Você está na página 1de 8

777

Genistein inhibits the development of


atherosclerosis via inhibiting NF-kB and VCAM-1
expression in LDLR knockout mice
Juejin Wang, Rongjian Zhang, Youhua Xu, Hong Zhou, Bin Wang, and Shengnan Li

Abstract: Diet can be an important factor that influences risks for cardiovascular disease. Genistein (4’,5,7-
trihydroxyisoflavone), rich in soy, is one candidate that may benefit the cardiovascular system. Here, we explored the ef-
fect of genistein in atherosclerosis (AS) development in an in vivo mouse model. Low-density lipoprotein receptor
(LDLR) knockout mice were allocated to control, model, and genistein groups. Our results showed that genistein signifi-
cantly reduced the formation and development of atherosclerotic plaques ((4.68 ± 1.18) 106 versus (6.65 ± 1.51) 106
mm2, p < 0.05). In the genistein group, compared with the model group, total antioxidant capacity (TAC) level was 85.5 ±
15.6 versus 203.4 ± 32.6 mmol/L (p < 0.01); malondialdehyde (MDA) level was 3.79 ± 0.28 versus 3.06 ± 0.31 mmol/L
(p < 0.01), and superoxide dismutase (SOD) activity was 86.1 ± 6.1 versus 139.1 ± 25.1 U/mL (p < 0.01). Therefore, gen-
istein was able to enhance serum antioxidative ability in our mouse model. Genistein had no influence, however, on serum
cholesterol and lipid profiles. Genistein also markedly downregulated the expression of nuclear factor (NF)-kB and vascu-
lar cell adhesion molecule (VCAM)-1 in aortas of mice (p < 0.05). These observations suggest that genistein may inhibit
AS in LDLR–/– mice via enhancing serum antioxidation and downregulating NF-kB and VCAM-1 expression in the aorta.
Key words: genistein, atherosclerosis, NF-kB, VCAM-1, LDLR knockout mice.
Résumé : La diète peut être un facteur de risque important de maladies cardiovasculaires. La génistéine (4’,5,7-trihydro-
xyisoflavone), riche en soya, est un candidat qui pourrait être d’une grande utilité pour le système cardiovasculaire. Dans
le présent article, nous avons exploré l’effet de la génistéine sur le développement de l’athérosclérose (AS) dans des modè-
les in vivo. Nous avons réparti des souris au gène LDLR invalidé (knockout) dans trois groupes : témoin, modèle et génis-
téine. Nos résultats ont montré que la génistéine a diminué de façon significative la formation et le développement des
plaques d’athérosclérose ((4,6 ± 1,18)  10–6 contre (6,65 ± 1,51)  106 mm2, p < 0,05). Chez le groupe génistéine,
l’indice TAC a été de 85,5 ± 16,6 contre 203,4 ± 32,6 mmol/L (p < 0,01), le taux de MDA de 3,79 ± 0,28 contre 3,06 ±
0,31 mmol/L (p < 0,01) et l’activité SOD de 86,1 ± 6,1 contre 139,1 ± 25,1 U/mL (p < 0,01) par rapport au groupe mo-
dèle, respectivement. Ainsi, la génistéine a pu stimuler la capacité antioxydative sérique dans les modèles de souris. Toute-
fois, elle n’a pu influer sur les profils des lipides et du cholestérol sériques. La génistéine a aussi diminué
considérablement l’expression de NF-kB et de VCAM-1 dans les aortes de souris (p < 0,05). Ces observations donnent à
penser que la génistéine pourrait inhiber l’AS chez les souris LDLR–/– en stimulant l’antioxydation sérique et en diminuant
l’expression de NF-kB et de VCAM-1 dans l’aorte.
Mots-clés : génistéine, athérosclérose, NF-kB, VCAM-1, souris knockout LDLR.
[Traduit par la Rédaction]

