You are on page 1of 6

1892 Biol. Pharm. Bull. 32(11) 1892—1897 (2009) Vol. 32, No.

11

Estrogenic Activity of Xanthorrhizol Isolated from Curcuma xanthorrhiza


ROXB.
ANGGAKUSUMA,a YANTI,a,b Myoungsu LEE,a and Jae-Kwan HWANG*,a
a
Department of Biotechnology, Yonsei University; 134 Shinchon-dong, Seodaemun-gu, Seoul 120–749, South Korea: and
b
Faculty of Biotechnology, Atma Jaya Catholic University; Jl Jenderal Sudirman 51, Jakarta 12930, Indonesia.
Received May 13, 2009; accepted August 7, 2009; published online August 14, 2009

Plant-derived estrogen-like compounds, or phytoestrogens, are given much attention due to their potential
therapeutic use. In this study, xanthorrhizol, a natural sesquiterpenoid isolated from the rhizome of Curcuma
xanthorrhiza ROXB. (Zingiberaceae), was evaluated for its estrogenic activity. It has been known that compounds
acting as ligands for estrogen receptors (ERs) are considered to possess estrogenic activity. Therefore, the Gal-
4/ER transactivation assay in transiently transfected African green monkey kidney (COS-7) cells was used to ex-
amine the estrogenic activity of xanthorrhizol. Both subtypes of ERs, ERa and ERb , were involved in this assay.
Further transactivation assays and pS2 mRNA analysis were also conducted in estrogen receptor-positive human
breast cancer (MCF-7). Our results showed that xanthorrhizol significantly increased Gal-4/ER luciferase activ-
ity in a dose-dependent manner and induced the endogenous ER-estrogen response element (ERE) interaction in
MCF-7 cells. Xanthorrhizol also significantly enhanced the expression of the pS2 gene in MCF-7 cells. In con-
trast, treatment using ICI 182780, an ER antagonist, suppressed all activities induced by xanthorrhizol, indicat-
ing ER-dependant activities were involved. These results suggest that xanthorrhizol possesses estrogenic activity
and its estrogenic effects are mediated by estrogen-induced gene expression.
Key words xanthorrhizol; estrogenic activity; estrogen receptor; transactivation assay; COS-7 cell; MCF-7 cell

