Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

International Conference on Diet, Nutrition, and Cancer

Zingiberaceous and Citrus Constituents, 1ⴕ-Acetoxychavicol Acetate,


Zerumbone, Auraptene, and Nobiletin, Suppress Lipopolysaccharide-
Induced Cyclooxygenase-2 Expression in RAW264.7 Murine Macrophages
through Different Modes of Action1–3
Akira Murakami, Tomohiro Shigemori, and Hajime Ohigashi4

Downloaded from https://academic.oup.com/jn/article-abstract/135/12/2987S/4669952 by guest on 13 June 2020


Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University,
Kyoto 606-8502, Japan

ABSTRACT In the present study, we explored the suppressive activities of 1⬘-acetoxychavicol acetate (ACA), au-
raptene, nobiletin, and zerumbone toward LPS-induced cyclooxygenase (COX)-2 mRNA expression in mouse macro-
phages and the underlying molecular mechanisms. Pretreatment of RAW264.7 cells with LPS led to the activation of
mitogen-activated protein kinase (MAPK)s [p38, extracellular signal-regulated kinase (ERK)1/2, c-Jun NH2-terminal
kinase (JNK)1/2] and Akt, together with degradation of the inhibitor of nuclear factor-␬B (I␬B)-␣ protein and nuclear
translocation of nuclear factor (NF)-␬B p65, and the resultant activation of activator protein (AP)-1, NF-␬B, and
cAMP-responsive element-binding protein (CREB) transcription factors. ACA abrogated ERK1/2 and JNK1/2, but not
p38 MAPK, as well as the activation of those transcription factors. Although it allowed LPS-triggered phosphorylation
of those MAPKs and NF-␬B nuclear translocation, nobiletin suppressed the activation of AP-1, NF-␬B, and CREB.
Zerumbone had no effect on those transcription factors, though it attenuated COX-2 mRNA expression, suggesting that
it disrupts the stabilization of COX-2 mRNA. Conversely, zerumbone significantly accelerated spontaneous COX-2
mRNA decay, the potency of which was comparable with that of SB203580, an inhibitor of p38 MAPK, whose activation
has key roles in the proinflammatory mRNA stabilization processes. Because SB203580 but not zerumbone suppressed
LPS-induced p38 MAPK activation, the molecular targets of zerumbone may be MAPK-activated protein kinase-2 or
located downstream. However, auraptene suppressed the expression of COX-2 protein but not mRNA, implying that it
targets translation. We propose that these phytochemicals are promising chemopreventive agents for inflammation-
associated carcinogenesis. Their use in combination may enhance their efficacy because of their different modes of
action. J. Nutr. 135: 2987S–2992S, 2005.

KEY WORDS: ● cyclooxygenase-2 ● cancer prevention ● anti-inflammation ● macrophage


● dietary factor

Inflammation is a pathophysiological phenomenon that is cocktail of biochemical mediators, including reactive oxygen
involved in numerous diseases, including the development of and nitrogen species and proinflammatory chemokines and
neoplasms. Stromal activation of inflammatory cells induces cytokines, as well as eicosanoids such as prostaglandins (PGs).5
dormant tumor cells to grow and progress into malignant Cyclooxygenase (COX; PGH2 synthase) donates 2 oxygen
tumors. Upon stimulation, those cells produce and secrete a molecules to arachidonic acid to form PGG2 by peroxidation,
which in turn is reduced to PGH2. Nonsteroidal anti-inflam-
matory drugs such as aspirin have received considerable atten-
1
Published in a supplement to The Journal of Nutrition. Presented as part of tion for their ability not only to mitigate inflammatory re-
the International Research Conference on Food, Nutrition, and Cancer held in sponses but also potentially to prevent cancer incidence in the
Washington, DC, July 14 –15, 2005. This conference was organized by the Amer-
ican Institute for Cancer Research and the World Cancer Research Fund Inter-
human colon (1). Conversely, numerous animal experiments
national and sponsored by (in alphabetical order) California Avocado Commis- have demonstrated the cancer preventive efficacy of COX
sion; California Walnut Commission; Campbell Soup Company; The Cranberry
Institute; Danisco USA, Inc.; The Hormel Institute; National Fisheries Institute; The
Solae Company; and United Soybean Board. Guest editors for this symposium
were Vay Liang W. Go, Ritva R. Butrum, and Helen A. Norman. Guest Editor 5
Abbreviations used: ACA, 1⬘-acetoxychavicol acetate; AP-1, activator pro-
Disclosure: R. R. Butrum and H. Norman are employed by conference sponsor tein-1; COX, cyclooxygenase; CREB, cAMP-responsive element-binding protein;
American Institute for Cancer Research; V.L.W. Go, no relationships to disclose. DMSO, dimethylsulfoxide; ERK, extracellular signal-regulated kinase; FBS, fetal
2
Author Disclosure: No relationships to disclose. bovine serum; GPx, glutathione peroxidase; GST, glutathione S-transferase;
3
Supported by grants-in-aid from the Ministry of Agriculture, Forestry, and HPRT, hypoxanthine phosphoribosyl transferase; HRP, horseradish peroxidase;
Fisheries Food Research Project, Integrated Research on Safety and Physiolog- I␬B, inhibitor of NF-␬B; JNK, c-Jun NH2-terminal kinase; LPS, lipopolysaccharide;
ical Function of Food. MAPK, mitogen-activated protein kinase; MAPKAPK-2, MAPK-activated protein
4
To whom correspondence should be addressed. kinase-2; NF-␬B, nuclear factor ␬B; NQO1, NAD(P)H quinone oxidoreductase;
E-mail: ohigashi@kais.kyoto-u.ac.jp. PG, prostaglandin; Pi, proteinase inhibitor.