Introduction 1998; Wong et al. 1998), inhibited vascular smooth muscle


cell (VSMC) proliferation (Dubey et al. 1999), improved
Diet can be an important factor that influences the risks arterial compliance (Nestel et al. 1997), and had an anti-
for cardiovascular disease (Keys et al. 1986), and consump- oxidant action (Yamakoshi et al. 2000). Genistein (4’,5,7-
tion of a traditional Asian diet high in soy may play a key trihydroxyisoflavone), a dietary-derived isoflavonoid, has
role in preventing chronic diseases such as atherosclerosis various biological actions, including a weak estrogenic ef-
(AS) (Kolonel 1988). Among the active components of soy, fect by binding to estrogen receptors (ERs) (Kuiper et
isoflavones appear to have an important role in preventing al.1998) and inhibition of protein tyrosine kinases (PTK)
cardiovascular disease. Previous studies showed that soy iso- (Nakashima et al. 1991). In experimental studies, genistein
flavone decreased cholesterol concentration (Kirk et al. significantly prevented thrombotic vessel occlusion (Kondo
et al. 2002).
Received 3 April 2008. Accepted 25 August 2008. Published on
the NRC Research Press Web site at cjpp.nrc.ca on 6 November AS is recognized as a chronic vascular inflammatory re-
2008. sponse (Libby 2002), since inflammation is involved in all
stages of AS, from initiation through progression and, ulti-
J. Wang, R. Zhang, Y. Xu, H. Zhou, B. Wang, and S. Li.1 mately, the thrombotic complications of AS (Hansson et al.
Department of Pharmacology, Nanjing Medical University, 2006; Lucas et al. 2006). Many pathophysiologic compo-
Hanzhong Road 140, Nanjing 210029, China.
nents, which are mediated by various proinflammatory me-
1
Corresponding author (e-mail: snli@njmu.edu.cn). diators and cell adhesion molecules secreted by injured

Can. J. Physiol. Pharmacol. 86: 777–784 (2008) doi:10.1139/Y08-085 # 2008 NRC Canada
778 Can. J. Physiol. Pharmacol. Vol. 86, 2008