Plant-derived estrogenic compounds, known as phytoestro- ment24) while in MCF-7 breast cancer cells apoptosis was
gens, have drawn attention due to their potential therapeutic modulated through bcl-2, p53, and poly(ADP-ribose)poly-
use. From epidemiologic data, Asian people, who frequently merase-1 (PARP-1).25) In another study using HepG2 he-
consume phytoestrogen-rich diets such as soybean, have been patoma cells, xanthorrhizol induced the cells to undergo
found to have a lower rate of osteoporotic fractures, cardio- apoptosis via the mitochondrial pathway.26) In addition, it has
vascular diseases, postmenopausal symptoms (PMS), and been reported that the combination of xanthorrhizol and cur-
certain cancers than Western populations.1) These health ad- cumin promoted apoptosis in MDA-MB-231 breast cancer
vantages are notably decreased when Asians adopt a Western cells.27)
lifestyle and eating habits. In other studies, phytoestrogens In the present study, using the Gal4/ERs ligand-binding
have been thought to show beneficial effects in cardiovascu- transactivation assay, we examined whether xanthorrhizol
lar and Alzheimer treatments.2—5) Research involving phy- might possess an estrogenic activity. It was hoped that this
toestrogens found largely in legumes, seeds, fruits, and veg- assay would provide further information regarding xanthor-
etables has been extensively conducted.6—9) Red clover, the rhizol and its benefit as a phytoestrogen. The assay was con-
first plant known for its estrogenic activities, contains high ducted in transiently-transfected ER-negative COS-7 cells
amounts of formononetin and biochanin A.10) Both com- using both subtypes of ERs, ERa and ERb , and ligand bind-
pounds were found to interact with estrogen receptors (ERs) ing domains (LBD). To determine if xanthorrhizol could in-
due to their structural similarity to 17b -estradiol.11) Cur- duce endogenous ER-estrogen response element (ERE) inter-
rently, there are four groups of phenolic compounds classi- action in cells, a further ligand binding assay was conducted
fied as phytoestrogens: isoflavones, stilbenes, coumestans, in ER-positive MCF-7 cells. In parallel, pS2 mRNA analysis
and lignans.11) Based on current findings revealing several was performed to investigate whether xanthorrhizol could
new phytoestrogen candidates, it is believed that other, yet promote ER target gene expression (pS2) in MCF-7 cells.
unknown compounds might possess estrogen-like activities The ER antagonist, ICI 182780, was used as a method of
with beneficial therapeutic potential.12—14) confirmation.
Sesquiterpenoid xanthorrhizol, a major bioactive com-
pound isolated from Curcuma xanthorrhiza ROXB. commonly MATERIALS AND METHODS
known as Javanese turmeric, has often been cited for its great
potential in food and medical applications. Xanthorrhizol has Plant Material Dried rhizomes of C. xanthorrhiza
been shown to exert antioxidant,15,16) anti-inflammatory,16,17) ROXB. were collected in Jakarta, Indonesia, and identified by
antibacterial,18) neuroprotective,19) nephroprotective,20) and Dr. Nam-In Baek, Department of Oriental Medicinal Materi-
hepatoprotective activity.21,22) Furthermore, xanthorrhizol has als and Processing, Kyunghee University (Yongin, Korea). A
been shown to have efficacy as a tumor chemopreventive voucher specimen (H015) has been deposited in the Depart-
agent. Xanthorrhizol was shown to suppress 12-O-tetrade- ment of Biotechnology, Yonsei University (Seoul, Korea).
canoylphorbol-13-acetate (TPA)-stimulated tumor promotion Xanthorrhizol isolated from C. xanthorrhiza was dissolved in
in mouse skin cells22) and attenuate lung metastasis in an in 100% (v/v) dimethyl sulfoxide (DMSO).
vivo model.23) In HeLa cervical cancer cells, p53 and bax-de- Isolation of Xanthorrhizol Xanthorrhizol (Fig. 1) was
pendent apoptosis were stimulated with xanthorrhizol treat- isolated from the ethyl acetate fraction of the methanol ex-
∗ To whom correspondence should be addressed. e-mail: jkhwang@yonsei.ac.kr © 2009 Pharmaceutical Society of Japan
November 2009 1893