0022-3166/05 $8.00 © 2005 American Society for Nutrition.

2987S
2988S SUPPLEMENT

inhibitors. The COX enzyme consists of at least two isoforms, lated the relative expression levels of mRNA in the samples as the
COX-1 and COX-2. In contrast to COX-1, COX-2 protein is ratio of the amount of each gene mRNA to that of HPRT mRNA.
only slightly expressed in most normal mammalian tissues in The number of PCR cycles was optimized so that each band intensity
response to physical, chemical, and biological stimuli, includ- increased proportionally as the amount of cDNA increased. Each
ing UV light exposure, dioxin, and lipopolysaccharide insult. experiment was done at least 3 times.
Western blotting. RAW264.7 cells (2 ⫻ 106) were grown to
Recently, COX-2 has received the attention of numerous confluence in 4 mL DMEM with 10% FBS on a 60-mm dish, then
researchers regarding the relation of its expression to patho- incubated in an atmosphere containing 5% CO2 at 37°C for 13 h.
genesis. In particular, ample evidence exists of the involve- The cells were washed with PBS twice, after which the medium was
ment of COX-2 expression in carcinogenesis in many different exchanged with FBS- and phenol-red-free mediium (10 mL) contain-
target organs (2). ing samples dissolved in 20 ␮L DMSO. After 30 min of preincuba-
We screened Japanese (3) and subtropical vegetables and tion, the cells were treated with LPS (100 ␮g/L) for various times.
fruits (4 – 6) for their anti–tumor-promoting activities and After being washed with PBS twice, the cells were separated into
identified some active constituents [reviewed in Nakamura et nuclear and cytosol fractions using a kit (Bio Vision; Research Prod-
al. (7)]. It is notable that 1⬘-acetoxychavicol acetate (ACA, ucts). Protein concentrations were determined using a DC Protein

Downloaded from https://academic.oup.com/jn/article-abstract/135/12/2987S/4669952 by guest on 13 June 2020


