anti-oxidative activity of hydrolysate from rice bran ...download.xuebalib.com/3nclwbc3xtv.pdf ·...

27
Biological and Pharmaceutical Bulletin Advance Publication by J-STAGE DOI:10.1248/bpb.b16-00971 2017 The Pharmaceutical Society of Japan Advance Publication April 6, 2017 Biol. Pharm. Bull. Regular Article Anti-oxidative activity of hydrolysate from rice bran protein in HepG2 cells Chie Moritani a , Kayoko Kawakami a , Akiko Fujita b , Koji Kawakami b , Tadashi Hatanaka c and Seiji Tsuboi *, a a Department of Biochemistry, School of Pharmacy, Shujitsu University; 1-6-1 Nishigawara, Okayama 703-8516, Japan, b Satake Corporation, 2-30 Saijo Nishihonmachi; Higashi- Hiroshima-shi, Hiroshima, 739-8602, Japan and c Okayama Prefectural Technology Center for Agriculture, Forestry, and Fisheries, Research Institute for Biological Sciences (RIBS); 7549-1 Yoshikawa, Kibi-chuo, Okayama 716-1241, Japan *To whom correspondence should be addressed. Seiji Tsuboi: Department of Biochemistry, School of Pharmacy, Shujitsu University, 1-6-1 Nishigawara, Okayama 703-8516, Japan E-mail address: [email protected]

Upload: others

Post on 21-Oct-2020

8 views

Category:

Documents


0 download

TRANSCRIPT

  • Biological and Pharmaceutical Bulletin Advance Publication by J-STAGE DOI:10.1248/bpb.b16-00971

    Ⓒ 2017 The Pharmaceutical Society of Japan

    Advance Publication April 6, 2017

    Biol. Pharm. Bull.

    Regular Article

    Anti-oxidative activity of hydrolysate from rice bran protein in HepG2 cells

    Chie Moritania, Kayoko Kawakamia, Akiko Fujitab, Koji Kawakamib, Tadashi Hatanakac and

    Seiji Tsuboi*, a

    aDepartment of Biochemistry, School of Pharmacy, Shujitsu University; 1-6-1 Nishigawara,

    Okayama 703-8516, Japan, bSatake Corporation, 2-30 Saijo Nishihonmachi; Higashi-

    Hiroshima-shi, Hiroshima, 739-8602, Japan and cOkayama Prefectural Technology Center

    for Agriculture, Forestry, and Fisheries, Research Institute for Biological Sciences (RIBS);

    7549-1 Yoshikawa, Kibi-chuo, Okayama 716-1241, Japan

    *To whom correspondence should be addressed.

    Seiji Tsuboi: Department of Biochemistry, School of Pharmacy, Shujitsu University, 1-6-1

    Nishigawara, Okayama 703-8516, Japan

    E-mail address: [email protected]

  • Biological and Pharmaceutical Bulletin Advance Publication

    SUMMARY

    Glutathione (GSH) is an ubiquitous thiol-containing tripeptide, which plays important

    roles in cellular protection from oxidative stress. In our search for a dietary source that can

    increase glutathione (GSH) levels, we discovered that a 24 h treatment of HepG2 cells with

    rice bran protein hydrolysate (RBPH), prepared by Umamizyme G-catalyzed hydrolysis,

    increased the GSH content in a dose-dependent manner. RBPH elevated the expression levels

    ofγ-glutamylcysteine synthetase (γ-GCS), which constitutes the rate-limiting enzyme of

    GSH synthesis, and of another two enzymes, hemeoxygenase-1 (HO-1) and NAD(P)H

    quinone oxidoreductase1 (NQO1). This induction was preceded by the accumulation of Nrf2

    (nuclear factor erythroid 2-related factor 2) inside the nucleus, which is a key transcription

    factor for the expression of the γ-GCS, HO-1, and NQO1. Pre-treatment of cells with RBPH

    produced a significant protective effect against cytotoxicity caused by H2O2 or ethanol. These

    results indicate that RBPH exerts a protective effect against oxidative stress by modulating

    GSH levels and anti-oxidative enzyme expression via the Nrf2 pathway.

    KEYWORDS: rice bran; anti-oxidative; glutathione; nuclear factor erythroid 2-related factor

    2

  • Biological and Pharmaceutical Bulletin Advance Publication

    INTRODUCTION

    Rice is the most important cereal food in Japan. Rice bran (RB), which constitutes

    approximately 10% of the grain, is a major by-product of rice milling. RB is rich in protein,

    lipids, dietary fibers, and vitamins.1, 2) Recently, RB was recognized as a functional ingredient,

    containing such antioxidants as tocopherols, tocotorienols, and γ-oryzanol.3, 4) Moreover,

    enzymatically produced RB protein hydrolysates (RBPHs) were found to possess various

    biological functions with potential medical applications. In a previous study, we

    demonstrated that RBPH produced with Umamizyme G, a commercial protease from

    Aspergillus oryzae, from defatted RB protein exhibited the inhibitory activity of

    dipeptidylpetidase-IV (DPP-IV) that is a key regulator involved in the prevention and

    treatment of type 2 diabetes. 5) RBPHs produced with other peptidases were shown to have an

    anti-proliferative effect on cancer cells,6) and the ability to reduce micellar cholesterol

    levels.7)

    Excess generation of reactive oxygen species (ROS) leads to oxidative stress, a condition

    characterized by ROS attacks on proteins, lipids, and DNA, leading to cell-function disorders.

