrkB agonist with potent neurotrophic activities by 7,8-dihydroxyflavone. Proc Natl Acad Sci USA. 107:2687–2692. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Kobayashi M and Yamamoto M: Molecular mechanisms activating the Nrf2-Keap1 pathway of antioxidant gene regulation. Antioxid Redox Signal. 7:385–394. 2005. View Article : Google Scholar : PubMed/NCBI

36 

Jain AK, Mahajan S and Jaiswal AK: Phosphorylation and dephosphorylation of tyrosine 141 regulate stability and degradation of INrf2: a novel mechanism in Nrf2 activation. J Biol Chem. 283:17712–17720. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Lee OH, Jain AK, Papusha V and Jaiswal AK: An auto-regulatory loop between stress sensors INrf2 and Nrf2 controls their cellular abundance. J Biol Chem. 282:36412–36420. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

2014-April
Volume 33 Issue 4

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
Ryu MJ, Kang KA, Piao MJ, Kim KC, Zheng J, Yao CW, Cha JW, Chung HS, Kim SC, Jung E, Jung E, et al: 7,8-Dihydroxyflavone protects human keratinocytes against oxidative stress-induced cell damage via the ERK and PI3K/Akt-mediated Nrf2/HO-1 signaling pathways. Int J Mol Med 33: 964-970, 2014
APA
Ryu, M.J., Kang, K.A., Piao, M.J., Kim, K.C., Zheng, J., Yao, C.W. ... Hyun, J.W. (2014). 7,8-Dihydroxyflavone protects human keratinocytes against oxidative stress-induced cell damage via the ERK and PI3K/Akt-mediated Nrf2/HO-1 signaling pathways. International Journal of Molecular Medicine, 33, 964-970. https://doi.org/10.3892/ijmm.2014.1643
MLA
Ryu, M. J., Kang, K. A., Piao, M. J., Kim, K. C., Zheng, J., Yao, C. W., Cha, J. W., Chung, H. S., Kim, S. C., Jung, E., Park, D., Chae, S., Hyun, J. W."7,8-Dihydroxyflavone protects human keratinocytes against oxidative stress-induced cell damage via the ERK and PI3K/Akt-mediated Nrf2/HO-1 signaling pathways". International Journal of Molecular Medicine 33.4 (2014): 964-970.
Chicago
Ryu, M. J., Kang, K. A., Piao, M. J., Kim, K. C., Zheng, J., Yao, C. W., Cha, J. W., Chung, H. S., Kim, S. C., Jung, E., Park, D., Chae, S., Hyun, J. W."7,8-Dihydroxyflavone protects human keratinocytes against oxidative stress-induced cell damage via the ERK and PI3K/Akt-mediated Nrf2/HO-1 signaling pathways". International Journal of Molecular Medicine 33, no. 4 (2014): 964-970. https://doi.org/10.3892/ijmm.2014.1643