from Alpinia galanga, Zingiberaceae) and zerumbone (from Assay kit (Bio-Rad), with ␥-globulin employed as the standard. Next,
Zingiber zerumbet, Zingiberaceae) as well as auraptene and 10 or 20 ␮g of proteins (for the nuclear and cytosol fractions,
respectively) were separated on 10% polyacrylamide gels and elec-
nobiletin (from citrus fruits), which are readily available from trophoretically transferred onto polyvinylidene difluoride membranes
natural sources and/or chemically synthesized, prevented (Millipore). After blocking, the membranes were incubated with
chemical carcinogenesis in several organs (8 –16), including rabbit anti-proteinase inhibitor (Pi)-extracellular signal-regulated ki-
the colon, in mouse and rat experiments. The mechanisms of nase (ERK)1/2, rabbit anti-ERK1/2 (Promega), rabbit anti-Pi-c-Jun
action of these agents are not fully understood, though impor- NH2-terminal kinase (JNK)1/2 antibody, rabbit anti-JNK1/2 anti-
tant findings based on the results of cellular and animal ex- body, rabbit anti-Pi-p38 mitogen-activated protein kinase (MAPK)
periments have accumulated. Those mechanisms include the antibody, rabbit anti-p38 antibody (Cell Signaling), rabbit anti-Pi-
induction of phase II enzymes and apoptosis, attenuation of Ser473 Akt1/2/3 antibodies, rabbit anti-Akt1/2/3 antibody, rabbit
reactive oxygen and nitrogen species from inflammatory cells, anti–inhibitor of nuclear factor ␬B (I␬B)-␣ antibody, rabbit anti-
regulation of proinflammatory cytokines, and inhibition of nuclear factor (NF)-␬B p65 antibody, rabbit anti-MAPK-activated
mutagenesis. In addition, these dietary factors have shown a protein kinase (MAPKAPK)-2 (Santa Cruz Biotechnology), and goat
anti-␤-actin antibody (Biochemical Technologies) (1:1000 dilution
marked ability to suppress COX-2 expression both in vitro and each) and then with the corresponding secondary antibodies [horse-
in vivo (8,10,17,18), though their molecular mechanisms re- radish peroxidase (HRP)-conjugated anti-rabbit IgG, 1:1000 dilution
main largely unknown. In the present study, we investigated (Dako); or HRP-conjugated anti-goat IgG, 1:1000 dilution (Dako)].
the mechanisms by which ACA, auraptene, nobiletin, and The blots were developed using an ECL detection kit (Amersham
zerumbone attenuate LPS-induced COX-2 mRNA expression Life Science). Each experiment was done at least 3 times.
in RAW264.7 mouse macrophages. Reporter assays. The following reporter assays were performed
using a Mercury® Pathway Profiling System (Clontech Laboratories)
with some modifications. RAW264.7 cells (3 ⫻ 105 cells/mL) were
Materials and methods preincubated on a 24-well plate for 12 h. Next, 625 ␮L OPTI-MEM®
Reagents and cells. ACA (19), auraptene (20), nobiletin (12), (Invitrogen) and 37.5 ␮L LipofectAMINE Reagent® (Invitrogen)
and zerumbone (5) were purified as previously reported. DMEM and were mixed in a tube, after which 4 ␮g of either pNF-␬B-, pAP-1, or
fetal bovine serum (FBS) were purchased from Invitrogen. LPS (Esch- pcAMP-responsive element-binding protein (CREB)-luciferase vec-
erichia coli serotype 0127, B8) was purchased from Difco Labs. All tor, provided in the kit, and 4 ␮g of pRL-TK vector (Promega), which
other chemicals were purchased from Wako Pure Chemical Industries served as the internal standard, were added and allowed to stand at
unless specified otherwise. RAW264.7 murine macrophages were room temperature for 30 min, after which an additional 5 mL OPTI-
purchased from the American Type Culture Collection. MEM was added. After the cells were washed with Hanks’ buffer
RT-PCR. RAW264.7 cells (1 ⫻ 106) were grown to confluence twice, 250 ␮L transfection mixture was added to each well, and the
in 5 mL DMEM with 10% FBS on 6-well plates, then incubated in an cells were incubated at 37°C for 6 h. After being washed, the cells
atmosphere containing 5% CO2 at 37°C for 13 h. The cells were were incubated in 1 mL DMEM with 10% FBS for 12 h. The cells
washed with PBS twice, after which the medium was replaced with were washed again with Hanks’ buffer twice, then exposed to the
FBS- and phenol-red-free medium (2.5 mL) containing samples dis- vehicle (0.5% DMSO, v:v) or samples dissolved in DMSO in serum-
solved in 25 ␮L dimethylsulfoxide (DMSO). After 30 min of prein- free DMEM for 30 min. After stimulation of the cells with LPS (100
cubation, the cells were treated with LPS (100 ␮g/L). After a 6-h ␮g/L) for 12 h, luciferase activity in the cell lysate was determined
incubation, the cells were lysed, and total RNA was extracted using using a Dual-Luciferase Reporter Assay Kit® (Promega).
kits (RNeasy® minikit and QIAshredder®, Qiagen). One microgram COX-2 mRNA decay. RAW264.7 cells (2 ⫻ 106) were grown to
of total RNA was reverse transcribed using an RNA PCR Kit® confluence in 3 mL DMEM with 10% FBS on 6-well plates, then
(Takara) with an oligo dT-adaptor primer, as recommended by the incubated in an atmosphere containing 5% CO2 at 37°C for 13 h.
supplier. Then, PCR assays were performed using a thermal cycler The cells were washed with PBS twice, after which the medium was
(PTC-0100; MJ Research) with hypoxanthine phosphoribosyl trans- exchanged with FBS- and phenol-red-free medium (3 mL) containing
ferase (HPRT) and COX-2 using primers synthesized by Proligo with LPS (100 ␮g/L) and incubated for 4.5 h. Then the cells were treated
1 ␮L of a cDNA preparation, 45 ␮L of Platinum® PCR SuperMix with actinomycin D (1 ␮g/mL final concentration) for 30 min,
(Invitrogen), and 2 ␮L of each primer (1 ␮mol/L) as follows: HPRT followed by treatment with the vehicle or sample for 6 h. RT-PCR
(5⬘-gTAATgATCAgTCAACggggAC-3⬘ and 5⬘-CCAgCAAgCTT- was performed as described above.
gcAACCTTAACCA-3⬘), 20 cycles at 94°C for 2 min, 58°C for 2 In vitro kinase assay. A cell-free kinase assay was performed
min, and 72°C for 2 min; and COX-2 (5⬘-gCATTCTTTgCCCAg- using a Kinase-Glo® Luminescent Kinase Assay (Promega). To a
CACTT-3⬘ and 5⬘-AgACCAggCACCAgACCAAAgA-3⬘), 20 cy- disposable culture tube (12 ⫻ 75 mm; Iwaki), 1.5 ␮L recombinant
cles at 94°C for 30 s, 59°C for 30 s, and 72°C for 30 s. The PCR MAPKAPK-2 (PanVera), 0.2 ␮L recombinant active p38␣ (Pan-
products were separated on 2% NuSieve® 3:1 agarose (BioWhittaker Vera), and 23.3 ␮L kinase buffer (25 mmol/L HEPES, pH 7.6; 2
Molecular Applications), and each band was visualized using 0.01% mmol/L dithiothreitol; 20 mmol/L MgCl2; 0.1 mmol/L NaVO4) were
SYBR Gold® stain (Molecular Probes). The amplified products were added. After 1 ␮L of each sample and 25 ␮L ATP solution (4
photographed with a digital camera, and the band intensities were ␮mol/L) were added, the reaction mixture was incubated at 28°C for
analyzed using NIH Image software. For each target gene, we calcu- 3 h. Thereafter, 50 ␮L Kinase-Glo Reagent (Promega) was added,
ZINGIBERACEOUS AND CITRUS CONSTITUENTS 2989S

induction of COX-2 mRNA as detected by RT-PCR. The


concentration of each dietary factor (Fig. 1A) was nonlethal
up to the highest concentration tested. Both zerumbone and
ACA (20 ␮mol/L each) abolished LPS-induced COX-2 ex-
pression, whereas nobiletin, but not auraptene, moderately
attenuated that expression (Fig. 1B).
Effects on activation of MAPKs and NF-␬B system. LPS
stimulation induced the time-dependent phosphorylation of
p38 MAPK from 0 to 60 min, whereas that of JNK1/2 and
ERK1/2 peaked at 30 min after the LPS challenge. Activation
of p38 was not attenuated by zerumbone, ACA, or nobiletin
(Fig. 2A). However, ACA substantially blocked both JNK1/2
and ERK1/2 activation, whereas both zerumbone and nobile-
tin allowed LPS-induced activation of those protein kinases.