endothelial cells (ECs), such as nuclear factor kappa B Atherosclerotic lesion analysis
(NF-kB), vascular cell adhesion molecule-1 (VCAM-1), The proximal portion of the thoracic aorta up to the aortic
thrombin, and tumor necrosis factor-a (TNF-a), play key origin (aortic arch) was isolated and cleaned of the adherent
roles in the development of AS (Libby 2002; Pearson et al. connective tissue, and samples were cut into 10-micrometre
2003). sections. Five consecutive sections were taken at
Emerging evidence indicates that genistein exerts multi- 120-micrometre intervals from each mouse. Sections were
faceted antiinflammatory effects in vasculature (Chacko et fixed with 10% neutral buffered formalin for 30 min,
al. 2005; Marotta et al. 2006), suggesting that genistein may washed with PBS, and then treated with 60% isopropyl
prevent AS by suppressing vascular inflammation. More- alcohol for 1 min. The sections were then stained with
over, the protective effects of soy isoflavones in AS hematoxylin–eosin (HE). Images were captured with an
included a series of mechanisms such as prevention of low- Olympus BX417 microscope and an Olympus digital
density lipoprotein (LDL) oxidation, improvement of vascu- camera (Tokyo, Japan). The lipid-stained areas were ana-
lar reactivity, inhibition of proinflammatory cytokines or lyzed with Adobe Photoshop software and Scion Image
cell adhesion proteins, and reduction of platelet aggregation software. The lipid-stained area was reported as the mean
(Wenzel et al. 2008). Recent studies have demonstrated that area of the 5 sections from each mouse.
genistein inhibits hyperpermeability of cultured vascular
ECs (Liu et al. 2005) and acts as antiinflammatory and anti- Plasma measurements
atherogenic agents in human ECs (Simoncini et al. 2008). Mice were euthanized by CO2 inhalation. Blood was col-
Our previous data also showed that genistein inhibits lected from the retroorbital venous plexus. Plasma was sepa-
angiotensin-converting enzyme (ACE) (Xu et al. 2006), rated, and total cholesterol, triglycerides, LDL cholesterol,
which is a key enzyme in the development of AS (Lonn et and high-density lipoprotein (HDL) cholesterol were deter-
al. 2001). In particular, genistein was shown to be able to mined by using commercially available kits (Jiancheng Bio-
inhibit NF-kB and VCAM-1 expression in vitro (Baxa and technology, Nanjing, China). Colorimetric changes were
Yoshimura 2003; Choi et al. 2003; Dijsselbloem et al. measured at 500 nm. Total antioxidant capacity (TAC), ma-
2007; Mukherjee et al. 2003; Simoncini et al. 2008), which londialdehyde (MDA), and superoxide dismutase (SOD)
is direct proof of an antiatherosclerotic effect of genistein. were also measured by commercially available kits (Jian-
Furthermore, previous observations indicated genistein or cheng Biotechnology, Nanjing, China).
isoflavone has an antiatherosclerotic effect in rabbits (Lee
et al. 2004; Yamakoshi et al. 2000); however, these mecha- Immunohistochemistry
nisms of genistein are still not clarified in mice models in To determine the expression of NF-kB and VCAM-1 in
vivo. the vessel wall of the LDLR knockout mice, cryosections
In the present study, we studied the effect of genistein on 10 mm thick were fixed in acetone. In brief, the sections
atherogenesis in LDL receptor (LDLR) knockout mice. The were blocked in goat serum in calcium- and magnesium-
effect of genistein on lesion progression and plasma lipid free PBS plus 0.005% Triton X-100 for 30 min. Tissue
levels was observed. NF-kB and VCAM-1 expression in the sections were then incubated with anti-rabbit NF-kB p50
aortic atherosclerotic lesions was also analyzed in LDLR–/– antibody or anti-rabbit VCAM-1 antibody (Santa Cruz Bio-
mice. technology, Santa Cruz, USA) for 1 h at 37 8C. The areas
that were stained brown by anti-NF-kB p50 or anti-VCAM-1
antibodies were regarded as their respective localization and
Materials and methods were measured by the image analyzer. Quantitative analysis
Animals and treatment was measured as a percentage of the positive-stained area in
Male LDLR knockout mice (Ishibashi et al. 1994) on the total atherosclerotic lesion area.
C57BL/6 background were obtained from the Model Animal
Research Center of Nanjing University. Experiments were Tissue protein extraction and NF-kB p50 assay
performed on age-matched mice at 2.5–4 months of age. Aortas were subjected to nuclear protein extraction for the
Mice in the control group (n = 6) were maintained on stand- determination of activation of the p50 subunit, an index of
ard rat chow. Mice in the model and genistein experimental NF-kB activation (Borrás et al. 2005), following the instruc-
groups were put on a western diet consisting of 21% fat by tions of the nuclear and cytoplasmic extraction kit (Pierce,
weight, 0.15% cholesterol by weight, and no cholic acid Rockford, USA). The efficiency of extraction was deter-
(made by the Model Animal Research Center). The experi- mined by measuring the relative activity of a cytosolic en-
mental genistein group (n = 6) was given genistein (Sigma, zyme (lactate dehydrogenase) in tissues. The active form of
St. Louis, USA) at 0.3 mg/kg in 100 mL of a mixture of the p50 subunit was then detected in extracts by ELISA ac-
DMSO (1.25%) and PEG-400 (98.75%) s.c. daily. The cording to the manufacturer’s instructions (TransAM NF-kB
model group (n = 6) received genistein vehicle (100 mL of p50 Chemi, Active Motif, Carlsbad, USA).
a mixture of DMSO (1.25%) and PEG-400 (98.75%) s.c.
daily), and the control group was given placebo (normal sal- Reverse transcription (RT)-PCR amplification
ine) s.c. daily. Treatment lasted 8 weeks. Body weight and The aortas of mice were dissected and kept in liquid ni-
food intake were monitored at regular intervals. The experi- trogen. Total RNA was then extracted from the aortas by us-
ments were conducted in accordance with the official rec- ing Trizol reagent (Invitrogen, Carlsbad, USA). Two
ommendations of the Chinese Community Guidelines hundred microliters of chloroform were added to the homo-
concerning care and use of experimental animals. genates, and these were then incubated for 10 min at room
# 2008 NRC Canada
Wang et al. 779

Fig. 1. Representative micrographs showing HE-stained lesions


(A) control, (B) model, and (C) genistein LDLR–/– mice
(magnification  400). (D) Lesion size in the aortic arch stained by
HE in LDLR–/– mice. Data are means ± SD. **, Significant at p <
0.01 vs. control; #, p < 0.05 vs. model, n = 6 mice per group, 8 sec-
tions per mouse. HE, hematoxylin–eosin; LDLR, low-density lipo-
protein receptor.