and PlusTM Reagent (Invitrogen, Carlsbad, CA, U.S.A.). After


5 h transfection, the medium was changed to fresh medium
containing the indicated amount of experimental treatment.
In the MCF-7 experiment, the cells were seeded at concen-
tration 3105 cells/ml into 6-well plates in phenol red-free
Fig. 1. Structure of Xanthorrhizol RPMI-csFBS. An ERE-luciferase plasmid and pFR-b gal
plasmid were transfected into the cells using the previously
tract of C. xanthorrhiza ROXB., as described previously.18) mentioned method. Medium replacement was done after 12 h
Briefly, the dried rhizomes of C. xanthorrhiza (100 g) were transfection. After 18 h incubation, cells were lysed and the
ground and extracted with 75% MeOH (v/v; 400 ml). The cell lysates were used in activities measurements. The rela-
methanol extract was fractionated consecutively with ethyl tive luciferase unit (RLU) was measured using a Micro-
acetate, n-butanol, and water. The yield at each step was 4.8, LumatPlus LB 96V luminometer (Berthold, Wildbad, Ger-
1.7, and 1.1 g, respectively. The ethyl acetate fraction was many) and the b gal activity was measured using the 2-nitro-
separated chromatographically by applying the fraction to a phenyl b -D-galactopyranoside (ONPG) method (Sigma-
silica gel column (70—230 mesh, Merck & Co., Whitehouse Aldrich, St. Louis, MO, U.S.A.). Final results were obtained
Station, NJ, U.S.A.) and eluting with an n-hexane–ethyl ace- from the RLU normalized against b gal activity. In this exper-
tate solution (10 : 1, v/v). The final product, xanthorrhizol, iment, the Gal4-ERa plasmid was a kind gift from Dr. Yoon
was obtained as a single compound with a yield of 0.2 g. Kwang Lee (Kent State University, Kent, Ohio, U.S.A.) and
Cell Cultures African green monkey kidney cells, COS- the Gal4-ERb and ERE-luciferase plasmids were kindly pro-
7, and human breast cancer cells, MCF-7, were obtained vided by Dr. Hinrich Gronemeyer (Institute of Genetics and
from the American Type Culture Collection (Manassas, VA, Molecular and Cellular Biology, Illkirch Cedex, France).
U.S.A.). COS-7 cells were cultured in Dulbecco’s modified Both pFR reporter plasmids were purchased from Stratagene
Eagle’s medium (DMEM) and MCF-7 cells were cultured in (La Jolla, CA, U.S.A.).
RPMI-1640 medium (Gibco, Rockville, MD, U.S.A.). In rou- Semi-quantitative RT-PCR for pS2 Expression MCF-
tine culture, both cell types were supplemented with 10% 7 cells were grown in 6-well plates in phenol red-free RPMI-
fetal bovine serum (FBS) (Gibco, Rockville, MD, U.S.A.) csFBS. Cells were treated with the indicated amount of
and antibiotics (100 U/ml penicillin A and 100 m g/ml of reagent for 48 h. Total RNA from MCF-7 cells was extracted
streptomycin), and stored at 37 °C in a humidified incubator using Trizol reagent (Invitrogen Corp., Carlsbad, CA,
containing 5% CO2. When cells reached about 80% conflu- U.S.A.) according to the manufacturer’s protocol and quanti-
ence, the medium was changed to phenol red-free DMEM or fied spectrophotometrically at 260/280 nm. cDNA was syn-
RPMI supplemented with 1% charcoal dextran-stripped FBS thesized by using 1 m g total RNA and oligo dT reverse tran-
(csFBS) (Thermo Fisher Scientific, Logan, UT, U.S.A.) for scriptase premix (Elpis-Biotech, Taejeon, Korea) in a 20 m l
2 d of hormone starvation. After hormone starvation, cells total reaction volume. Reverse transcription was performed
were ready to use for assays. as follows: 70 °C for 5 min, 42 °C for 60 min, and 94 °C for
Cytotoxicity of Xanthorrhizol on COS-7 and MCF-7 5 min. The cDNA products were directly used in PCR reac-
Cells Cell viability was determined by the MTT colorimet- tions conducted using Takara LA Taq polymerase (Takara
ric assay (3-[4,5-diethylthiazol-2-yl]-2,5-dipheniltetrazolium Bio Inc., Shiga, Japan). The primers used for pS2 and glycer-
bromide; Sigma-Aldrich, St. Louis, MO, U.S.A.) according aldehyde 3-phosphate dehydrogenase (GAPDH) were 5-
to the method of Mosmann. After hormone starvation, the CACCATGGAGAACAAGGTGA-3 (pS2 forward), 5-CC-
cells were seeded at a concentration of 4104 cells/ml in 96- GAGCTCTGGGACTAATCA-3 (pS2 reverse), 5-CAAT-
well plates (Microtest 96, Becton Dickinson Co., Franklin GACCCCTTCATTGACC-3 (GAPDH forward), and 5-
Lakes, NJ, U.S.A.) and cultured in phenol red-free DMEM- TGAGTCCTTCCACGATACCA-3 (GAPDH reverse) to
csFBS at 37 °C in a 5% CO2 atmosphere. After 24 h of yield products of 323 and 425 bp, respectively. PCR con-
attachment, culture medium was discarded and the cells were sisted of 25 amplification cycles of 1 min at 94 °C, 1 min at
treated with various concentrations of xanthorrhizol or a annealing temperature, and 1 min at 72 °C in a thermal cycler
combination of xanthorrhizol and ICI 182780 (Sigma- (Gene Amp PCR System 2700, Applied Biosystems, CA,
Aldrich, St. Louis, MO, U.S.A.) in 200 m l serum-free and U.S.A.). The human GAPDH housekeeping gene was used as
phenol red-free DMEM or RPMI. The COS-7 cells were an internal control. PCR products were separated elec-
treated for 24 h and the MCF-7 cells were treated for 48 h. trophoretically in a 2% agarose DNA gel and stained with
After treatment, cells were incubated with 50 m l of MTT ethidium bromide. The stained gel was visualized by Gel-
(1 mg/ml) for 4 h at 37 °C. The insoluble formazan dye was Doc Quantity One software (Bio-Rad Laboratories, Hercules,
solubilized in 200 m l of DMSO and the spectrophotometric CA, U.S.A.).
absorbance was measured at 550 nm with a tunable mi- Statistical Analysis Triplicate experiments were per-
croplate reader (Versa Max, Sunnyvale, CA, U.S.A.). formed throughout this study. All data are presented as the
Transient Transfection and Luciferase Ligand Binding meanstandard deviation (S.D.). The significant difference
Transactivation Assays COS-7 cells were seeded at con- between the control (DMSO) and treated groups were statis-
centration 1106 cells/ml in 6-well plates in phenol red-free tically analyzed by the paired Student’s t-test (p0.05).
DMEM-csFBS. After seeding, a Gal4 DNA binding domain
(DBD)-ERa ligand binding domain (LBD) or Gal4 DBD- RESULTS
ERb LBD plasmid, pFR-luciferase, and pFR-b gal reporter
plasmid were transfected into the cells using Lipofectamine Cytotoxicity of Xanthorrhizol on COS-7 and MCF-7
1894 Vol. 32, No. 11