Downloaded from https://academic.oup.com/jn/article-abstract/135/12/2987S/4669952 by guest on 13 June 2020


Furthermore, the phosphorylation of Akt (protein kinase B) at
Ser473 was detectable in nontreated cells and enhanced by LPS
in a time-dependent manner (Fig. 2B). None of the 3 com-
pounds demonstrated any inhibition. LPS-induced NF-␬B ac-
tivation is mediated through the degradation of I␬B, a sup-
pressive partner of NF-␬B, leading to the nuclear translocation
of NF-␬B for transactivation of numerous proinflammatory
FIGURE 1 (A) Chemical structure of dietary factors. (B) Suppres- and antiapoptotic genes. As shown in Figure 2B, I␬B protein
sive effects of dietary factors on LPS-induced COX-2 mRNA expres- was scarcely seen at 30 min after the LPS challenge and was
sion detected by RT-PCR, as described in Materials and Methods. restored within 60 min. Conversely, NF-␬B p65 was signifi-
ACA, 5 ␮mol/L; AUR, auraptene, 20 ␮mol/L; BL, negative control
cantly translocated into the nucleus from 30 to 60 min. ACA
treated with vehicle; NOB, nobiletin, 200 ␮mol/L; ZER, zerumbone, 20
␮mol/L.
did not allow those LPS-induced biochemical processes to
occur, whereas both zerumbone and nobiletin were virtually
inactive, except that the nuclear protein level of p65 in the
and the culture was incubated at room temperature for 15 min before nobiletin-treated cells was comparable with that in the non-
the determination of chemiluminescence. treated cells at 60 min.
Statistical analysis. Each experiment was done 3 times unless Effects on transcriptional activity of NF-␬B, AP-1, and
specified otherwise, with values shown as means ⫾ SD. The signifi-
cance of differences between groups in each assay was assessed using CREB. We transfected the NF-␬B-, AP-1-, and CREB-
a Student’s t-test (2-sided) that assumed unequal variance. luciferase vectors to examine the effects of dietary factors on
those transcriptional activities (Fig. 3). LPS treatment mark-
Results edly elevated the activities of the transcription factors by 2.1-
to 6.8-fold. It is notable that ACA and nobiletin significantly
Effects on COX-2 mRNA expression. Treatment of suppressed the 3 transcription factors, whereas zerumbone had
RAW264.7 macrophages with LPS for 6 h caused a marked no effect.

FIGURE 2 Effects of dietary fac-


tors on LPS-induced phosphorylation
of p38, JNK1/2, and ERK1/2 (A); and
Akt (Ser473) activation, I␬B protein deg-
radation, and NF-␬B p65 nuclear
translocation (B); detected by Western
blotting assay, as described in Materi-
als and Methods. ACA, 5 ␮mol/L;
NOB, nobiletin, 200 ␮mol/L; ZER,
zerumbone, 20 ␮mol/L.
2990S SUPPLEMENT

FIGURE 3 Effects of dietary fac-


tors on LPS-induced NF-␬B (A), AP-1
(B), and CREB (C) activation, detected
by luciferase reporter assays, as de-
scribed in Materials and Methods. *Dif-
ferent from BL, P ⬍ 0.01; **different
from BL, P ⬍ 0.05; ***different from
LPS, P ⬍ 0.01; ****different from LPS,
P ⬍ 0.05; Student’s t test. ACA, 5
␮mol/L; BL, negative control treated
with vehicle; NOB, nobiletin, 200
␮mol/L; RLU, relative luciferase unit;
ZER, zerumbone, 20 ␮mol/L.

Downloaded from https://academic.oup.com/jn/article-abstract/135/12/2987S/4669952 by guest on 13 June 2020


