You are on page 1of 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/284713715

Cytoprotective Effect of Makgeolli Lees on Paraquat Induced Oxidative Stress


in A549 Cells via Activation of NRF2 and Antioxidant Genes

Article  in  Journal of Microbiology and Biotechnology · November 2015


DOI: 10.4014/jmb.1510.10093

CITATIONS READS

5 260

3 authors:

Miso Jeon Naimur Rahman


Soonchunhyang University Purdue University
11 PUBLICATIONS   82 CITATIONS    12 PUBLICATIONS   82 CITATIONS   

SEE PROFILE SEE PROFILE

Yong-Sik Kim
College of Medicine, Soonchunhyang University
84 PUBLICATIONS   1,886 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Cancer cell apoptosis View project

The function of receptor-associated protein 80 (RAP80) View project

All content following this page was uploaded by Miso Jeon on 15 March 2016.

The user has requested enhancement of the downloaded file.


jmb
J. Microbiol. Biotechnol. (2016), 26(2), 277–286
http://dx.doi.org/10.4014/jmb.1510.10093 Research Article

Review

Cytoprotective Effect of Makgeolli Lees on Paraquat Induced Oxidative


Stress in A549 Cells via Activation of NRF2 and Antioxidant Genes
Miso Jeon1, Naimur Rahman1,2, and Yong-Sik Kim1,2*
1
Department of Microbiology, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea
2
Institute of Tissue Engineering, College of Medicine, Soonchunhyang University, Cheonan 31151, Republic of Korea

Received: October 29, 2015


Revised: November 11, 2015 Makgeolli lees (ML) has several physiological effects such as antioxidant, antidiabetic, and
Accepted: November 12, 2015
anticancer properties, but its biological functions have not been determined definitively. Here,
we tested whether ML has a cytoprotective effect on paraquat (PQ)-induced oxidative stress in
the human lung carcinoma cell line A549. At 0.1 mg/ml ML, viability of PQ-exposed A549
First published online cells was restored by 12.4%, 18.5%, and 48.6% after 24, 48, and 72 h, respectively. ML also
November 24, 2015 reduced production of the intracellular reactive oxygen species (ROS) that were generated by
*Corresponding author PQ treatment. Further experiments revealed that ML treatment enhanced the expression and
Phone: +82-41-570-2413; nuclear translocation of nuclear factor erythroid 2–related factor 2 (NRF2) as well as ARE-GFP
Fax: +82-41-575-2412;
E-mail: yongsikkim@sch.ac.kr
reporter activity. ML treatment also effectively increased the expression of NRF2’s target
genes NAD(P)H dehydrogenase quinone 1 (NQO1) and heme oxygenase 1 (HO-1). Moreover,
we found that expression of cytoprotective genes, including glutathione peroxidases (GPXs),
superoxide dismutase (SOD1), catalase (CAT), peroxiredoxin 3 (PRDX3), and peroxiredoxin 4
(PRDX4), was greatly enhanced by treatment with ML during PQ exposure. Taken together,
the data suggest that treatment of PQ-exposed A549 cells with ML ameliorates cytotoxicity
through induction of NRF2 expression and its target genes HO-1, NQO1, and other antioxidant
pISSN 1017-7825, eISSN 1738-8872 genes. Thus, ML may serve as a functional food applicable to ROS-mediated human diseases.
Copyright © 2016 by
The Korean Society for Microbiology Keywords: Makgeolli lees, paraquat, oxydative stress, NRF2, antioxidant response element, A549
and Biotechnology

Introduction such as wine, cider, and beer [30]. During the brewing of
makgeolli by means of nuruk or koji (a Korean fermentation
Wines and their derivatives can delay tumor onset [8], starter), a massive amount of makgeolli lees (ML) is formed
protect from coronary heart disease [28], and reduce [20]. At present, ML is used as a cheap animal feed and
susceptibility to LDL oxidation and aggregation [12]. For fertilizer. As an industrial waste, ML is considered a cause
instance, unlike other alcoholic beverages, makgeolli is of environmental pollution if not properly processed [2, 7].
regarded as a functional food because of its high nutritious Therefore, it would be useful to find an alternate use for
value and a wide range of physiological functions; its ML.
consumption has markedly increased in Korea and other ML that is used in this study was extracted from makgeolli
countries [6]. residue with ethanol at the final yield of 6.62% and
Makgeolli is a traditional and popular rice wine in Korea. contains 1.0 ± 0.1 (mean ± SD) mg/g total flavonoids, 35.0
It contains a relatively low concentration of alcohol (6–8%), ± 1.1 mg/g polyphenols, 394.1 ± 17.0 mg/g total sugar, and
and is rather rich in proteins, minerals, vitamins, dietary 367.8 ± 15.0 mg/g reducing sugar [19]. Choi et al. [5]
fiber, organic acids, and uncharacterized bioactive compounds demonstrated that ML still contains considerable amounts
[11]. Makgeolli contains the byproduct “lees,” which is of dietary fiber, proteins, minerals, vitamins, and organic
formed during fermentation and aging of alcoholic drinks acids. It has been also reported that ML has antioxidant,