    Oxidative stress is thought to be involved in the pathogenesis of various diseases, e.g.

    cancer,8) diabetes,9) cardiovascular diseases,10) and neurodegenerative disorders.11) Thus,

    maintenance or restoration of the mammalian cell balance between ROS generation and

    detoxification through the action of anti-oxidative molecules and enzymes that decrease

    oxidative stress, may be important in the prevention of these pathological conditions.12)

    Glutathione (GSH), a ubiquitous thiol-tripeptide, is a major cellular anti-oxidative

    molecule, as it has the ability, by itself or in combination with GSH peroxidase, to scavenge

    H2O2, other peroxides and free radicals. GSH is biosynthesized from glutamate, cysteine, and

    glycine through a two-step ATP-dependent reaction. The first rate-limiting step is catalyzed

    by the enzyme γ-glutamylcysteine synthetase (γ-GCS), which is a dimer consisting of a heavy

  • Biological and Pharmaceutical Bulletin Advance Publication

    chain (γ-GCSh) and a light chain (γ-GCSl).13) The expression of γ-GCS and various other

    antioxidant and phase-2 enzymes, such as hemeoxygenase 1 (HO-1), NAD(P)H quinone

    oxidoreductase 1 (NQO1), catalase, and GSH S-transferase, is mainly regulated by the

    transcription factor Nrf2 (Nuclear factor erythroid 2-related factor 2).14, 15) Under normal

    oxidation conditions, Nrf2 is located in the cytoplasm, bound to the Kelch-like ECH

    associated protein 1 (Keap1), which inhibits Nrf2 translocation to the nucleus. In response to

    oxidative stress, Nrf2 is released from Keap1, translocates to the nucleus, and activates the

    expression of the aforementioned genes, exerting an anti-oxidative cytoprotective effect.16)

    Therefore, activation of Nrf2 contributes to the regulation of GSH levels and the maintenance

    of normal redox status in cells.

    Various hydrolysates derived from dietary proteins have been shown to exert biological

    functions, including anti-oxidative, anti-hypertensive, anti-diabetic, and immuno-modulating

    activities.17, 18) After studying the biological functions of RBPH, we discovered that it can

    increase GSH levels in HepG2 cells, and exerts a protective effect against cytotoxicity

    induced by oxidative stress, through the induction of the Nrf2 pathway.

    MATERIALS AND METHODS

    Materials Defatted RB was a gift from SATAKE Co. Ltd. (Higashi-Hiroshima, Japan).

    Umamizyme G was obtained from AMANO Enzyme Co. Ltd. (Nagoya, Japan), soybean

    protein (FUJIPRO E) from FUJI OIL Co. Ltd. (Osaka, Japan), and collagen peptide from

    Nippi (Tokyo, Japan). The protein assay kit was purchased from Bio-Rad Laboratories Inc.

    (Hercules, CA, USA). 7-Benzo-2-oxa-1,3-diazole-4-sulfonic acid (SBD-F) and 4-

    (aminosulfonyl)-7-fluoro-2,1,3-benzoxadiazole (ABD-F) were obtained from Dojindo Labs

    (Kumamoto, Japan), while 5,5′-dithiobis-(2-nitrobenzoic acid) (DTNB) was bought from

    Wako Pure Chemical (Osaka, Japan).

  • Biological and Pharmaceutical Bulletin Advance Publication

    Preparation of RBPH RBPH was prepared as described previously.5) In brief, defatted RB

    was solubilized in distilled water, whose pH had been adjusted to 12.5 using NaOH, by

    stirring for 2 h at 45 °C. After centrifugation at 2,000 × g for 15 min, the supernatant was

    collected, and the pH was adjusted to 4.0 with 1 M HCl. After a new centrifugation, the solid

    residue (RB proteins) was dried in a vacuum oven, overnight at 40 °C. The obtained proteins

    were hydrolyzed with a 1% (w/w) solution of Umamizyme G, overnight at 45 °C. After a 30

    min incubation at 80 °C for protease inactivation, the hydrolysate was centrifuged at 2,000 ×

    g for 30 min. The supernatant (RBPH) was divided into aliquots and freeze-dried. The RBPH

    was dissolved in 25 mM Hepes-NaOH before further analysis. RB protein without hydrolysis

    was prepared by a similar method to RBPH, except for the absence of Umamizyme G.

    Cell cultures HepG2 and COS7 cells were purchased from the RIKEN Cell Bank (Tsukuba,

    Japan), and maintained in MEM and DMEM, respectively. Both media were supplemented

    with 10% fetal bovine serum (FBS), 100 µg/ml streptomycin, 100 µg/ml penicillin, and 0.56

    µg/ml amphotericin B. The cells were cultured at 37 °C in a humidified atmosphere

    containing 5 % CO2.

    Measurement of intracellular GSH levels Cells were seeded into 6-well plates at a

    concentration of 1.5·105 cells / well. After 48 h, the culture medium was replaced with

    medium containing RBPH. After incubating for the indicated periods of time, cells were

    rinsed twice with PBS and collected. Following homogenization and deproteinization, the

    obtained supernatants were used for measuring the GSH content. The concentrations of GSH

    and its oxidized form, glutathione disulfide (GSSG), were determined simultaneously by

    HPLC-fluorescence detection, after labeling with ABD-F and SBD-F, respectively.19) The

    total GSH content was determined by the enzymatic recycling method using GSH reductase

  • Biological and Pharmaceutical Bulletin Advance Publication

    and DTNB.20)

    Real-time PCR analysis Total RNA from RBPH-treated cells was extracted with the

    RNeasy Plus Mini Kit (QIAGEN, Hilden, Germany), according to the manufacturer’s

    instructions. First strand cDNA was synthesized from 1 µg of total RNA using the

    PrimeScript RT-PCR kit (Takara, Tokyo, Japan). Real-time PCR was performed using SYBR

    Premix EX Taq (Takara), and fluorescence was quantified with the ABI PRISM 7700

    sequence detection system (Thermo Fisher Scientific, Waltham, MA, USA). The cDNA

    levels of the house keeping gene, β -actin, were used as an endogenous control. The

    sequences of the primers used in the PCR were as follows: β-GCSh forward primer, 5′-