Cells The cytotoxicity of xanthorrhizol on COS-7 and


MCF-7 cells was determined by MTT assay. In the COS-7
assay, cells were treated with xanthorrhizol at concentrations
ranging from 1—50 m M for 24 h. At these concentrations,
xanthorrhizol did not affect the cell viability (Fig. 2A). Thus,
xanthorrhizol concentrations within this range were used in
further COS-7 transactivation assays. In contrast, in the
MCF-7 assay, cells demonstrated more sensitivity to xanthor-
rhizol than COS-7 cells. Xanthorrhizol at 1—7.5 m M resulted
in cell viability of more than 80% after incubation for 24 and
48 h (Fig. 2B). At 1 m M, xanthorrhizol slightly decreased cell
viability (13%) during a 48 h incubation. Therefore, we
used xanthorrhizol at concentrations of 0.5—5 m M for the as-
says involving MCF-7 cells.
Induction of Luciferase Activity by Xanthorrhizol in
Transiently Transfected COS-7 Cells In this study, ER-
negative COS-7 cells were transiently transfected with the
ERa and ERb reporter systems. As shown in Figs. 3A and B,
treatment of these cells with xanthorrhizol at various concen-
trations increased luciferase activity in a dose-dependent
manner. In ERa -transfected cells, 10 m M xanthorrhizol in-
duced luciferase activity by about 517% compared to control
treatment. Interestingly, luciferase activity was similar to that Fig. 2. Effects of Xanthorrhizol on COS-7 and MCF-7 Cell Viability
obtained with 5 m M estradiol (504%) (Fig. 3A). In ERb trans- COS-7 cells were treated with xanthorrhizol at concentrations of 1—50 m M for 24 h
fected cells, 10 m M xanthorrhizol induced luciferase activity (A) and MCF-7 cells were treated at concentrations of 1—25 m M for both 24 h and 48 h
(B). The cells treated with vehicle only (DMSO) were used as the control. Cell viability
by 832% compared with the vehicle (Fig. 3B). However, this was determined by the MTT assay. Data are shown as meanS.D. from triplicate ex-
luciferase activity was much lower than obtained with 5 m M periments. ∗∗ p0.01 compared with the control.
estradiol (5783%). Furthermore, to confirm that the agonistic
activity of xanthorrhizol was ER-mediated, ERa transfected