February 2016 ⎪ Vol. 26 ⎪ No. 2


278 Jeon et al.

antidiabetic, antiobesity, anticancer, and antihypertensive Materials and Methods


properties [6]. In addition, it is known that ML contributes
to a reduction in blood cholesterol and the incidence of Reagents
atherosclerosis [15, 21]. Therefore, ML may be an inexpensive ML was obtained from Kooksoondang Brewery Co., Ltd.
nutraceutical supplement for many human diseases. Despite (Seongnam, South Korea). Paraquat dichloride (1,1’-dimethyl-4,4’-
the high nutritious value and various useful biological bipyridinium dichloride; PQ), 3-(4,5-dimethylthiazol-2-yl)-2,5-
diphenyltetrazolium bromide (MTT), and trypan blue dye, 2’,7’-
effects, practical applications of ML have so far been limited.
dichlorofluorescein diacetate (DCF-DA) were purchased from
Paraquat (1,1’-dimethyl-4,4’-bipyridinium dichloride; PQ)
Sigma-Aldrich (St. Louis, MO, USA). Hydrogen peroxide was
is a well-known potent herbicide that is banned in many
obtained from Junsei Chemicals Co., Ltd. (Tokyo, Japan).
countries but is still used in developing countries owing to Lipofectamine 2000 was purchased from Invitrogen (Life
its low cost and high effectiveness. PQ is known to exert its Technologies, Coventry, UK). DMEM/F12 (Dulbecco’s modified
toxic effects via oxidative stress, although the exact Eagle’s medium supplemented with nutrient mixture F-12 [Ham])
molecular mechanism is still unknown [9]. On the other and the Opti-MEM serum-free medium were purchased from
hand, PQ is a useful inducer of reactive oxygen species Gibco (Grand Island, NY, USA). Fetal bovine serum was purchased
(ROS); however, chronic exposure to PQ is associated with from Atlas Biologicals (Fort Collins, CO, USA). A penicillin-
liver damage, kidney failure, and Parkinsonian lesions [4]. streptomycin solution was purchased from Hyclone Laboratories,
Moreover, poisoning with PQ in mammals and other Inc. (South Logan, NY, USA). Antibodies against NRF2, HO-1,
animals mainly causes serious lung injury such as bronchial and NQO1 were purchased from Santa Cruz Biotechnology (Santa
Cruz, CA, USA), and anti-β-actin, anti–peroxiredoxin 3, anti–
or alveolar hemorrhage, interstitial edema, hypoxemia, and
peroxiredoxin 4, and anti–superoxide dismutase 1 antibodies were
leukocyte infiltration via production of intracellular ROS
purchased from Abcam (Cambridge, MA, USA). Horseradish
[3].
peroxidase–conjugated anti–rabbit immunoglobulin G (IgG)
Nuclear factor erythroid 2–related factor 2 (NRF2) is a antibody and anti–mouse IgG antibody were also obtained from
pivotal transcription factor in ROS-mediated pathophysiological Santa Cruz Biotechnology.
processes and is located in the cytoplasm, where it is
degraded by Kelch-like ECH-associated protein (Keap1). Cell Culture
Under oxidative conditions caused by one of many oxidants A human lung carcinoma cell line, A549, was purchased from
or electrophilic stimuli, NRF2 dissociates from inhibitory ATCC (Manassas, VA, USA) and grown in DMEM/F12 containing
complexes and is translocated to the nucleus, where it serves 10% (v/v) of heat-inactivated fetal bovine serum and 1% (v/v) of
as a transcription factor through binding to the antioxidant the penicillin-streptomycin solution (10,000 U/ml penicillin and
response element (ARE) in the promoter regions of target 10,000 µg/ml streptomycin) in a humidified atmosphere (incubator)
containing 5% of CO2 at 37°C.
genes [34]. It has been well established that transcriptional
regulation by NRF2 via binding to ARE is a crucial event in
Preparation of ML
modulation of oxidative stress and cytoprotection against
The procedure for preparation of ML was described previously
pro-oxidant stimuli. If oxidative stress is present, NRF2 by Kim et al. [19]. Briefly, Makgeolli residue was added to 10
binds to the ARE in the promoter region of phase II volumes of 95% ethanol and incubated at room temperature for
detoxification enzymes and cytoprotective genes and 24 h; then the supernatant was collected and this extraction
activates such enzymes and antioxidant genes, including process was repeated three times. The resulting ethanol extract
glutathione peroxidases (GPXs), NAD(P)H dehydrogenase was dried at 55oC and concentrated in an evaporator under reduced
quinone 1 (NQO1), and heme oxygenase 1 (HO-1) [26]. In pressure (Eyela Rotary evaporator N-1000; Tokyo Rikakikai Co.,
addition, many antioxidant enzymes, such as superoxide Ltd., Japan) to obtain a powder; the final yield was 6.62%. The ML
dismutase (SOD), catalase (CAT), glutathione peroxidase powder was then dissolved in dimethyl sulfoxide (DMSO) and
(GPX), and peroxiredoxin (PRDX) have been reported to stored at -20oC for subsequent use.
inactivate ROS directly; the activation of these enzymes is
Cell Viability Assay
regulated by NRF2 [22, 33].
A549 cells were seeded in a 96-well plate at the density of 104/well
The aim of this study was to test whether ML has a
and grown overnight to ~80% confluence. The cells were then
cytoprotective effect on PQ-induced oxidative stress, and if incubated with various doses of ML or PQ for indicated periods.
so, whether this effect is mediated by activation of the After completion of the incubation with ML, PQ, or ML with PQ,
NRF2–ARE detoxification axis. an MTT solution was added to each well of the 96-well plate at the