    TGCTGTCTCCAGGTGACATTC-3′ and reverse primer, 5′-cccagcgacaatcaatgtct-3′)21);

    β-GCSl forward primer 5′-TCCAGTTCCTGCACATCTACCA-3′ and reverse primer,

    5 ′ -TCATCGCCCCACTTGAGAA-3 ′ ); HO-1 forward primer, 5 ′ -

    GCAACCCGACAGCATGC-3′ and reverse primer, 5′-TGCGGTGCAGCTCTTCTG-3′

    22); NQO1 (forward primer, 5′-CATGAATGTCATTCTCTGGCCA-3′ and reverse primer,

    5 ′ -CTGGAGTGTGCCCAATGCTA-3 ′ ); Nrf-2 forward primer, 5 ′ -

    TGCTTTATAGCGTGCAAACCTCGC-3 ′ and reverse primer, 5 ′ -

    ATCCATGTCCCTTGACAGCACAGA-3 ′ 23); β -actin forward primer, 5 ′ -

    CCTGGCACCCAGCACAAT-3 ′ and reverse primer, 5 ′ -

    GCCGATCCACACGGAGTACT-3′.

    Western blotting Treated cells were washed twice with PBS and harvested using a cell

    scraper. Harvested cells were lysed using the RIPA Lysis Buffer System (Santa Cruz

  • Biological and Pharmaceutical Bulletin Advance Publication

    Biotechnology, Dallas, TX, USA). Cells were incubated in the lysis buffer for 30 min on ice.

    After centrifugation at 13,000 × g for 15 min at 4 °C, the supernatants were collected as cell

    lysates.

    Nuclear extracts were prepared as described.24) Briefly, harvested cells were suspended in

    200 µl of extraction buffer containing 10 mM HEPES, pH 7.5, 150 mM NaCl, 0.6 % Nonidet

    P-40, 1 mM EDTA, 5 mM DTT, supplemented with a proteinase-inhibitor cocktail (Roche

    Applied Science, Penzberg, Germany) just before use. After a 20-min incubation on ice,

    nuclei were pelleted by centrifugation at 13,000 × g for 15 min at 4 °C. The nuclear pellet

    was extracted with a solution containing 10 mM HEPES, pH 7.9, 420 mM NaCl, 1.5 mM

    MgCl2, 0.2 mM EDTA, 5 mM DTT, supplemented with a proteinase-inhibitor cocktail.

    Nuclear fractions were collected after a 15 min centrifugation at 13,000 g, at 4 °C.

    Samples of cell lysates and nuclear extracts containing 20 µg of protein were separated by

    SDS-polyacrylamide gel electrophoresis and transferred onto a PVDF membrane (Bio-Rad).

    The membrane was incubated with primary antibodies against γ -GCSh (Santa Cruz

    Biotechnology), γ-GCSl, Nrf2 (Santa Cruz Biotechnology), HO-1 (Enzo Life Science),

    Lamin B2 (Santa Cruz Biotechnology) or β-actin (Sigma, St. Louis, MO, USA), followed by

    incubation with horseradish peroxidase-linked second antibodies. The immune complexes on

    the membrane were detected with the Amersham ECL Prime western blotting detection

    reagent (GE Healthcare, Chicago, IL, USA) in a LAS-1000 imager (Fuji, Tokyo, Japan).

    Band intensities were analyzed using ImageJ software (Public Domain).

    Lactate dehydrogenase (LDH) cytotoxicity assay The activity of LDH released from

    damaged cells into the medium was measured using the Cytotoxic Detection Kit (Roche

    Applied Science). HepG2 cells seeded into 96-well microplates at a concentration of 1.5 · 104

  • Biological and Pharmaceutical Bulletin Advance Publication

    cells / well were treated with RBPH for 24 h. After incubation, the medium was replaced with

    FBS-free medium containing H2O2 or ethanol. The culture medium was collected after

    incubation periods of 1 h and 24 h for H2O2 and ethanol, respectively, and used to measure

    the activity of released LDH (sample). The activity of the total LDH in the culture (LDHhigh

    control) was determined by lysing cells in 1 % Triton X-100, while the LDH activity from the

    medium of untreated cells was defined as LDHlow control. After the subtraction of background

    absorbance from all other values, the cytotoxicity was calculated as follows:

    % = – ℎ ℎ – x100

    RESULTS

    Effect of various protein hydrolysates on the intracellular GSH level We examined the

    effect of hydrolysates of various proteins on the intracellular GSH levels of HepG2 cells after

    24 h incubations with each hydrolysate, at a concentration of 5 mg/ml. Cultures treated with

    RBPH displayed about double the amount of GSH compared to the untreated controls (Fig.

    1A). Soybean protein hydrolysate produced by Umamizyme G and collagen peptide did not

    cause statistically significant changes in intracellular GSH levels. Treatment of the cells with

    RB protein without hydrolysis did not increase the intracellular GSH levels (Fig. 1B). A dose-

    response experiment revealed that RBPH elevated the intracellular GSH levels in HepG2 and

    COS7 cells in a dose-dependent fashion (Fig. 2A, 2B). On the other hand, the ratio of GSH to

    GSSG, which reflects the redox status in the cell, was not significantly affected (Fig. 2C).

    The time course analysis indicates that the increase of the intracellular GSH levels was

    relatively slow and tended to reach a plateau at 16 h (Fig. 2D).