Zerumbone promotion of COX-2 mRNA degredation. A oral and topical administration of zerumbone suppressed phor-
number of proinflammatory genes have highly conserved AU- bol ester– and azoxymethane-induced COX-2 protein expres-
rich elements in the proximal regions of the 3⬘-untranslated sion in mouse skin (6) and rat colon (10), respectively, though
region, which play critical roles in their mRNA stabilization the molecular mechanisms were not fully elucidated. The
processes via activation of the p38 MAPK pathway (21). present results provide some preliminary but useful insights
RAW264.7 cells were pretreated with LPS to induce COX-2 into the molecular mechanisms of these dietary factors (Fig.
mRNA expression and then exposed to actinomycin D to 5). Auraptene may disrupt the translation step of COX-2
block gene transcription. The cells were then treated with the mRNA, because it was reported to suppress expression of the
vehicle, zerumbone, or SB203580, an inhibitor of p38 MAPK, protein in LPS-treated RAW264.7 cells (18), whereas it did
after which total RNA was extracted and RT-PCR was per- not suppress mRNA expression in our experiment (Fig. 1). In
formed. Zerumbone accelerated the vehicle-induced degrada- addition, we recently found that auraptene suppresses the
tion of COX-2 mRNA, and its potency was comparable with expression of protein but not mRNA of matrix metallopro-
that of SB203580 (Fig. 4A). Further, the p38-induced phos-
phorylation of MAPKAPK-2 was abolished by SB203580 and
slightly attenuated by zerumbone (Fig. 4B). A cell-free in vitro
kinase assay for p38 MAPK activity also revealed that zerum-
bone did not inhibit the p38 enzyme up to a concentration of
100 ␮mol/L, whereas SB203580 demonstrated complete inhi-
bition at 1 ␮mol/L (Fig. 4C).

Discussion
The dietary factors examined in the present study were
shown to be cancer preventive in mouse and rat models
(8 –16). Although the mechanisms of action have not been
fully elucidated, some of our previous findings in regard to
their biological and biochemical activities may be helpful. For
example, oral feeding of ACA and auraptene led to a marked
elevation of phase II drug metabolizing enzymes, including
glutathione S-transferase (GST) and NAD(P)H quinone ox-
idoreductase (NQO1), in the colon and liver of rats (14,22).
Similarly, zerumbone induced the activation of transcription
factor Nrf2 in RL34 rat hepatocytes for upregulating the an-
tioxidative and self-protecting enzymes ␥-glutamylcysteine
synthetase, glutathione peroxidase (GPx), and hemeoxygen-
ase-1 (23). These results are consistent with our other in vivo
observations that a topical application of zerumbone enhanced
or induced mRNA expression by the manganese superoxide
dismutase gene as well as the GPx, GST-P1, and NQO1 genes
without affecting the expression levels of cytochrome P450
1A1 and 1B1 mRNA in mouse epidermis (8). Furthermore, FIGURE 4 (A) Blots for RAW 264.7 cells treated with LPS and
the antioxidative and antinitrosative properties of that com- incubated for 4.5 h, then treated with actinomycin D (1 ␮g/mL) for 30
min, followed by treatment with the vehicle or sample for 6 h. RT-PCR
pound, which are related to the attenuation of endotoxin- or
was performed as described in Materials and Methods. (B) Blots for
phorbol ester–induced superoxide anion and nitric oxide gen- RAW 264.7 cells treated with either zerumbone (ZER; 20 ␮mol/L) or
eration in inflammatory cells, were described in other cell SB203580 (SB; 20 ␮mol/L) for 30 min, then exposed to LPS for 30 or 60
culture and animal model studies (7,12,17–20). min, followed by Western blotting assay, as described in Materials and
ACA, auraptene, nobiletin, and zerumbone were found to Methods. (C) Graphed results of cell-free kinase assay, performed as
suppress COX-2 protein production in macrophages, whereas described in Materials and Methods.
ZINGIBERACEOUS AND CITRUS CONSTITUENTS 2991S

FIGURE 5 Proposed molecular


mechanisms by which dietary factors
attenuate the expression of LPS-in-
duced COX-2 protein production in
RAW264.7 mouse macrophages. ACA
targets both JNK1/2 and ERK1/2, and
nobiletin may interfere with coactiva-
tors such as CBP/p300 that suppress
the transactivation of NF␬B, AP-1, and
CREB. Because zerumbone allowed
LPS-induced MAPKs/Akt activation
and transcriptional activation of those
transcription factors while it abrogated
COX-2 mRNA induction, it may target

Downloaded from https://academic.oup.com/jn/article-abstract/135/12/2987S/4669952 by guest on 13 June 2020


MK-2 or downstream molecules for
destabilizing mRNA. Auraptene may
disturb the translation process of
COX-2 mRNA, because it suppressed
COX-2 protein but not mRNA produc-
tion. AUR, auraptene; CBP, CREB-
binding protein; IKK, I␬B kinase; MK-2,
MAPK-activated protein kinase-2; NOB,
nobiletin; PKA, protein kinase A; SB,
SB203580; TLR, toll-like receptor; ZER,
zerumbone.