J. Microbiol. Biotechnol.
ML Protects PQ-Exposed A549 Cells 279

final concentration of 0.5 mg/ml and incubated for 4 h at 37°C. construct constitutively expressing GFP, was purchased from
After incubation with MTT, the resulting formazan crystals were Qiagen. Briefly, cells were seeded at a density of 105/well in a 24-
dissolved in 100 µl of DMSO, and absorbance was measured on a well plate and grown overnight to ~70–80% confluence; then, the
Victor X3 multilabel reader (Perkin Elmer, Waltham, MA, USA) at cells were transiently transfected with various ARE-GFP reporter
590 nm wavelength. constructs. The construct constitutively expressing GFP was used to
visually verify transfection efficiency. The transfection experiments
Measurement of Intracellular ROS Production were performed using the Lipofectamine 2000 reagent. After 24 h
This analysis was conducted by a DCF-DA assay. In brief, A549 of the transfection, the cells were incubated with PQ, ML, or PQ
cells were seeded in a black 96-well flat-bottom plate at a density with ML for 24 h. Then, fluorescence microscopy was used to
of 5 × 104/well and grown overnight to ~90% confluence. Next, measure the GFP intensity. Fluorescence intensity was analyzed
the cells were incubated with 100 µl of 25 µM DCF-DA in 1× PBS by ImageJ software (National Institutes of Health, Bethesda, MD,
for 30 min followed by two washes with 1× PBS. After that, the USA).
cells were incubated with 0.1 mg/ml ML, 0.2 mM PQ, or in
combination, and fluorescence intensity of the formed DCF was Whole-Cell Protein Extraction
measured on the Victor X3 multi plate reader, at excitation and A549 cells were seeded in a 60 mm cell culture dish at the
emission wavelengths of 485 and 535 nm, respectively, for 30 min density of 4 × 105/well and incubated with various doses of ML,
in every 5 min. PQ, or ML with PQ for various periods. The cells were harvested
by scraping and washed with 1×PBS; then total protein was
Quantitative Real-Time Reverse-Transcriptase PCR (qRT-PCR) extracted with RIPA lysis buffer containing a protease inhibitor
A549 cells were seeded in a 6-well plate at the density of 4 × cocktail, 2 mM phenylmethylsulfonyl fluoride, and 1 mM sodium
105/well and grown overnight to ~90% confluence. Then, the cells orthovanadate (Santa Cruz Biotechnology). The protein concentration
were incubated with different doses of ML, PQ, or ML with PQ for was determined using the BCA protein assay kit (Pierce, Rockford,
indicated periods. After that, the cells were washed with 1× cold IL, USA).
PBS. Total RNA was isolated with an RNA extraction kit (Qiagen,
Valencia, CA, USA), and the RNA quality was analyzed using a Preparation of Nuclear Extracts
scandrop spectrophotometer (Analytik Jena AG; Jena, Germany). Nuclear extracts were prepared according to a nuclear extraction
A total of 1 µg of RNA was used to synthesize cDNA by means of kit (Active Motif, Carlsbad, CA, USA). Briefly, media were
the Maxime RT PreMix kit (Intron Biotechnology, Seoul, Korea), aspirated from the cell culture dishes, and the cells were washed
and the reaction was run in a Veriti 96-Well Thermal Cycler twice with 1× PBS containing phosphatase inhibitors. The cells
(Applied Biosystems, Singapore). Quantitative real-time PCR was were then scraped and centrifuged at 200 ×g for 5 min at 4oC. The
performed by means of the iQ SYBR Green Supermix Kit (Bio- cell pellets were gently resuspended in a 1× hypotonic solution
Rad, Singapore) on a CFX96 Real-Time PCR detection system containing a detergent, and homogenized by pipetting up and
(Bio-Rad). The primer sequences are listed in Table 1. The down several times, after which the samples were incubated for
expression data were normalized to GAPDH. 15 min on ice. The cytoplasmic fraction was separated by
centrifuging the homogenized pellet at 14,000 ×g for 1 min at 4oC.
ARE Reporter Assay After that, the remaining nuclear pellet was resuspended in lysis
A green fluorescent protein (GFP)-based ARE reporter assay buffer containing 10 mM dithiothreitol and protease inhibitors,
kit, including an inducible transcription factor–responsive GFP incubated for 30 min on ice on a rocking platform at 150 rpm,
reporter, a negative control (in which GFP expression is controlled vortexed for 30 sec at the maximal setting, and centrifuged at
by a minimal promoter), and a positive control containing a 14,000 ×g for 10 min at 4oC. The nuclear extracts were collected

Table 1. qRT-PCR primer sets for this study.


Gene Sequences Amplicon sizes (bp)
Nrf2 (F) 5’-GCGACGGAAAGAGTATGAC-3’ 99
Nrf2 (R) 5’-GTTGGCAGATCCACTGGTTT-3’
HO-1 (F) 5’-GCAACCCGACAGCATGC-3’ 245
HO-1 (R) 5’-TGCGGTCGAGCTCTTCTG-3’
NQO1(F) 5’-CGCAGACCTTGTGATATTCCAG-3’ 249
NQO1 (R) 5’-CGTTTCTTCCATCCTTCCAGG-3’
GAPDH (F) 5’-TCCCATCACCATCTTCCA-3’ 380
GAPDH (R) 5’-CATCACGCCACAGTTTCC-3’

February 2016 ⎪ Vol. 26 ⎪ No. 2


280 Jeon et al.

and stored at -80oC until analysis. value < 0.05 were considered statistically significant.