    Effect of RBPH on the expression of γ-GCSh and γ-GCSl In an effort to determine the

  • Biological and Pharmaceutical Bulletin Advance Publication

    mechanism behind RBPH's ability to increase intracellular GSH levels, we determined the

    mRNA levels of the γ-GCSh and γ-GCSl genes, that encode the two subunits comprising

    the rate-limiting enzyme of GSH synthesis, γ-GCS mRNA levels of both genes began to

    increase at 3 h after the addition of RBPH, reaching a statistically significant increase of

    about 50% at 8 h (Fig. 3A, 3B). Protein levels of both subunits were found to be decreased at

    1 and 3 h after addition, but this reduction had disappeared at the samples collected at 8 h and

    the protein levels increased at 24 h though without significance (Fig. 3C, 3D). In addition, the

    increase of the intracellular GSH levels was inhibited by about 35% after treatment with 1

    mM methionine sulfoximine, which is a known inhibitor of γ-GCS (data not shown).25)

    Effect of RBPH on NQO1 and HO-1 expression levels To determine the effect of RBPH

    on the expression of anti-oxidant and phase II detoxifying enzymes, the expression of NQO1

    and HO-1 was investigated at various times after RBPH addition. At 3 h, mRNA levels of

    NQO1 were increased by about 50%, and remained roughly at this level until the end of the

    experiment (24 h) (Fig. 4A). RBPH also induced the mRNA levels of HO-1, but more slowly.

    The levels had statistically significantly increased by about 100% at 8 h, and kept up to 24 h

    (Fig. 4B). The HO-1 protein levels were also increased by 150% at 24 h (Fig. 4C).

    Effects of RBPH on Nrf2 Nrf2 is known as a key regulator for anti-oxidant and phase II

    detoxifying enzymes such as γ -GCS, HO-1 and NQO1. Since mRNA levels of these

    enzymes were induced by RBPH, we decided to examine the expression and nuclear

    translocation of Nrf2. mRNA levels of Nrf2 appeared increased about 1.5-fold at 3 h after

    RBPH addition. At 8 h they had fallen to about 50 % the initial value, and remained low until

    the end of the experiment (Fig. 5A). The Nrf2 protein levels in total cell lysate also reached a

  • Biological and Pharmaceutical Bulletin Advance Publication

    maximum increase (about 9-fold) at 3 h, only to fall afterwards, correlating with mRNA

    expression (Fig. 5B). On the other hand, the nuclear Nrf2 protein levels began to increase at 3

    h, indicating that RBPH treatment induces the translocation of Nrf2 into the nucleus (Fig. 5C).

    The levels of Nrf2 in the cytosolic fraction were generally unchanged, although they

    increased slightly after 3 h.

    Protective effects of RBPH against cytotoxicity induced by oxidative stress Cells treated

    with RBPH, as well as untreated controls, were incubated with H2O2 or ethanol. Afterwards,

    cytotoxicity was evaluated by measuring the activity of released LDH in the medium.

    Treatment of control cells with 100 μM or 200 μM H2O2 1 h had a cytotoxicity of 10 % and

    25 %, respectively. In contrast, at the RBPH-pretreated cells cytotoxicity had been decreased

    to less than 5 %, and this reduction was statistically significant fashion. Pretreatment of RB

    protein without hydrolysis did not show the cytoprotective effect (Fig. 6A, 6B). Ethanol

    treatment was also included in the study, as ROS are known to be produced during ethanol

    metabolism, especially through the action of CYP 2E1.26) Pre-treatment of control cultures

    with 200 mM or 500 mM of ethanol caused cytotoxicity levels of 20 % and 24 %,

    respectively, whereas pre-treatment with RBPH reduced these levels to less than 1% at 200

    mM, and to 7% at 500 mM (Fig. 6C).

    DISCUSSION

    In this study, we demonstrated that RBPH increases intracellular GSH levels in a dose-

    and time- dependent manner (Fig. 2), whereas soybean protein hydrolysate or collagen

    peptide did not exhibit a similar effect (Fig. 1). The mRNA levels of both γ-GCS subunits

    began to increased at 3 h after addition (Fig. 3). In addition to γ-GCS, HO-1 and NQO1

  • Biological and Pharmaceutical Bulletin Advance Publication

    were induced, suggesting that RBPH activated the Nrf2 pathway (Fig. 4). Nrf2 expression

    temporarily increased at 3 h after RBPH treatment. Moreover, the increased Nrf2 nuclear

    accumulation started at 3 h after the addition of RBPH, which corresponded to the expression

    of anti-oxidant enzymes (Fig. 5). As our results indicated that RBPH was able to activate the

    Nrf2 pathway, we assumed that it might protect against cell damage caused by oxidative

    stress. The hypothesis was verified because pre-treatment of cells with RBPH did offer

    significant protection against damage brought about by H2O2 and ethanol (Fig. 6). HO-1

    converts heme into biliverdin, releasing free iron and carbon monoxide. Biliverdin is rapidly

    metabolized to the antioxidant bilirubin.27) NQO1 detoxifies quinones, which protects the cell

    against oxidative stress, and reduces the antioxidants vitamin E and coenzyme Q to their

    active form.28) In addition to the induction of HO-1 and NQO1, other anti-oxidant enzymes

    that are regulated by Nrf2, including catalase and SOD, could contribute to the protection of

    the cell from oxidative stress. Taken together, our results strongly suggest that RBPH may be

    able to suppress oxidative stress in cells not only through the up-regulation of GSH

    biosynthesis, but also by increasing of expression of other antioxidant enzymes.