teinase-7 in HT-29 human colon cancer cells (K. Kawabata et (37). Those genes, including the COX-2 gene, contain
al., unpublished data, 2005). highly conserved AU-rich elements in the proximal regions
In the present study, ACA substantially suppressed LPS- of the 3⬘-untranslated region (23). Some binding proteins
induced activation of both ERK1/2 and JNK1/2, which is for AU-rich elements were identified and shown to highly
probably associated with its ability to abrogate COX-2 mRNA affect COX-2 mRNA stability in RAW264.7 macrophages
via repression of AP-1, NF-␬B, and CREB, as shown previ- (38). Of importance, the binding abilities of those proteins
ously for the involvement of ERK1/2 and JNK1/2 with those are regulated by the p38 MAPK pathway (39). We found
transcriptional factors. For example, ERK activation is in- that zerumbone (20 ␮mol/L) abrogated LPS-induced COX-2
volved in the phosphorylation of p65, thereby activating mRNA expression (Fig. 1B), whereas it did not show any
NF-␬B (24); a similar observation was made for RAW264.7 suppression of the transcriptional activation of NF-␬B,
cells (25). Furthermore, the importance of Ser536 as the phos- AP-1, and CREB. It is likely that CCAAT/enhancer bind-
phorylation site of p65 was proposed (26). Chen et al. (27) ing protein ␤ and ␦ also may not be suppressed, because
reported that ERK activation led to NF-␬B activation that was their production depends on NF-␬B transcriptional activity
related to COX-2 expression, and other lines of evidence exist (40). This raises the possibility that zerumbone suppresses
for crosstalk activity between the ERK and CREB pathways COX-2 mRNA expression via a posttranscriptional mech-
(28 –30); the effects of ACA on protein kinase A activation anism. In the present study, zerumbone accelerated sponta-
should be explored in the future. Ample evidence shows that neous degradation of COX-2 mRNA (Fig. 4A), was virtu-
activation of the JNK pathway leads to AP-1 transcriptional ally inactive for suppressing p38 MAPK activation (Fig. 2),
activation, because phosphorylated c-Jun, one of the direct
and slightly attenuated MK2 phosphorylation (Fig. 4B)
targets of JNK, is a component of the AP-1 complex (31),
whereas our in vitro kinase assay revealed that it was
suggesting that ACA suppresses AP-1 activation by blocking
JNK activation, as shown in the present study (Fig. 5). The definitely inactive regarding inhibition of p38 MAPK ac-
direct targets of ACA, which are probably located upstream of tivity (Fig. 4C). Based on these findings, we hypothesize
ERK/JNK, should be investigated in the future. that zerumbone inhibits MK2 activity or targets unidenti-
Transcription coactivators, including CREB-binding pro- fied molecules located downstream of MK2 (Fig. 5).
tein and its homologue p300, are known to play major roles in In conclusion, ACA targets both JNK1/2 and ERK1/2, and
various stimuli-induced or -repressed intracellular signaling nobiletin may interfere with coactivators, such as CREB-
pathways (32) and were shown to potentiate the transcrip- binding protein/p300, that suppress the transactivation of NF-
tional activities of AP-1 (33), NF-␬B (34), and CREB (35). In ␬B, AP-1, and CREB. Zerumbone allowed LPS-induced
addition, ASC-2 has emerged as a novel coactivator partici- MAPKs/Akt activation and transcriptional activation of those
pating in the transcriptional activation of NF-␬B and AP-1 transcription factors, whereas it abrogated COX-2 mRNA
(36). Thus, nobiletin may disrupt the binding of those coac- induction. Thus, zerumbone may target MK-2 or downstream
tivators with their corresponding partner transcription factors, molecules for destabilizing mRNA. In addition, auraptene may
because we found that it suppressed NF-␬B, AP-1, and CREB disturb the translation process of COX-2 mRNA. because it
activation, whereas it was virtually inactive in suppression of suppressed COX-2 protein but not mRNA production. These
the activation of MAPKs and the Akt-NF-␬B pathway (Figs. results indicate that the dietary factors studied here have
2, 3, and 5). distinct molecular mechanisms for COX-2 protein suppression
The mRNA expression of many, if not all, proinflamma- and that their use in combination may lead to synergistic
tory genes is regulated by posttranscriptional mechanisms results with higher efficacy and lower toxicity.
2992S SUPPLEMENT

LITERATURE CITED 20. Murakami A, Kuki W, Takahashi Y, Yonei H, Nakamura Y, Ohto Y, Ohi-
gashi H, Koshimizu K. Auraptene, a citrus coumarin, inhibits 12-O-tetradecanoyl-
1. Kune GA, Kune S, Watson LF. Colorectal cancer risk, chronic illnesses, phorbol-13-acetate-induced tumor promotion in ICR mouse skin, possibly
operations, and medications: case control results from the Melbourne Colorectal through suppression of superoxide generation in leukocytes. Jpn J Cancer Res.
Cancer Study. Cancer Res. 1998;48:4399 – 404. 1997;88:443–52.
2. Subbaramaiah K, Dannenberg AJ. Cyclooxygenase 2: a molecular target 21. Lasa M, Mahtani KR, Finch A, Brewer G, Saklatvala J, Clark AR. Regu-
for cancer prevention and treatment. Trends Pharmacol Sci. 2003;24:96 –102. lation of cyclooxygenase 2 mRNA stability by the mitogen-activated protein
3. Koshimizu K, Ohigashi H, Tokuda H, Kondo A, Yamaguchi K. Screening kinase p38 signaling cascade. Mol Cell Biol. 2000;20:4265–74.
of edible plants against possible anti-tumor promoting activity. Cancer Lett. 22. Tanaka T, Kawabata K, Kakumoto M, Makita H, Matsunaga K, Mori H,
1988;39:247–57. Satoh K, Hara A, Murakami A, et al. Chemoprevention of azoxymethane-induced
4. Murakami A, Morita H, Safitri R, Ramlan A, Koshimizu K, Ohigashi H. rat colon carcinogenesis by a xanthine oxidase inhibitor, 1⬘-acetoxychavicol
Screening for in vitro anti-tumor-promoting activities of edible plants from Indo- acetate. Jpn J Cancer Res. 1997;88:821–30.
nesia. Cancer Detect Prev. 1998;22:516 –25. 23. Nakamura Y, Yoshida C, Murakami A, Ohigashi H, Osawa T, Uchida K.
5. Murakami A, Takahashi M, Jiwajinda S, Koshimizu K, Ohigashi H. Iden- Zerumbone, a tropical ginger sesquiterpene, activates phase II drug metabolizing
tification of zerumbone in Zingiber zerumbet Smith as a potent inhibitor of enzymes. FEBS Lett. 2004;572:245–50.
12-O-tetradecanoylphorbol-13-acetate-induced Epstein-Barr virus activation. 24. Zhang L, Ma Y, Zhang J, Cheng J, Du J. A new cellular signaling
Biosci Biotechnol Biochem. 1999;63:1811–2. mechanism for angiotensin II activation of NF-␬B: an I␬B-independent RSK-
6. Murakami A, Jiwajinda S, Koshimizu K, Ohigashi H. Screening for in vitro mediated phosphorylation of p65. Arterioscler Thromb Vasc Biol. 2005;25:1148 –