Western Blot Analysis Results


Equal amounts of protein (50 µg) were loaded into each well of
a 4–20% sodium dodecyl sulfate polyacrylamide gradient gel (Mini- ML Protects Cytotoxicity of PQ-Exposed A549 Cells
PROTEAN Precast Gel; Bio-Rad) and separated by electrophoresis
To evaluate the effect of ML on A549 cell viability, we
and then transferred to a nitrocellulose membrane. After blocking
incubated the cells with different doses of ML (from 0.1 to
with 5% skim milk in Tris-buffered saline (TBS) containing 0.1%
1.0 mg/ml) for 24, 48, or 72 h, and then the MTT assay was
Tween 20, we incubated the membrane with primary antibodies
overnight at 4°C and HRP-conjugated secondary antibodies for 1 h
performed. Our results revealed that ML did not reduce
at room temperature. Immunoreactive proteins were visualized the viability of A549 cells but instead enhanced cell
by means of the chemiluminescent ECL assay (Advansta, Melno viability at the concentration of 0.1 mg/ml even after a
Park, CA, USA). Skim milk and primary and secondary antibodies prolonged incubation period, such as 72 h (Fig. 1A).
were diluted in 1× TBS containing 0.1% Tween 20. All washing Because ML showed a beneficial effect on cell viability, we
steps involved 1× TBS containing 0.1% Tween 20, lasted 5 min, next assessed the cytoprotective effect of ML on PQ-
and were repeated three times. β-Actin served as an internal exposed A549 cells. Before this assay, we incubated A549
control. The western blot data were quantified in the ImageJ cells with PQ at different concentrations (0.1–0.7 mM) for
software. various periods (24, 48, or 72 h) to evaluate the cytotoxicity
of PQ toward A549 cells. In line with our previous studies,
Statistical Analysis
PQ strongly reduced the viability of A549 cells. As shown
All data were expressed as the mean ± standard deviation (SD).
in Fig. 1B, PQ exerted its cytotoxic effects in both dose- and
Significance of the differences among the treatment groups was
evaluated by Student’s t test and two-way analysis of variance,
time-dependent manners. Accordingly, we next cotreated
followed by the post hoc Bonferroni test. Differences with a p the cells with 0.1–1.0 mg/ml ML and 0.2 mM PQ (the

Fig. 1. Protective effect of makgeolli lees (ML) on paraquat (PQ)-induced cytotoxicity toward A549 cells.
(A) Effects of ML (0.1–1.0 mg/ml) on A549 cells viability. (B) Effects of different doses of PQ (0.1–0.7 mM) on A549 cell viability. All cell treatments
were performed as indicated in the figure, and viability was assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)
assay. The data are presented as the mean ± SD of three independent experiments. *denotes significant differences between control and treatment
groups (*p < 0.05, **p < 0.01). (C) The protective effect of ML on PQ-treated A549 cells. *Statistically significant differences between groups “PQ
only” and “PQ plus ML cotreatment” (*p < 0.05).

J. Microbiol. Biotechnol.
ML Protects PQ-Exposed A549 Cells 281

standard lethal concentration) to analyze the effects of ML


on the PQ-induced cytotoxicity. As shown in Fig. 1C,
treatment with 0.2 mM PQ reduced cell viability by 16%,
43.5%, and 53.6% after 24, 48, and 72 h, respectively, but
cotreatment with 0.1 mg/ml ML significantly attenuated
the PQ-induced cytotoxicity by 12.4%, 18.5%, and 48.6%
after 24, 48, and 72 h, respectively. ML was somewhat less
effective at 0.25 mg/ml: this dose attenuated the PQ-
induced cytotoxicity by 11.87%, 25.5%, and 37.1% after 24,
48, and 72 h, respectively. Thus, our data suggested that 0.1
and 0.25 mg/ml were effective doses of ML for protection
against PQ-induced cytotoxicity. We chose 0.1 mg/ml ML
for evaluation of the cytoprotection against PQ in further
experiments. Fig. 2. Makgeolli lees (ML) reduces the paraquat (PQ)-induced
intracellular production of reactive oxygen species (ROS).
ML Ameliorates PQ-Induced Oxidative Stress by Scavenging A549 cells were incubated with 100 µM H2O2, 0.2 mM PQ, and 0.1 mg/ml
Intracellular ROS ML alone or in combination with 0.2 mM PQ for 30 min. Then,
Next, we tested whether ML protects A549 cells from PQ- intracellular ROS levels were measured by the 2’,7’-dichlorofluorescein
induced oxidative stress through scavenging of intracellular diacetate (DCF-DA) assay. The data are presented as the mean ± SD
ROS or via other mechanisms. To quantify the intracellular of three independent experiments (***p < 0.001).