    Even though our results show that RBPH increase on GSH levels takes place through a

    mechanism related to the Nrf2 pathway, this response is relatively slow. Furthermore, the

    levels of the γ-GCS protein actually decreased during the first few hours of treatment,

    before the induction. In LC2 cells, intracellular GSH levels were found to increase at 6 h after

    exposure to 2,3-dimethoxy-1,4-naphthoquinone, which is known to generate ROS,29) whereas

    ionizing radiation and TNF-α have been shown to increase intracellular GSH levels with a

    peak appearing at 6 or 3 h.30) A 3-h treatment with pyrrolidine dithiocarbamate induces γ-

    GCS expression by an Nrf2-associated mechanism.15) After treatment with tert-

    butylhydroquinone31), the nuclear import of Nrf2 started as early as 1 h and Nrf2 was present

  • Biological and Pharmaceutical Bulletin Advance Publication

    in the nucleus between 1 h and 4 h. After stimulation with dieckol, a hexamer of

    phloroglucinol with known anti-oxidant activity, the nuclear translocation of Nrf2 was

    induced at 1 h.32) Changes in GSH levels, γ-GCS levels, and Nrf2 nuclear translocation to

    these oxidants and anti-oxidants are faster than those to RBPH. These observations suggest

    that activation of the Nrf2 pathway by RBPH is mediated through an additional step: RBPH

    might induce a weak oxidative stress in cells, such as an imbalance of the GSH/GSSG ratio,

    which subsequently triggered the activation of the Nrf2 pathway. However, the mechanism of

    the RBPH-induced decrease of γ -GCS levels is unclear. As it has been reported that

    inhibition of NO synthesis leads to a decrease in GSH levels through downregulation of γ-

    GCS expression,33) a rational hypothesis would be that RBPH might inhibit NO synthesis.

    Several studies have demonstrated the anti-oxidative effects of phytochemicals isolated

    from dietary sources.34, 35) Protein hydrolysates derived from soy, egg, milk, whey etc., have

    also been reported to exhibit anti-oxidative activity.36) Both, scavenging of ROS and free

    radicals as well as sequestering pro-oxidative metals through chelation, have been described

    as the primary mechanisms of anti-oxidative activity. To this day, only a few peptides have

    been shown to exert anti-oxidative action via increasing the expression levels or the activity

    of anti-oxidative enzymes. Egg-derived peptides have been reported to increase the

    intracellular GSH levels and upregulate anti-oxidative enzymes in Caco-2 cells37) whereas,

    peptides derived from chickpeas have been reported to increase the expression of anti-

    oxidative enzymes including γ-GCS, HO-1, NQO1, and Nrf2, in Caco-2 and HT-29 cells.38)

    Thus, RBPH is one of the few anti-oxidants that exert their anti-oxidative activity by

    regulating the oxidative defense system.

    Many studies have shown that oxidative stress is related to the progression or the

    aggravation of diseases including cancer, neurodegenerative disorders, and diabetes.8-11)

  • Biological and Pharmaceutical Bulletin Advance Publication

    Supplementation with exogenous GSH has been suggested as a novel treatment for

    Parkinson’s disease,39) psychiatric disorders,40) and diabetes.41) In our previous study, the

    apparent molecular weight of RBPH was estimated around 300 Da, corresponding with the

    molecular weight of di- or tripeptides, and inhibitory peptides of DPP-IV from RBPH were

    successfully identified.5) In this case, even though the sequences of the bioactive peptides in

    RBPH have not yet been identified, our results suggest that novel anti-oxidative agents

    among them may prevent the development and progression of disorders caused by oxidative

    stress such as those mentioned above.

    In summary, RBPH increased intracellular GSH levels and induced the expression levels

    of anti-oxidative enzymes such as γ-GCS, HO-1 and NQO1 through an activation of the

    Nrf2 pathway. In addition, RBPH provided the cytoprotective effect against oxidative stress.

    Acknowledgements We would like to acknowledge the contribution Mr. Yoshikazu Inoue,

    who was one of our colleagues and passed away suddenly in 2015. This project was

    financially supported by the Iijima Memorial Foundation for the promotion of food science

    and technology.

    Conflict of interest Chie Moritani, Kayoko Kawakami and Seiji Tsuboi received a research

    grant from Satake Corporation. Akiko Fujita and Koji Kawakami are employees of Satake

    Corporation. Tadashi Hatanaka has no conflict of interest.

  • Biological and Pharmaceutical Bulletin Advance Publication

    REFERENCES

    1) Bandyopadhyay K, Misra G, Ghosh S. Preparation and characterization of protein

    hydrolysates from Indian defatted rice bran meal. J. Oleo Sci., 57, 47–52 (2008).

    2) Parrado J, Miramontes E, Jover M, Gutierrez JF, Terán LC, Bautista J. Preparation of a

    rice bran enzymatic extract with potential use as functional food. Food Chem., 98, 742–

    748 (2006).

    3) Islam MS, Nagasaka R, Ohara K, Hosoya T, Ozaki H, Ushio H, Hori M. Biological

    abilities of rice bran-derived antioxidant phytochemicals for medical therapy. Curr. Top.

    Med. Chem., 11, 1847–1853 (2011).

    4) Min B, McClung AM, Chen MH. Phytochemicals and antioxidant capacities in rice

    brans of different color. J. Food Sci., 76, C117–126 (2011).

    5) Hatanaka T, Inoue Y, Arima J, Kumagai Y, Usuki H, Kawakami K, Kimura M,

    Mukaihara T. Production of dipeptidyl peptidase IV inhibitory peptides from defatted

    rice bran. Food Chem., 134, 797–802 (2012).

    6) Kannan A, Hettiarachchy NS, Lay JO, Liyanage R. Human cancer cell proliferation

    inhibition by a pentapeptide isolated and characterized from rice bran. Peptides, 31,

    1629–1634 (2010).

    7) Zhang H, Yokoyama WH, Zhang H. Concentration-dependent displacement of

    cholesterol in micelles by hydrophobic rice bran protein hydrolysates. J. Sci. Food

    Agric., 92, 1395–1401 (2012).

    8) Trachootham D, Alexandre J, & Huang P. Targeting cancer cells by ROS-mediated

    mechanisms: a radical therapeutic approach? Nat. Rev. Drug Discov., 8, 579–591 (2009).

    9) Evans JL, Goldfine ID, Maddux BA, Grodsky GM. Oxidative stress and stress-activated

    signaling pathways: a unifying hypothesis of type 2 diabetes. Endocr. Rev., 23, 599–622

    (2002).

  • Biological and Pharmaceutical Bulletin Advance Publication

    10) Lefer DJ, Granger DN. Oxidative stress and cardiac disease. Am. J. Med., 109, 315–323

    (2000).