temperature. After centrifugation at 12 000g for 10 min,


500 mL of isopropyl alcohol was added. The samples were
then incubated for 10 min at room temperature and centri-
fuged at 12 000g for 5 min. RNA pellets were washed with
75% ethanol and dried. RT was performed with oligo(dT).
The following oligonucleotide primer pairs were examined:
b-actin sense, 5’-GAC GGC CAG GTC ATC ACT AT-3’,
and antisense, 5’-CTT CTG CAT CCT GTC AGC AA-3’;
VCAM-1 sense, 5’-CGT CGC GAG GTT GTT TAG AGT-
3’, and antisense, 5’-CAA CAG TCA GTC CAA GCA ACA
CT-3’. PCR assays were performed for 35 cycles. Each
cycle consisted of the following steps: denaturation at 94 8C
for 30 s, annealing at 56 8C for 45 s, and extension at 72 8C
for 1 min. PCR products were analyzed in 2% agarose gel
containing ethidium bromide. Each mRNA level was ex-
pressed as a ratio to b-actin mRNA.

Statistical analysis
The data were represented as means ± SD. The compari-
son of more than 2 groups was determined by one-way
ANOVA, followed by Bonferroni test as appropriate, using
Stata 7.0 (Stata, College Station, USA). Statistical signifi-
cance was considered at p < 0.05.

Results
Reduced atherosclerotic lesions in genistein group mice
To examine the effect of genistein in vivo, genistein was
administrated to mice for 8 weeks. As a result, genistein sig-
nificantly reduced the area of atherosclerotic lesions in this
group. Figure 1 shows examples of the different stages of
AS lesion development observed on examination by light
microscope; sections of aorta also revealed significant dif-
ferences in lesion area between the genistein and model
groups. Counting by 8 consecutive sections occupied by
HE-stained areas, lesions were (6.65 ± 1.51) 106 mm2 in
the model group versus (4.68 ± 1.18) 106 mm2 in the gen-
istein group (p < 0.05).

Food intake, body weight, and plasma cholesterol and


lipid profiles
After 8 weeks of treatment, food intake and body weight
did not differ among the 3 groups (data not shown).
Although serum cholesterol and lipid profiles play an impor-
tant role in the development of AS, there is disagreement on
the effect of genistein on plasma cholesterol and lipid pro-
files. In this study, genistein had no effect on the plasma
lipid profile, which is consistent with previous reports
(Atteritano et al. 2007; Nagarajan et al. 2008). Table 1
shows that plasma total cholesterol, HDL cholesterol, LDL
cholesterol, and triglycerides were not different from those
of the model group after chronic treatment with genistein.
# 2008 NRC Canada
780 Can. J. Physiol. Pharmacol. Vol. 86, 2008

Table 1. Plasma lipid levels (mmol/L) in LDLR–/– mice main- Fig. 2. (A) TAC level in the presence or absence of genistein.
tained for 8 weeks on a high-fat diet in the presence or absence of (B) MDA level in the presence or absence of genistein. (C) SOD
genistein. activity in the presence or absence of genistein in LDLR–/– mice.
Data are means ± SD. *, Significant at p < 0.05 and **, p < 0.01
Control Model Genistein vs. control; ##, p < 0.01 vs. model, n = 6 per group. TAC, total
Total cholesterol 8.8±1.8 16.7±2.1** 15.9±1.1** antioxidant capacity; MDA, malondialdehyde; SOD, superoxide
HDL cholesterol 0.48±0.06 0.62±0.13* 0.68±0.07** dismutase.
LDL cholesterol 10.2±1.3 12.9±1.4** 13.2±1.2**
Triglycerides 0.39±0.05 0.58±0.07** 0.62±0.04**
Note: LDLR–/–, low-density lipoprotein receptor knockout. Control mice
received a standard diet. Model (vehicle) and genistein groups received a
high-fat diet. Data are means ± SD. *, Significant at p < 0.05 and **, p <
0.01 vs. control, n = 6 per group.