ROS production, the DCF-DA assay was performed


(Fig. 2). Here, 100 µM H2O2 was used as a positive control
(ROS inducer). The results showed that both 0.2 mM PQ the NRF2 mRNA (2.5-fold, p < 0.05) and protein levels (p <
and 100 µM H2O2 greatly increased intracellular ROS 0.01). Next, we analyzed the distribution of NRF2 in the
production by 69.54% and 41.1%, respectively. Incubation cytoplasm and nucleus. As shown in Fig. 3D, the protein
of untreated cells with 0.1 mg/ml ML alone produced no level of NRF2 was significantly increased in the nuclear
significant change in intracellular ROS, as compared with fraction in A549 cells exposed to 0.1 mg/ml ML alone (p <
the control. In contrast, cotreatment with ML and PQ 0.05) or in combination with PQ (p < 0.01). These data
significantly reduced the production of intracellular ROS showed that ML can enhance NRF2 expression and
by 11% in comparison with PQ treatment (p < 0.05). Thus, promote its nuclear translocation.
these data suggested that ML has an intracellular ROS- To dissect the regulation of antioxidant-related gene
scavenging effect, and this action may contribute to expression that is governed by activation of NRF2 during
cytoprotection under oxidative stress. PQ-induced oxidative stress, we performed a reporter
assay that is based on an ARE-containing reporter, where
ML Induces the Expression of NRF2 and Enhances ARE- the ARE reporter encodes a GFP gene under the control of
GFP Reporter Activity a minimal (m)CMV promoter and tandem repeats of the
The serious toxicity of PQ toward A549 cells and the ARE transcriptional response element (Figs. 4A and 4B).
partial restoration of cell viability by ML treatment raised Transfection of the inducible ARE-GFP and constitutively
the question whether NRF2 is involved in this effect. It is active ARE-GFP vectors into the cells caused activation of
well known that activation of NRF2 can prevent cell death the ARE reporter activity, in contrast to the non-inducible
caused by various environmental toxins that can induce ARE-GFP control vector. On the other hand, ML alone and
oxidative stress [33]. To determine whether ML can induce the combination of ML with PQ strongly enhanced the
NRF2 signaling, expression of NRF2 and its nuclear ARE-GFP reporter activity, in comparison with cells
translocation were examined in A549 cells that were treated with PQ only. Therefore, our results suggested that
exposed to ML, PQ, or the combination of both. As shown ML can effectively enhance the binding of NRF2 to the ARE
in Figs. 3A–3C, treatment of A549 cells with 0.1 mg/ml ML reporter.
significantly increased both mRNA (3-fold, p < 0.05) and
protein levels of NRF2 (p < 0.01). The cotreatment with ML Activates Antioxidant-Related Gene Expression
0.1 mg/ml ML and 0.2 mM PQ also effectively increased The binding of NRF2 to the ARE in the promoter region

February 2016 ⎪ Vol. 26 ⎪ No. 2


282 Jeon et al.

Fig. 3. Makgeolli lees (ML) enhances the expression of NRF2 and promotes its nuclear translocation.
(A) The mRNA level of NRF2 in cells incubated with paraquat (PQ), ML, or ML with PQ was measured by quantitative real-time reverse-
transcriptase PCR. The data are presented as the mean ± SD of at least three independent experiments (*p < 0.05). (B) The protein level of NRF2 in
the cells incubated for 24 h with the indicated doses of ML. The protein levels were measured by western blotting, and band intensity was
quantified in the ImageJ software. *Statistically significant differences between the control and treatment groups (*p < 0.05). (C) Protein levels of
NRF2 in the cells treated with PQ, ML, or ML plus PQ for 24 h (**p < 0.01). (D) Protein levels of NRF2 in nuclear and cytosolic fractions of the cells
treated with PQ, ML, or ML plus PQ for 24 h. The protein levels were quantified by western blotting, and the data were normalized to β-actin and
lamin B for the cytosolic and nuclear proteins, respectively (*p < 0.05, **p < 0.01).

Fig. 4. Makgeolli lees (ML) effectively activates an antioxidant response element (ARE)-green fluorescent protein (GFP) reporter.
Cells were seeded to attain 20–30% confluence on the following day. Next, the cells were transfected with an inducible ARE-responsive GFP
reporter, a construct constitutively expressing GFP, and a GFP reporter construct in which GFP expression is controlled by a minimal promoter.
Then, 6 h after the transfection, the cells were incubated with ML, paraquat (PQ), or ML plus PQ. After 24 h of the incubation, fluorescent images
were acquired to analyze the GFP activity. (A) Representative fluorescent images showing the effects of PQ, ML, or PQ plus ML on the binding of
NRF2 to the ARE-responsive GFP reporter. (B) Quantification of fluorescence intensity. The data are presented as the mean ± SD of at least three
independent experiments (**p < 0.01).

J. Microbiol. Biotechnol.
ML Protects PQ-Exposed A549 Cells 283

Fig. 5. Makgeolli lees (ML) enhances the expression of NRF2’s target genes and other cytoprotective genes.
(A) The protein level of NQO1 and HO-1 in the cells treated with 0.1 mg/ml ML for indicated periods of time. *Statistically significant differences
between the control and treatment groups (*p < 0.05). (B) The mRNA level of NQO1 and HO-1 in the cells incubated with paraquat (PQ), ML, or
PQ plus ML for 24 h. (C) The protein levels of NQO1 and HO-1 in the cells treated with PQ, ML, or PQ plus ML for 24 h. (D) The mRNA level of
GPXs. (E) The mRNA levels of CAT and SOD1. (F) The mRNA level of PRDXs. (G) The protein levels of PRDX3, PRDX4, and SOD1 in the cells
treated with PQ, ML, or PQ plus ML for 24 h. The mRNA and protein levels were analyzed by quantitative real-time reverse-transcriptase PCR
and western blotting, respectively. The data are presented as the mean ± SD of at least three independent experiments. In panels B–G, the asterisks
denote statistically significant differences among the cells treated with PQ only and cells treated with ML with or without PQ (*p < 0.05).