    11) Shukla V, Mishra SK, Pant HC. Oxidative stress in neurodegeneration. Adv. Pharmacol.

    Sci., 2011, Article ID 572634 (2011).

    12) Birben E, Sahiner UM, Sackesen C, Erzurum S, Kalayci O. Oxidative stress and

    antioxidant defense. World Allergy Organ J., 5, 9–19 (2012).

    13) Griffith OW, Mulcahy RT. The enzymes of glutathione synthesis: γ-glutamylcysteine

    synthetase. Adv. Enzymol. Relat. Areas Mol. Biol., 73, 209–267 (1999).

    14) Itoh K, Tong KI, Yamamoto M. Molecular mechanism activating Nrf2-Keap1 pathway

    in regulation of adaptive response to electrophiles. Free Radical Biol. Med., 36, 1208–

    1213 (2004).

    15) Wild AC, Moinova HR, Mulcahy RT. Regulation of γ-glutamylcysteine synthetase

    subunit gene expression by the transcription factor Nrf2. J. Biol. Chem., 274, 33627–

    33636 (1999).

    16) Kobayashi M, Yamamoto M. Nrf2-Keap1 regulation of cellular defense mechanisms

    against electrophiles and reactive oxygen species. Adv. Enzyme Regul., 46, 113–140

    (2006).

    17) Saadi S, Saari N, Anwar F, Hamid AA, Ghazali MH. Recent advances in food

    biopeptides: production, biological functionalities and therapeutic applications.

    Biotechnol. Adv., 333, 80–116 (2015).

    18) Walther B, Sieber R. Bioactive proteins and peptides in foods. Int. J. Vitam. Nutr. Res.,

    81, 181–192 (2011).

    19) Toyo’oka T, Uchiyama S, Saito Y. Simultaneous determination of thiols and disulfides

    by high-performance liquid chromatography with fluorescence detection. Anal. Chim.

    Acta, 205, 29–41 (1988).

  • Biological and Pharmaceutical Bulletin Advance Publication

    20) Matsumoto S, Teshigawara M, Tsuboi S, Ohmori S. Determination of glutathione and

    glutathione disulfide in biological samples using acrylonitrile as a thiol-blocking reagent.

    Anal. Sci., 12, 91–95 (1996).

    21) Andringa KK, Coleman MC, Aykin-Burns N, Hitchler MJ, Walsh SA, Domann FE,

    Spitz DR. Inhibition of glutamate cysteine ligase activity sensitizes human breast cancer

    cells to the toxicity of 2-deoxy-D-glucose. Cancer Res., 66, 1605–1610 (2006).

    22) Chen N, Shao W, Lv P, Zhang S, Chen Y, Zhu L, Lu Y, Shen Y. Hemin-induced Erk1/2

    activation and heme oxygenase-1 expression in human umbilical vein endothelial cells.

    Free Radic. Res., 41, 990–996 (2007).

    23) Wu TY, Khor TO, Saw CL, Loh SC, Chen AI, Lim SS, Park JH, Cai L, Kong AN. Anti-

    inflammatory/Anti-oxidative stress activities and differential regulation of Nrf2-

    mediated genes by non-polar fractions of tea Chrysanthemum zawadskii and licorice

    Glycyrrhizauralensis. AAPS J., 13, 1–13 (2011).

    24) Ramyaa P, Krishnaswamy R, Padma VV. Quercetin modulates OTA-induced oxidative

    stress and redox signalling in HepG2 cells - up regulation of Nrf2 expression and down

    regulation of NF-κB and COX-2. Biochim. Biophys. Acta, 1840, 681–692 (2014).

    25) Meister A. Glutathione metabolism and its selective modification. J. Biol. Chem., 263,

    17205-17208 (1988).

    26) Das SK, Vasudevan DM. Alcohol-induced oxidative stress. Life Sciences, 27, 177–187

    (2007).

    27) Ryter SW, Otterbein LE, Morse D, Choi AM. Heme oxygenase/carbon monoxide

    signaling pathways: regulation and functional significance. Mol. Cell. Biochem., 234-

    235, 249-263 (2002).

  • Biological and Pharmaceutical Bulletin Advance Publication

    28) Siegel D, Bolton EM, Burr JA, Liebler DC, Ross D. The reduction of alpha-

    tocopherolquinone by human NAD(P)H: quinone oxidoreductase: the role of alpha-

    tocopherolhydroquinone as a cellular antioxidant. Mol. Pharmacol. 52, 300–305 (1997).

    29) Shi MM, Kugelman A, Iwamoto T, Tian L, Forman HJ. Quinone-induced oxidative

    stress elevates glutathione and induces γ-glutamylcysteinesynthetase activity in rat

    lung epithelial L2 cells. J. Biol. Chem., 269, 26512–26517 (1994).

    30) Kondo T, Higashiyama Y, Goto S, Iida T, Cho S, Iwanaga M, Mori K, Tani M, Urata Y.

    Regulation of γ-glutamylcysteinesynthetase expression in response to oxidative stress.

    Free Radic. Res., 31, 325–334 (1999).

    31) Jain AK, Bloom DA, Jaiswal AK. Nuclear import and export signals in control of Nrf2.

    J. Biol. Chem., 280, 29158–29168 (2005).

    32) Lee MS, Lee B, Park KE, Utsuki T, Shin T, Oh CW, Kim HR. Dieckol enhances the

    expression of antioxidant and detoxifying enzymes by the activation of Nrf2-MAPK

    signalling pathway in HepG2 cells. Food Chem., 17, 538-546 (2015)

    33) Kuo PC, Abe KY, Schroeder RA. Interleukin-1-induced nitric oxide production

    modulates glutathione synthesis in cultured rat hepatocytes. Am. J. Physiol., 271, C851–

    C862 (1996).