Downloaded from https://academic.oup.com/jn/article-abstract/135/12/2987S/4669952 by guest on 13 June 2020


anti-tumor promoting activities of edible plants from Thailand. Cancer Lett. 1995; 53.
95:139 – 46. 25. Jung SW, Lee MH, Kim YC, Paik SG, Choi YH, Kim YS, Kang KI.
7. Nakamura Y, Murakami A, Ohigashi H. Search for naturally-occurring Modulation of the transactivation function of nuclear factor-␬B by lipopolysac-
antioxidative chemopreventors on the basis of the involvement of leukocyte- charide in RAW264.7 macrophages. Int J Oncol. 2004;25:1081–7.
derived reactive oxygen species in carcinogenesis. Asian Pac J Cancer Prev. 26. Hu J, Nakano H, Sakurai H, Colburn N.H. Insufficient p65 phosphorylation
2000;1:115–20. at S536 specifically contributes to the lack of NF-␬B activation and transformation
8. Murakami A, Tanaka T, Lee JY, Surh YJ, Kim HW, Kawabata K, Nakamura in resistant JB6 cells. Carcinogenesis. 2004;25:1991–2003.
Y, Jiwajinda S, Ohigashi H. Zerumbone, a sesquiterpene in subtropical ginger, 27. Chen BC, Yu CC, Lei HC, Chang MS, Hsu MJ, Huang CL, Chen MC, Sheu
suppresses skin tumor initiation and promotion stages in ICR mice. Int J Cancer. JR, Chen TF, et al. Bradykinin B2 receptor mediates NF-␬B activation and
2004;110:481–90. cyclooxygenase-2 expression via the Ras/Raf-1/ERK pathway in human airway
9. Suzuki R, Kohno H, Murakami A, Koshimizu K, Ohigashi H, Yano M, epithelial cells. J Immunol. 2004;173:5219 –28.
Tokuda H, Nishino H, Tanaka T. Citrus nobiletin inhibits azoxymethane-induced 28. Impey S, Obrietan K, Wong ST, Poser S, Yano S, Wayman G, Deloulme
large bowel carcinogenesis in rats. Biofactors 2004;22:111– 4. JC, Chan G, Storm DR. Cross talk between ERK and PKA is required for Ca2⫹
10. Tanaka T, Shimizu M, Kohno H, Yoshitani S, Tsukio Y, Murakami A, Safitri stimulation of CREB-dependent transcription and ERK nuclear translocation.
R, Takahashi D, Yamamoto K, et al. Chemoprevention of azoxymethane-induced Neuron. 1998;21:869 – 83.
rat aberrant crypt foci by dietary zerumbone isolated from Zingiber zerumbet. Life
29. Davis S, Vanhoutte P, Pages C, Caboche J, Laroche S. The MAPK/ERK
Sci. 2001;69:1935– 45.
cascade targets both Elk-1 and cAMP response element-binding protein to
11. Kohno H, Yoshitani S, Tsukio Y, Murakami A, Koshimizu K, Yano M,
control long-term potentiation-dependent gene expression in the dentate gyrus in
Tokuda H, Nishino H, Ohigashi H, et al. Dietary administration of citrus nobiletin
vivo. J. Neurosci. 2000;20:4563–72.
inhibits azoxymethane-induced colonic aberrant crypt foci in rats. Life Sci. 2001;
30. Chen JC, Huang KC, Wingerd B, Wu WT, Lin WW. HMG-CoA reductase
69:901–13.
inhibitors induce COX-2 gene expression in murine macrophages: role of MAPK
12. Murakami A, Nakamura Y, Torikai K, Tanaka T, Koshiba T, Koshimizu K,
cascades and promoter elements for CREB and C/EBP␤. Exp. Cell Res. 2004;
Kuwahara S, Takahashi Y, Ogawa K, et al. Inhibitory effect of citrus nobiletin on
301:305–19.
phorbol ester-induced skin inflammation oxidative stress and tumor promotion in
31. Eferl R, Wagner EF. AP-1: a double-edged sword in tumorigenesis. Nat.
mice. Cancer Res. 2000;60:5059 – 66.
13. Kobayashi Y, Nakae D, Akai H, Kishida H, Okajima E, Kitayama W, Denda Rev. Cancer 2003;3:859 – 68.
A, Tsujiuchi T, Murakami A, et al. Prevention by 1⬘-acetoxychavicol acetate of the 32. Chakravarti D, LaMorte VJ, Nelson MC, Nakajima T, Schulman IG, Ju-
induction but not growth of putative preneoplastic glutathione S-transferase guilon H, Montminy M, Evans RM. Role of CBP/P300 in nuclear receptor signal-
placental form-positive focal lesions in the livers of rats fed a choline-deficient ling. Nature 1996;383:99 –103.