of antioxidant genes gives rise to the expression of several associated with cytoprotection. ML treatment in combination
antioxidant genes and phase II detoxification enzymes [26, 31]. with PQ significantly increased the mRNA expression of
Accordingly, we evaluated the effect of ML on the GPX3 (Fig. 5D), CAT, SOD1 (Fig. 5E), PRDX4, and PRDX5
expression of genes that counteract oxidative damage. As (Fig. 5F) as compared with the cells treated with PQ only.
shown in Fig. 5A, we observed an increase in the protein Moreover, the protein levels of PRDX3, PRDX4, and SOD1
levels of NQO1 and HO-1 during treatment with 0.1 mg/ml were also increased by ML alone and in cotreatment with
ML in a time-dependent manner. Similarly, our results PQ, as compared with the cells treated with PQ only
showed that cotreatment with 0.1 mg/ml ML and 0.2 mM (Figs. 5G and 5H).
PQ significantly increased both mRNA and protein levels
of NQO1 and HO-1 in A549 cells as compared with the Discussion
cells treated with PQ only (Figs. 5B and 5C). We also
analyzed the expression of some other antioxidant genes In this study, we examined the cytoprotective effect of

February 2016 ⎪ Vol. 26 ⎪ No. 2


284 Jeon et al.

ML on PQ-induced oxidative stress in human lung alveolar [31]. These enzymes include NQO1, HO-1, SOD isoforms,
A549 cells. One of the most salient findings of this study is CAT, PRDX isoforms, GPX, GST isoforms, and glutathione
that ML activates the NRF2–ARE regulatory axis, which is reductase [17, 35]. Several studies have shown that
a master regulator of the mechanisms of defense against moderate upregulation of HO-1 (less than 5-fold) is
oxidative stress. associated with protection from oxidative stressors [1, 10].
PQ-induced cellular toxicity is solely mediated by It has been revealed that a 3-fold overexpression of HO-1 is
oxidative stress, judging by the production of ROS during involved in protection from heme-mediated damage [1],
the redox cycle. The pulmonary toxicity pattern of PQ is in and 1.8-fold overexpression of HO-1 protein contributes to
many ways similar to that of several other lung toxins such the protection from oxygen toxicity [10].
as oxygen, nitrofurantoin, and bleomycin, which mainly We also observed a significant increase in the mRNA and
cause severe tissue damage followed by fibrosis after protein levels of HO-1 and NQO1 as a result of ML
systemic or respiratory administration [3]. Therefore, PQ treatment during PQ-induced oxidative stress. Therefore,
can be used as a model compound for studies on pulmonary we can assume that moderate upregulation of HO-1 is
toxicity and fibrosis exclusively mediated by oxidative involved in the ML-mediated cytoprotection against PQ-
stress in lung tissue. In the present study, PQ exposure was induced oxidative stress. The promoter region of the NQO1
shown to cause oxidative stress through accumulation of gene contains AREs, which are essential for both constitutive
intracellular ROS, which caused a dramatic reduction in and xenobiotic-induced NQO1 activity [27]. In contrast,
the viability of lung alveolar A549 cells. Our results are also basal NQO1 expression is nearly abrogated in cells and
supported by several reports showing the involvement of tissues of NRF2 knockout mice [23]. Thus, the upregulation
ROS in PQ-induced damage to lung cells [13, 18]. of NQO1 as a result of ML treatment during PQ-induced
Intracellular ROS levels are tightly regulated by oxidative stress is likely associated with regulation of the
inducible antioxidant pathways that respond to various NRF2–ARE axis.
cellular stressors and are mainly regulated by the nuclear Emerging evidence suggests that in addition to HO-1 and
transcription factor NRF2 [16, 26]. NRF2 is a crucial player NQO1, some other antioxidant genes such as GPXs, SOD1,
in cellular homeostasis; this protein is sequestered in the CAT, PRDX3, and PRDX4 are activated by the NRF2–ARE
cytoplasm in the inactive form and is translocated into the regulatory system and perform crucial functions in cellular
nucleus in the active form to regulate an antioxidant defense against oxidative stress [18, 24]. GPXs are a key
response upon the exposure of cells to chemical or group of peroxide-scavenging enzymes, and inactivation of
oxidative stress [29]. It has been reported that once NRF2 is their activity leads to excess accumulation of cytotoxic
activated, it relocates to the nucleus from the cytoplasm, binds peroxide and causes cellular damage [25]. Consequently,
to the ARE sites in target promoter regions, and regulates maintenance of the GPX activity is crucial for the protection
the expression of its downstream genes: antioxidant and of cells from oxidative stress.
detoxification-related genes. The transcriptional regulatory PRDXs are enzymes that exert their antioxidant action by
antioxidant response element ARE has structural and biological catalyzing reduction of hydrogen peroxide [24]. It is well
features that characterize its unique responsiveness to known that SOD plays a critical role in the reduction of
oxidative stress. Alteration of the cellular redox status after excess ROS in living organisms by converting the superoxide
intracellular accumulation of ROS triggers the ARE- radical to hydrogen peroxide, which is subsequently
dependent transcriptional response, which is mainly converted to water by the action of CAT and GPX [18].
governed by NRF2 [26]. CAT, another regulator of antioxidant defense mechanisms,
In this study, we found that ML treatment induces NRF2 exerts its action by degrading hydrogen peroxide and
expression and promotes its nuclear translocation. prevents formation of the hydroxyl radical by the Fenton
Furthermore, our results show that ML treatment during reaction. One report showed that overexpression of CAT
oxidative stress markedly increases the binding of NRF2 to makes cells more resistant to the toxic effects of hydrogen
an ARE-GFP reporter, suggesting that the cytoprotective peroxide and other types of oxidant-mediated injury [32].
mechanism of ML may involve the NRF2–ARE regulatory Conversely, mice deficient in CAT are more susceptible to
axis. hyperoxia-induced lung injury [14]. Therefore, in our
To control and neutralize pro-oxidative stress caused by study, the induction of these genes or enzymes either at the
xenobiotics, higher animals possess well-orchestrated gene transcriptional or translational level by ML alone or during
regulatory systems, including phase II detoxification enzymes cotreatment with PQ must be strongly associated with the