Fig. 3. Ligand Binding Activity of Xanthorrhizol in the Gal4/ERs Luciferase Transactivation Assays
COS-7 cells were transfected with the Gal4/ERa (A) or Gal4/ERb (B) fusion reporter system and treated with DMSO control, estradiol (5 m M), or xanthorrhizol (1, 5, 10 m M) for
18 h. (C) In the confirmation assay, transfected cells were co-treated with a combination of 5 m M xanthorrhizol and 1 m M ICI 182780. Final results were obtained from the RLU
which had been normalized against b gal activity. Cell viability for the co-treatment assay in COS-7 cells was also checked with 10 m M xanthorrhizol co-treated with various con-
centrations of ICI 182780 (1—25 m M). Cells treated with vehicle only (DMSO) were used as controls. Data are shown as meanS.D. from triplicate experiments. ∗ p0.05 com-
pared with the control.
November 2009 1895

cells were co-treated with 1 m M ICI 182780. These results pS2 mRNA expression was assayed. As shown in Fig. 4B,
showed that the luciferase activity induced by 5 m M xanthor- after 48 h sample treatment at 1 m M and 5 m M concentrations,
rhizol was completely diminished by ICI 182780 (Fig. 3C). the cells increased expression of pS2 mRNA compared to
The quenching of luciferase expression was not a result of control treatment. This result also confirmed the previous
reduced cell viability because co-treatment with both agents assay result. Finally, co-treatment of transiently transfected
did not cause COS-7 cell death (Fig. 3D). These results indi- MCF-7 cells with 1 m M ICI 182780 could completely abolish
cate that the induction of luciferase activity is possibly medi- both the luciferase activity and pS2 mRNA expression (Figs.
ated through ER LBD-ligand interaction. 4C, D). Nevertheless, both of the activities were not abol-
Effects of Xanthorrhizol on ERE Activation and pS2 ished due to the cell death. Since, as shown in Fig. 4E, the
mRNA Expression in MCF-7 Cells We investigated cell viability under treatment of 1 m M xanthorrhizol com-
whether xanthorrhizol activated endogenous ER in MCF-7 bined with 1 m M ICI 182780 was still remaining high
cells by transiently transfecting the cells with an ERE-lu- (80%) and similar with the 48 h treatment of 1 m M xanthor-
ciferase reporter gene. As seen in Fig. 4A, xanthorrhizol in- rhizol or ICI 182780 alone. This result implied that xanthor-
duced ERE-dependent luciferase activity by about 244% at rhizol exerted estrogenic effects through ER binding to ERE.
1 m M 24 h after treatment compared to controls. This result
meant that xanthorrhizol could activate the ER-ERE interac- DISCUSSION
tion followed by expression of the target gene. In a parallel
assay, to show if the original ER gene target in MCF-7 cells Phytoestrogens, a term coined to describe plant-derived
can also be induced by xanthorrhizol treatment, the level of chemicals that exert estrogenic activity, involve numerous va-