    34) Stefanson AL, Bakovic M. Dietary regulation of Keap1/Nrf2/ARE pathway: focus on

    plant-derived compounds and trace minerals. Nutrients, 6, 3777–3801 (2014).

    35) Surh YJ. Cancer chemoprevention with dietary phytochemicals. Na.t Rev. Cancer, 3,

    768–780 (2003).

    36) Samaranayaka AGP, Li-Chan ECY. Food-derived peptidic antioxidants: A review of

    their production, assessment, and potential applications. J. Funct. Foods, 3, 229–254

    (2011).

    37) Shi Y, Kovacs-Nolan J, Jiang B, Tsao R, Mine Y. Peptides derived from eggshell

  • Biological and Pharmaceutical Bulletin Advance Publication

    membrane improve antioxidant enzyme activity and glutathione synthesis against

    oxidative damage in Caco-2 cells. J. Funct. Foods, 11, 571–580 (2014).

    38) Guo Y, Zhang T, Jiang B, Miao M, Mu W. The effects of an antioxidative pentapeptide

    derived from chickpea protein hydrolysates on oxidative stress in Caco-2 and HT-29 cell

    lines. J. Funct. Foods, 7, 719–726 (2014).

    39) Martin HL, Teismann P. Glutathione--a review on its role and significance in Parkinson's

    disease. FASEB J., 23, 3263–3272 (2009).

    40) Berk M, Ng F, Dean O, Dodd S, Bush AI. Glutathione: a novel treatment target in

    psychiatry. Trends Pharmacol. Sci., 29, 346-351 (2008).

    41) Trocino RA, Akazawa S, Ishibashi M, Matsumoto K, Matsuo H, Yamamoto H, Goto S,

    Urata Y, Kondo T, Nagataki S. Significance of glutathione depletion and oxidative

    stress in early embryogenesis in glucose-induced rat embryo culture. Diabetes, 44, 992–

    998 (1995).

  • Biological and Pharmaceutical Bulletin Advance Publication

    Figure legends

    Figure 1. Effects of hydrolysates of various proteins on the intracellular GSH levels in

    HepG2 cells. Cells were treated with 5 mg/ml of RB protein hydrolysate (RBPH), or soybean

    protein hydrolysate (Soybean), or collagen peptide (Collagen) (A), or RB protein without

    hydrolysis (RB/−) (B), for 24 h. Values are the means ± SD (n=3). *p < 0.05, **p < 0.01 vs.

    control group.

    Figure 2. Dose- and time-dependent dependent effects of RB protein hydrolysate on the

    intracellular GSH levels. HepG2 (A) or COS7 (B) cells were incubated with the indicated

    concentrations of RB protein hydrolysate for 24 h, followed by measurement of GSH levels.

    (C) Effects of RB protein hydrolysate on GSH/GSSG ratios. HepG2 cells were treated with

    the indicated concentrations of RB protein hydrolysate for 24 h. (D) Time-dependent effect of

    RB protein hydrolysate on GSH levels. Cells were treated with 5 mg/ml of RB protein

    hydrolysate for the indicated times. Values are the means ± SD (n=3). *p < 0.05, ***p <

    0.001 vs. control group.

    Figure 3. Effects of RB protein hydrolysate on γ-GCS expression levels. mRNA levels of

    heavy (A) and light (B) subunits were estimated by real-time PCR analysis after HepG2 cells

    were treated with 5 mg/ml RB protein hydrolysate for the indicated times. Values are the

    means ± SD (n=3). *p < 0.05 vs. control. Protein levels of each subunit (C, D) were analyzed

    by western blotting using corresponding antibodies. Values are the means ± SD (n=3).

    Figure 4. Effects of RB protein hydrolysate on NQO1 and HO-1 expression levels. mRNA

    levels of NQO1 (A) and HO-1 (B) were estimated by real-time PCR analysis after HepG2

    cells were treated with 5 mg/ml RB protein hydrolysate for the indicated times. (C) HO-1

  • Biological and Pharmaceutical Bulletin Advance Publication

    protein levels were analyzed by western blotting using corresponding antibodies. Values are

    the means ± SD (n=3). *p < 0.05, **p < 0.01 vs. control group.

    Figure 5. Effects of RB protein hydrolysate on Nrf2 expression. (A) mRNA levels of Nrf2

    were estimated by real-time PCR analysis after HepG2 cells were treated with 5 mg/ml RB

    protein hydrolysate for the indicated times. Values are the means ± SD (n=3). Nrf2 protein

    levels in cell lysate (B) and the cytosolic or nuclear fraction (C) were analyzed by western

    blotting using corresponding antibodies. Values are the means ± SD (n=3). *p < 0.05 and **p

    < 0.01 vs. control group.

    Figure 6. Protective effect of RB protein hydrolysate against cell damage caused by

    oxidative stress in HepG2 cells. (A) Cells were treated with 5 mg/mL RB protein with

    (RBPH) or without hydrolysis (RB/−) for 24 h and subsequently exposed to 200 µM H2O2 for

    1 h (n=4). (B, C) Cells treated with 5 mg/mL RB protein hydrolysate for 24 h, as well as

    untreated controls, were exposed to various concentrations of H2O2 for 1 h (n=3-4) and

    various concentrations of ethanol for 24 h (n=3-4). The cytotoxicity was determined by

    measurement of LDH activity released from damaged cells into the medium. Values are the

    means ± SD. *p < 0.05 and **p < 0.01 vs. the group that was not pre-treated with RB protein

    hydrolysate.

  • 0

    100

    200

    300

    400

    500

    600To

    tal G

    SH

    (nm

    ol/m

    g pr

    otei

    n)*

    Control CollagenRBPH Soybean

    Figure 1. Effects of hydrolysates of various proteins on the intracellular GSH levels in HepG2 cells.