J. Microbiol. Biotechnol.
ML Protects PQ-Exposed A549 Cells 285

protection of A549 cells from PQ-induced oxidative stress. transgenic mice fed an amino acid-based diet supplemented
Collectively, our results demonstrated that ML enhances with red wine solids. Am. J. Clin. Nutr. 64: 748-756.
expression of NRF2, promotes its nuclear translocation, 9. Day BJ, Patel M, Calavetta L, Chang LY, Stamler JS. 1999. A
and facilitates its binding to an ARE in the promoter region mechanism of paraquat toxicity involving nitric oxide
synthase. Proc. Natl. Acad. Sci. USA 96: 12760-12765.
of the target genes, HO-1 and NQO. Additionally, ML can
10. Dennery PA, Wong HE, Sridhar KJ, Rodgers PA, Sim JE,
effectively upregulate several cytoprotective genes such as
Spitz DR. 1996. Differences in basal and hyperoxia-associated
SOD1, PRDX3, and PRDX4, which contribute to protection
HO expression in oxidant-resistant hamster fibroblasts. Am.
of the lung cell line A549 from PQ-induced toxicity by J. Physiol. 271: L672-L679.
decreasing the intracellular ROS level. Thus, the findings of 11. Ha J, Wang Y, Jang H, Seog H, Chen X. 2014. Determination
this study provide new efficacies of ML usage that might of E,E-farnesol in makgeolli (rice wine) using dynamic
lead to the development of a promising nutraceutical agent headspace sampling and stir bar sorptive extraction coupled
for the prevention or treatment of oxidative-stress-related with gas chromatography-mass spectrometry. Food Chem.
diseases. 142: 79-86.
12. Hayek T, Fuhrman B, Vaya J, Rosenblat M, Belinky P, Coleman
Acknowledgments R, et al. 1997. Reduced progression of atherosclerosis in
apolipoprotein E-deficient mice following consumption of
red wine, or its polyphenols quercetin or catechin, is
This study was supported by a research fund of
associated with reduced susceptibility of LDL to oxidation
Soonchunhyang University and by the Basic Science
and aggregation. Arterioscler. Thromb. Vasc. Biol. 17: 2744-2752.
Research Program through the National Research Foundation
13. He X, Wang L, Szklarz G, Bi Y, Ma Q. 2012. Resveratrol
of Korea (NRF) funded by the Ministry of Education inhibits paraquat-induced oxidative stress and fibrogenic
(2015R1A6A1A03032522). response by activating the nuclear factor erythroid 2-related
factor 2 pathway. J. Pharmacol. Exp. Ther. 342: 81-90.
References 14. Ho YS, Xiong Y, Ma W, Spector A, Ho DS. 2004. Mice
lacking catalase develop normally but show differential
1. Abraham NG, Lavrovsky Y, Schwartzman ML, Stoltz RA, sensitivity to oxidant tissue injury. J. Biol. Chem. 279: 32804-
Levere RD, Gerritsen ME, et al. 1995. Transfection of the 32812.
human heme oxygenase gene into rabbit coronary microvessel 15. Jeong JW, Park KJ. 2006. Quality characteristics of loaf bread
endothelial cells: protective effect against heme and hemoglobin added takju powder. Kor. J. Food Sci. Technol. 38: 52-58.
toxicity. Proc. Natl. Acad. Sci. USA 92: 6798-6802. 16. Jones CI, 3rd, Zhu H, Martin SF, Han Z, Li Y, Alevriadou
2. Bae SH, Jung EY, Kim SY, Shin KS, Suh HJ. 2010. BR. 2007. Regulation of antioxidants and phase 2 enzymes
Antioxidant and immune-modulating activities of Korean by shear-induced reactive oxygen species in endothelial
traditional rice wine, takju. J. Food Biochem. 34: 233-248. cells. Ann. Biomed. Eng. 35: 683-693.
3. Bus JS, Gibson JE. 1984. Paraquat: model for oxidant- 17. Joo CR, Cheng M, Shik KY. 2014. Desoxyrhapontigenin up-
initiated toxicity. Environ. Health Perspect. 55: 37-46. regulates Nrf2-mediated heme oxygenase-1 expression in
4. Castello PR, Drechsel DA, Patel M. 2007. Mitochondria are a macrophages and inflammatory lung injury. Redox Biol.
major source of paraquat-induced reactive oxygen species 2: 504-512.
production in the brain. J. Biol. Chem. 282: 14186-14193. 18. Kim H, Lee SW, Baek KM, Park JS, Min JH. 2011.
5. Choi YS, Park KS, Choi JH, Kim HW, Song DH, Kim JM, et al. Continuous hypoxia attenuates paraquat-induced cytotoxicity
2010. Physico-chemical properties of chicken meat emulsion in the human A549 lung carcinoma cell line. Exp. Mol. Med.
systems with dietary fiber extracted from makgeolli lees. J. 43: 494-500.
Kor. Soc. Food Sci. Anim. Resour. 30: 910-917. 19. Kim MS, Lee YS, Kim JS, Shin WC, Sohn HY. 2015.
6. Choi JS, Seo HJ, Lee YR, Kwon SJ, Moon SH, Park SM, Sohn Evaluation of in-vitro anti-thrombosis and anti-oxidation
JH. 2014. Characteristics and in vitro anti-diabetic properties activity of lees of takju (wookukseng). J. Life Sci. 24: 843-850.
of the Korean rice wine, makgeolli fermented with Laminaria 20. Kim SM, Cho WK. 2006. Effects of takju (Korean turbid rice
japonica. Prev. Nutr. Food Sci. 19: 98-107. wine) lees on the serum glucose levels in streptozotocin-
7. Choi YS, Park KS, Kim HY, Kim HW, Song DH, Chung HJ, induced diabetic rats. Kor. J. Food Cult. 21: 636-643.
et al. 2011. Interactions between chicken salt-soluble meat 21. Lee HS, Hong KH, Kim JY, Kim DH, Yoon CH, Kim SM.
proteins and makgeolli lees fiber in heat-induced gels. J. Kor. 2009. Blood pressure lowering effect of Korean turbid rice
Soc. Food Sci. Anim. Resour. 31: 817-826. wine (takju) lees extracts in spontaneously hypertensive rat
8. Clifford AJ, Ebeler SE, Ebeler JD, Bills ND, Hinrichs SH, (SHR). Kor. J. Food Cult. 24: 338-343.
Teissedre PL, Waterhouse AL. 1996. Delayed tumor onset in 22. Li N, Alam J, Venkatesan MI, Eiguren-Fernandez A,