Fig. 4. Effects of Xanthorrhizol on ER Activation in MCF-7 Cells


(A) MCF-7 cells were transfected with the ERE-luciferase system and treated with vehicle, estradiol (1 m M), or xanthorrhizol (0.5, 1, 5 m M). (B) The confirmation assay where
transfected cells were co-treated with a combination of 1 m M xanthorrhizol and 1 m M ICI 182780. (C) The affect of xanthorrhizol on pS2 mRNA expression was assayed using semi-
quantitative RT-PCR. Cells were treated with vehicle, estradiol (1 m M) or xanthorrhizol (1, 5 m M). (D) The confirmation assay where transfected cells were co-treated with a combi-
nation of 1 m M xanthorrhizol and 1 m M ICI 182780. GAPDH mRNA was used as an internal control. (E) Cell viability for the co-treatment assay in MCF-7 cells was determined
using various concentration of xanthorrhizol (1, 5, 10 m M) co-treated with various concentrations of ICI 182780 (1—25 m M). Cells treated with vehicle were used as controls.
∗ p0.05 compared with the control.
1896 Vol. 32, No. 11

rieties of structurally diverse compounds and are grouped fects through the ER and follows the classical pathway of ER
into isoflavones, stilbenes, coumestans, and lignans. They activation.
can regulate the actions of endogenous estrogens, usually by To prove our hypothesis, xanthorrhizol was also tested in
binding to ERs.28,29) ER itself is a member of the nuclear re- MCF-7 cells, an estrogen positive cell line.28) Results showed
ceptor superfamily, which acts by regulating the transcription that, xanthorrhizol could enhance expression of both lu-
process. The classical pathway of ER action involves ligand ciferase and pS2 mRNAs compared to controls. However,
binding to receptor in the nucleus, followed by dimerization both signals appeared weaker than that obtained with estra-
and binding to specific response elements known as EREs, diol treatment (Figs. 4A, C). This was due to the fact that
located in the promoter regions of target genes.30) In humans, xanthorrhizol, at the higher concentration range used previ-
two distinct estrogen receptors, ERa and ERb , can be de- ously in the MCF-7 assay (1—10 m M) exhibited strong cyto-
tected in a broad spectrum of tissues. In some organs, both toxic activity.25) Finally, the up-regulated luciferase and pS2
receptor subtypes are expressed at similar levels, whereas in expressions could be reversed by ICI 182780 co-treatment.
others, one or the other subtypes predominates.31) Both ER The complete reversal of both signals showed that xanthor-
subtypes have functional differences, depending on the tissue rhizol exerted its estrogenic activity through the ER in MCF-
type. For example, ERb has been known for its potential as a 7 cells.
tumor suppression gene in breast tissue, while ERa plays an Our studies show that xanthorrhizol, isolated from C. xan-
important role in vessel wall pathophysiology in the cardio- thorrhiza ROXB. has estrogenic activity because it binds to
vascular system. ERa has also been known for playing an ERs and activates its target genes through the classical ER
important role in postpubertal bone elongation, while ERb pathway. This finding expands the potential uses for xanthor-
is responsible for repression of the ERa -mediated bone rhizol. From preliminary assays, we have determined that
growth-promoting effect. Furthermore, in adipocytes, ERa is xanthorrhizol does not interact with the glucocorticoid recep-
known to decrease adipose tissue mass by increasing lypoly- tor (data not shown). Therefore, we will explore if xanthor-
sis.31) Both ER subtypes bind phytoestrogen, although their rhizol may also activate other steroidal hormone receptors
binding patterns may vary and their affinities for phytoestro- (androgen receptor (AR) and mineral receptors (MR)). Fur-
gen are 100- to 10000-fold lower than that of estradiol.32) thermore, as a phytoestrogen, it will be determined if xan-
Based on recent reviews, phytoestrogens have frequently thorrhizol exhibits direct action on osteoblasts by inducing