    0

    50

    100

    150

    200

    Control RBPH RB/-Tot

    al G

    SH

    (nm

    ol/m

    g pr

    otei

    n)A

    B

    **

    Biological and Pharmaceutical Bulletin Advance Publication

  • *

    0

    50

    100

    150

    200

    250

    300

    0 1.25 2.5 5Tot

    al G

    SH

    (nm

    ol/m

    g pr

    otei

    n)

    RB protein hydrolysate (mg/mL)

    AHepG2

    0

    50

    100

    150

    200

    250

    300

    Tota

    l GSH

    (nm

    ol/m

    g pr

    otei

    n)

    RB protein hydrolysate (mg/mL)0 2.5 5

    *

    ***

    BCOS7

    0

    20

    40

    60

    80

    100

    0 1.25 2.5 5

    GSH

    /GSSG

    RB protein hydrolysate(mg/mL)

    0

    20

    40

    60

    80

    0 10 20

    Tota

    lGSH

    (nm

    ol/m

    g pr

    otei

    n)

    Time (h)

    *** ***C D

    Figure 2. Dose- and time-dependent dependent effects of RB protein hydrolysate on the intracellular GSH levels.

    Biological and Pharmaceutical Bulletin Advance Publication

  • C

    0.0

    0.5

    1.0

    1.5

    2.0

    0 3 8 24

    Rel

    ativ

    e ex

    pres

    sion

    leve

    l

    Time (h)

    *

    A γ-GCSh B

    0.0

    0.5

    1.0

    1.5

    2.0

    0 3 8 24Rel

    ativ

    e ex

    pres

    sion

    leve

    lTime (h)

    *

    γ-GCSl

    D

    0.0

    0.5

    1.0

    1.5

    0 1 3 8 24

    γ-G

    CSh/

    β-ac

    tin (

    fold

    )

    Time (h)

    0 1 3 8 24 (h)

    β-actin

    γ-GCSh0 1 3 8 24 (h)

    β-actin

    γ-GCSl

    0.0

    0.5

    1.0

    1.5

    2.0

    0 1 3 8 24

    γ-G

    CSl/

    β-ac

    tin (

    fold

    )

    Time (h)

    Figure 3. Effects of RB protein hydrolysate on γ-GCS expression levels.

    Biological and Pharmaceutical Bulletin Advance Publication

  • 0 1 3 8 24 (h)

    0.0

    0.5

    1.0

    1.5

    2.0

    2.5

    0 3 8 24Rel

    ativ

    e ex

    pres

    sion

    leve

    l

    Time (h)

    HO-1

    ** **

    BA

    0.0

    0.5

    1.0

    1.5

    2.0

    0 3 8 24

    Rel

    ativ

    e ex

    pres

    sion

    leve

    l

    Time (h)

    NQO1

    ** *

    HO-1

    β-actin

    C

    0.0

    0.5

    1.0

    1.5

    2.0

    2.5

    3.0

    3.5

    0 1 3 8 24

    HO

    -1/β

    -act

    in (

    fold

    )

    Time (h)

    **

    *

    **

    Figure 4. Effects of RB protein hydrolysate on NQO1 and HO-1 expression levels.

    Biological and Pharmaceutical Bulletin Advance Publication

  • 0 1 3 8 24 (h)

    0 1 3 8 24 (h)

    0.0

    0.5

    1.0

    1.5

    2.0

    0 3 8 24

    Rel

    ativ

    e ex

    pres

    sion

    leve

    l

    Time (h)

    Nrf2

    Nrf2

    Lamin B

    β-actin

    A

    C

    B

    **

    **

    0

    2

    4

    6

    8

    10

    12

    0 1 3 8 24

    Tota

    l Nrf

    2/β-

    actin

    (fo

    ld)

    Time (h)

    Figure 5. Effects of RB protein hydrolysate on Nrf2 expression.

    Nrf2

    β-actinCytosol

    Nucleus0

    2

    4

    6

    8

    0 1 3 8 24Rel

    ativ

    e N

    rf2

    prot

    ein

    leve

    ls(f

    old)

    Time (h)

    Cytosol

    Nucleus*

    **

    **

    Biological and Pharmaceutical Bulletin Advance Publication

  • 0

    10

    20

    30

    40

    0 50 100 150 200

    Cyt

    otox

    icity

    (%)

    H₂O₂ (µM)

    0%

    10%

    20%

    30%

    40%

    0 200 400 600

    Cyt

    otox

    icity

    (%)

    Ethanol (mM)

    **

    Control

    RB protein hydrolysate

    Control

    RB protein hydrolysate40

    30

    20

    10

    0

    Cyt

    otox

    icity

    (%

    A

    Figure 6. Protective effect of RB protein hydrolysate against cell damage caused by oxidative stress in HepG2 cells.

    0

    10

    20

    30

    40

    Control RBPH RB/-

    Cyt

    otox

    icity

    (%

    **

    C

    B

    ***

    Biological and Pharmaceutical Bulletin Advance Publication

  • 本文献由“学霸图书馆-文献云下载”收集自网络,仅供学习交流使用。

    学霸图书馆(www.xuebalib.com)是一个“整合众多图书馆数据库资源,

    提供一站式文献检索和下载服务”的24 小时在线不限IP

    图书馆。

    图书馆致力于便利、促进学习与科研,提供最强文献下载服务。

    图书馆导航:

    图书馆首页 文献云下载 图书馆入口 外文数据库大全 疑难文献辅助工具

    http://www.xuebalib.com/cloud/http://www.xuebalib.com/http://www.xuebalib.com/cloud/http://www.xuebalib.com/http://www.xuebalib.com/vip.htmlhttp://www.xuebalib.com/db.phphttp://www.xuebalib.com/zixun/2014-08-15/44.htmlhttp://www.xuebalib.com/

    Anti-oxidative Activity of Hydrolysate from Rice Bran Protein in HepG2 Cells.学霸图书馆link:学霸图书馆