L-amino acid-defined diet. Carcinogenesis. 1998;19:1809 –14. 33. Horvai AE, Xu L, Korzus E, Brard G, Kalafus D, Mullen TM, Rose DW,
14. Tanaka T, Kawabata K, Kakumoto M, Hara A, Murakami A, Kuki W, Rosenfeld MG, Glass CK. Nuclear integration of JAK/STAT and Ras/AP-1 signal-
Takahashi Y, Yonei H, Maeda M, et al. Citrus auraptene exerts dose-dependent ing by CBP and p300. Proc Natl. Acad. Sci. U S A. 1997;94:1074 –9.
chemopreventive activity in rat large bowel tumorigenesis: the inhibition corre- 34. Ravi R, Mookerjee B, van Hensbergen Y, Bedi GC, Giordano A, El-Deiry
lates with suppression of cell proliferation and lipid peroxidation and with induc- WS, Fuchs EJ, Bedi A. p53-Mediated repression of nuclear factor-␬B RelA via the
tion of phase II drug-metabolizing enzymes. Cancer Res. 1998;58:2550 – 6. transcriptional integrator p300. Cancer Res. 1998;58:4531–36.
15. Tanaka T, Kawabata K, Kakumoto M, Matsunaga K, Mori H, Murakami A, 35. Lundblad JR, Kwok RP, Laurance ME, Harter ML, Goodman RH. Adeno-
Kuki W, Takahashi Y, Yonei H, et al. Chemoprevention of 4-nitroquinoline 1-ox- viral E1A-associated protein p300 as a functional homologue of the transcrip-
ide-induced oral carcinogenesis by citrus auraptene in rats. Carcinogenesis. tional co-activator CBP. Nature 1995;374:85– 8.
1998;19:425–31. 36. Lee SK, Na SY, Jung SY, Choi JE, Jhun BH, Cheong J, Meltzer PS, Lee
16. Tanaka T, Kawabata K, Kakumoto M, Makita H, Hara A, Mori H, Satoh K, YC, Lee JW. Activating protein-1 nuclear factor-␬B and serum response factor as
Hara A, Murakami A, et al. Citrus auraptene inhibits chemically induced colonic novel target molecules of the cancer-amplified transcription co-activator ASC-2.
aberrant crypt foci in male F344 rats. Carcinogenesis. 1997;18:2155– 61. Mol. Endocrinol. 2000;14:915–25.
17. Murakami A, Takahashi D, Kinoshita T, Koshimizu K, Kim HW, Yoshihiro 37. Bakheet T, Frevel M, Williams BR, Greer W, Khabar KS. ARED: human
A, Nakamura Y, Jiwajinda S, Terao J, et al. Zerumbone, a Southeast Asian ginger AU-rich element-containing mRNA database reveals an unexpectedly diverse
sesquiterpene, markedly suppresses free radical generation, proinflammatory functional repertoire of encoded proteins. Nucleic Acids Res. 2001;29:246 –54.
protein production, and cancer cell proliferation accompanied by apoptosis: the 38. Cok SJ, Acton SJ, Sexton AE, Morrison AR. Identification of RNA-binding
alpha,beta-unsaturated carbonyl group is a prerequisite. Carcinogenesis. 2002; proteins in RAW 264.7 cells that recognize a lipopolysaccharide-responsive ele-
23:795– 802. ment in the 3-untranslated region of the murine cyclooxygenase-2 mRNA. J. Biol.
18. Murakami A, Nakamura Y, Tanaka T, Kawabata K, Takahashi D, Ko- Chem. 2004;279:8196 –205.
shimizu K, Ohigashi H. Suppression by citrus auraptene of phorbol ester-and 39. Winzen R, Kracht M, Ritter B, Wilhelm A, Chen CY, Shyu AB, Muller M,
endotoxin-induced inflammatory responses: role of attenuation of leukocyte ac- Gaestel M, Resch K, et al. The p38 MAP kinase pathway signals for cytokine-
tivation. Carcinogenesis. 2000;21:1843–50. induced mRNA stabilization via MAP kinase-activated protein kinase 2 and an
19. Murakami A, Ohura S, Nakamura Y, Koshimizu K, Ohigashi H. 1⬘-Ace- AU-rich region-targeted mechanism. EMBO J. 1999;18:4969 – 80.
toxychavicol acetate, a superoxide anion generation inhibitor, potently inhibits 40. Wadleigh DJ, Reddy ST, Kopp E, Ghosh S, Herschman HR. Transcrip-
tumor promotion by 12-O-tetradecanoylphorbol-13-acetate in ICR mouse skin. tional activation of the cyclooxygenase-2 gene in endotoxin-treated RAW 264.7
Oncology. 1996;53:386 –91. macrophages. J. Biol. Chem. 2000;275:6259 – 66.

You might also like