been tested for their benefits. Phytoestrogens have been mineralization and if it has non-genomic estrogenic activity.
known for their ability to improve bone mass and reduce car-
diovascular risk factors in postmenopausal woman.33) Re- Acknowledgements This work was supported in part by
duced endometrial cancer risk has also been observed among the Yonsei Biomolecule Research Initiative of the two-step
those taking phytoestrogen. Phytoestrogens may also act as Brain Korea 21 Project.
antioxidants and inhibit blood vessel growth, essential for
tumor expansion (a similar function was also proposed for REFERENCES
xanthorrhizol). In Alzheimer’s disease research, phytoestro- 1) Adlercreutz H., Mazur W., Ann. Med., 29, 95—120 (1997).
gens showed beneficial effects on memory and central nerv- 2) Cruz M. N., Agewall S., Schenck-Gustafsson K., Kublickiene K.,
ous system function.34) Furthermore, in recent studies, phy- Atherosclerosis, 196, 49—58 (2008).
toestrogens have been assayed as cosmeceuticals as skin anti- 3) Cassidy A., Hooper L., J. Br. Menopause Soc., 12, 49—56 (2006).
aging and anti-carcinogenesis agents.35,36) 4) Tempfer C. B., Bentz E. K., Leodolter S., Tscherne G., Reuss F., Cross
H. S., Huber J. C., Fertil. Steril., 87, 1243—1249 (2007).
In this study, using the Gal4 fusion reporter system, xan- 5) Shi C., Xu J., Yew D. T. W., Neuroembryol. Aging, 4, 162—164
thorrhizol binding interaction to ERs was screened. From (2006).
our results, we found that xanthorrhizol could interact and 6) Lamikanra O., Grimm C. C., Rodin J. B., Inyang I. D., J. Agric. Food
saturate the ER ligand binding domain. The saturation was Chem., 44, 1111—1115 (1996).
7) Fremont L., Life Sci., 66, 663—675 (2000).
adequate to stimulate the luciferase signal in both ERa and
8) Frank A. A., Coster L. J., Cerna C. M., Navala K. K., J. Agric. Food
ERb systems. However, due to differences in the reporter Chem., 42, 1905—1913 (1994).
plasmids involved in this assay, the levels of luciferase signal 9) Setchell K. D., Lawson A. M., Borriello S. P., Harkness R., Gordon H.,
from both systems were different. The signals resulting from Morgan D. M., Lancet, 2, 4—7 (1981).
xanthorrhizol treatment, if compared with estradiol, were far 10) Bennettz H. V., Underwood E. J., Shier F. L., Aust. Vet. J., 22, 2—12
(1946).
lower in the ERb system than in the ERa system. In previous
11) Cornwell T., Cohick W., Raskin I., Phytochemistry, 65, 995—1016
studies, estradiol itself has been reported to have a lower (2004).
affinity to ERb than to ERa .37—39) Thus, it could be viewed 12) Hu Y., Hou T. T., Zhang Q. Y., Xin H. L., Zheng H. C., Rahman K.,
that xanthorrhizol might have a higher affinity for ERa be- Qin L. P., J. Pharm. Pharmacol., 59, 1307—1312 (2007).
cause it stimulated luciferase expression in a dose-dependent 13) Wang Z. Q., Lou Y. J., Eur. J. Pharmacol., 504, 147—153 (2004).
14) Usui T., Ikeda Y., Tagami T., Matsuda K., Moriyama K., Yamada K.,
manner and reached the same level of luciferase signal pro- Kuzuya H., Kohno S., Shimatsu A., J. Endocrinol., 175, 289—296
duced by 5 m M estradiol at a concentration of 10 m M (Fig. (2002).
3A). Although xanthorrhizol’s luciferase signal in the ERb 15) Park J. H., Kim J. H., Park K. K., Kim H. O., Hwang J. K., Chung W.
assay was also dose-dependent, and significant relative to Y., J. Korean Assoc. Cancer Prev., 8, 91—97 (2003).
controls, the signals were very low compared to that of estra- 16) Lim C. S., Jin D. Q., Mok H., Oh S. J., Lee J. U., Hwang J. K., Ha I.,
Han J. S., J. Neurosci. Res., 82, 831—838 (2005).
diol (Fig. 3B). Finally, in the co-treatment assay using the ER 17) Lee S. K., Hong C. H., Huh S. K., Kim S. S., Oh O. J., Min H. Y., Park
antagonist, ICI 182780, signal from the ERa system was K. K., Chung W. Y., Hwang J. K., J. Environ. Pathol. Toxicol. Oncol.,
completely reversed without affecting the cell viability (Figs. 21, 141—148 (2002).
3C, D). These results imply that xanthorrhizol exerts its ef- 18) Hwang J. K., Shim J. S., Pyun Y. R., Fitoterapia, 71, 321—323 (2000).
November 2009 1897