February 2016 ⎪ Vol. 26 ⎪ No. 2


286 Jeon et al.

Schmitz D, Di Stefano E, et al. 2004. Nrf2 is a key 29. Paul MK, Bisht B, Darmawan DO, Chiou R, Ha VL, Wallace
transcription factor that regulates antioxidant defense in WD, et al. 2014. Dynamic changes in intracellular ROS levels
macrophages and epithelial cells: protecting against the regulate airway basal stem cell homeostasis through Nrf2-
proinflammatory and oxidizing effects of diesel exhaust dependent Notch signaling. Cell Stem Cell 15: 199-214.
chemicals. J. Immunol. 173: 3467-3481. 30. Pérez-Bibbins B, Torrado-Agrasar A, Salgado JM, de Souza
23. Lin X, Yang H, Zhou L, Guo Z. 2011. Nrf2-dependent Oliveira RP, Domínguez JM. 2015. Potential of lees from
induction of NQO1 in mouse aortic endothelial cells wine, beer and cider manufacturing as a source of economic
overexpressing catalase. Free Radic. Biol. Med. 51: 97-106. nutrients: an overview. Waste Manage. 90: 1027-1039
24. Miyamoto N, Izumi H, Miyamoto R, Kondo H, Tawara A, 31. Reuland DJ, Khademi S, Castle CJ, Irwin DC, McCord JM,
Sasaguri Y, Kohno K. 2011. Quercetin induces the expression Miller BF, Hamilton KL. 2013. Upregulation of phase II
of peroxiredoxins 3 and 5 via the Nrf2/NRF1 transcription enzymes through phytochemical activation of Nrf2 protects
pathway. Invest. Ophthalmol. Vis. Sci. 52: 1055-1063. cardiomyocytes against oxidant stress. Free Radic. Biol. Med.
25. Miyamoto Y, Koh YH, Park YS, Fujiwara N, Sakiyama H, 56: 102-111.
Misonou Y, et al. 2003. Oxidative stress caused by inactivation 32. Santanam N, Augé N, Zhou M, Keshava C, Parthasarathy S.
of glutathione peroxidase and adaptive responses. Biol. Chem. 1999. Overexpression of human catalase gene decreases
384: 567-574. oxidized lipid-induced cytotoxicity in vascular smooth
26. Nguyen T, Nioi P, Pickett CB. 2009. The Nrf2-antioxidant muscle cells. Arterioscler. Thromb. Vasc. Biol. 19: 1912-1917.
response element signaling pathway and its activation by 33. Surh YJ, Kundu JK, Na HK. 2008. Nrf2 as a master redox
oxidative stress. J. Biol. Chem. 284: 13291-13295. switch in turning on the cellular signaling involved in the
27. Nioi P, McMahon M, Itoh K, Yamamoto M, Hayes JD. 2003. induction of cytoprotective genes by some chemopreventive
Identification of a novel Nrf2-regulated antioxidant response phytochemicals. Planta Med. 74: 1526-1539.
element (ARE) in the mouse NAD(P)H:quinone oxidoreductase 34. Theodore M, Kawai Y, Yang J, Kleshchenko Y, Reddy SP,
1 gene: reassessment of the ARE consensus sequence. Villalta F, Arinze IJ. 2008. Multiple nuclear localization
Biochem. J. 374: 337-348. signals function in the nuclear import of the transcription
28. Pace-Asciak CR, Hahn S, Diamandis EP, Soleas G, Goldberg factor Nrf2. J. Biol. Chem. 283: 8984-8994.
DM. 1995. The red wine phenolics trans-resveratrol and 35. Thimmulappa RK, Mai KH, Srisuma S, Kensler TW,
quercetin block human platelet aggregation and eicosanoid Yamamoto M, Biswal S. 2002. Identification of Nrf2-regulated
synthesis: implications for protection against coronary heart genes induced by the chemopreventive agent sulforaphane
disease. Clin. Chim. Acta 235: 207-219. by oligonucleotide microarray. Cancer Res. 62: 5196-5203.

J. Microbiol. Biotechnol.

View publication stats

You might also like