19) Kim S. H., Hong K. O., Hwang J. K., Park K. K., Food Chem. Toxicol., 30) Bjornstrom L., Sjoberg M., Mol. Endocrinol., 19, 833—842 (2005).
43, 117—122 (2005). 31) Dahlman-Wright K., Cavailles V., Fuqua S. A., Jordan V. C.,
20) Lin S. C., Lin C. C., Lin Y. H., Supriyatna S., Teng C. W., Am. J. Chin. Katzenellebogen J. A., Korach K. S., Maggi A., Muramatsu M., Parker
Med., 23, 243—254 (1995). M. G., Gustafsson J. A., Pharmacol. Rev., 58, 773—781 (2006).
21) Kim S. H., Hong K. O., Chung W. Y., Hwang J. K., Park K. K., Toxi- 32) Kuiper G. G., Lemmen J. G., Carlson B., Carton J. C., Safe S. H., van
col. Appl. Pharmacol., 196, 346—355 (2004). der Saag P. T., van der Burg B., Gustafsson J. A., Endocrinology, 139,
22) Chung W. Y., Park J. H., Kim M. J., Kim H. O., Hwang J. K., Lee S. 4252—4263 (1998).
K., Park K. K., Carcinogenesis, 28, 1224—1231 (2007). 33) Sirtori C. R., Arnoldi A., Johnson S. K., Ann. Med., 37, 423—438
23) Choi M. A., Kim S. H., Chung W. Y., Hwang J. K., Park K. K., (2005).
Biochem. Biophys. Res. Commun., 326, 210—217 (2005). 34) Usui T., Endocr. J., 53, 7—20 (2006).
24) Ismail N., Pihie A. H., Nallapan M., Anticancer Res., 25, 2221—2228 35) Wei H., Saladi R., Lu Y., Wan Y., Lebwohl M. G., J. Nutr., 133,
(2005). 3811S—3819S (2003).
25) Cheah Y. H., Azimahtol H. L., Abdullah N. R., Anticancer Res., 26, 36) Huang Z. R., Hung C. F., Lin Y. K., Fang J. Y., Int. J. Pharm., 364,
4527—4534 (2006). 36—44 (2008).
26) Handayaan T., Sakinah S., Nallapan M., Pihie A. H., Anticancer Res., 37) Burow M. E., Bove S. M., Collins-Burrows, Mclachlan J. A., J. Clin.
27, 965—971 (2007). Endocrinol., 86, 1750—1758 (2001).
27) Cheah Y. H., Nordin F. J., Tee T. T., Azimahtol H. L., Abdullah N. R., 38) Fokialakis N., Lambrinidis G., Mitsiou D. J., Alexis M. N., Chem.
Ismail Z., Anticancer Res., 28, 3677—3689 (2008). Biol., 11, 397—406 (2004).
28) Harris D. M., Besselink E., Henning S. M., Go V. L. W., Heber D., 39) Papoutsi Z., Kassi E., Halabalaki M., Mitakou S., Moutsatsou P., Toxi-
Exp. Biol. Med., 230, 558—568 (2005). col. in Vitro, 21, 364—370 (2006).
29) Setchell K. D., Am. J. Clin. Nutr., 68, 1333S—1346S (1998).

You might also like