Plasma TAC, MDA, and SOD levels


To examine the antioxidant ability of genistein in vivo,
plasma of mice was separated to examine TAC, MDA, and
SOD levels. Compared with the model group, TAC level in
the genistein group was 85.5 ± 15.6 versus 203.4 ±
32.6 mmol/L (p < 0.01) (Fig. 2A), MDA level was 3.79 ±
0.28 versus 3.06 ± 0.31 mmol/L (p < 0.01) (Fig. 2B), and
SOD activity was 86.1 ± 6.1 versus 139.1 ± 25.1 U/mL
(p < 0.01) (Fig. 2C).

Expression of NF-kB and VCAM-1 in aorta


To investigate the mechanism of genistein’s effect on the
development of AS, we next examined the expression of
NF-kB p50 and VCAM-1. There was minimal NF-kB p50
reaction produced in the ECs of aorta in control animals.
But in model animals, the localization of NF-kB p50 was
observed in ECs of aorta, even in VSMCs. Genistein de-
creased the localization of NF-kB p50 reaction production.
As shown in Fig. 3, limited NF-kB reaction production was
observed in ECs of aorta. In addition, treatment with genis-
tein also decreased the level of the p50 subunit of NF-kB in
lysis derived from aorta (p < 0.05) (Fig. 3E).
There was minimal VCAM-1 expression in control
LDLR–/– mice. The immunostaining of this protein became
obvious, however, in the model LDLR–/– mice. Genistein
markedly reduced immunoactive VCAM-1 expression (p <
0.05) (Fig. 4). The level of VCAM-1 mRNA in LDLR–/–
mice treated with genistein (0.28 ± 0.13) was also signifi-
cantly reduced compared with that of the model group
(0.75 ± 0.15) (p < 0.01) (Fig. 5). These observations indi-
cated that genistein inhibited the development of AS, which
may be attributed to the suppression of NF-kB and VCAM-1
expression in the aorta.
During the past decade, extensive studies have largely fo-
Discussion cused on elucidating the effect of genistein on lipid profiles
Genistein is known to have a number of antiatherogenic because hyperlipidemia contributes to AS (Vitolins et al.
actions, as described in the Introduction. In vitro, genistein 2001). In our study, however, there were no differences be-
inhibited the migration and proliferation of SMCs (Pan et tween the model and genistein groups on the plasma lipids
al. 2001; Shimokado et al. 1994), which is important in pro- and lipoproteins (Table 1). This result is consistent with pre-
motion and progression of the atherosclerotic process. Geni- vious reports. Atteritano et al. (2007) showed that genistein
stein may also suppress thrombus formation by inhibiting administration did not affect blood lipid levels in postmeno-
platelet activation (Kuruvilla et al. 1993; Murphy et al. pausal women; Nagarajan et al. (2008) also indicated that
1993) and aggregation (Asahi et al. 1992; Kondo et al. feeding soy protein isolate (SPI) in mice did not affect
2002) and by reducing platelet serotonin uptake (Helmeste plasma concentrations of HDL, LDL, and total cholesterol
and Tang 1995). In the present study, our in vivo data indi- or triglycerides, suggesting that the antiatherogenic effect of
cated that genistein could decrease the area of atheroscler- genistein is not due to a change in the plasma lipids.
otic lesion in LDLR–/– mice (Fig. 1). Lipid peroxidation due to a hyperlipidemic diet may be
# 2008 NRC Canada
Wang et al. 781

Fig. 3. Immunohistochemical staining for NF-kB p50 in the aorta of LDLR–/– mice, (A) control, (B) model, and (C) genistein
(magnification  400). (D) Comparison of the NF-kB p50 protein expression among groups in LDLR–/–mice. Data are means ± SD. **, Sig-
nificant at p < 0.01 vs. control; ##, p < 0.01 vs. model, n = 6 per group. (E) Levels of the active p50 subunit of NF-kB were measured in
lysis from aorta treated with genistein. Data are means ± SD for 6 different experiments. *, Significant at p < 0.05 vs. control; #, p < 0.05
vs. model. NF-kB, nuclear factor kB; LDLR, low-density lipoprotein receptor.

the predominant factor of endothelial injury in the LDLR–/– plaques and in cultured cells (Collins and Cybulsky 2001;
mice model. Genistein has antiatherogenic effects, one po- Kutuk and Basaga 2003). Clearly, inhibition of NF-kB could
tential mechanism of which is that genistein has antioxidant attenuate the development of AS in apoE/LDLR double-
properties, including scavenging of hydrogen peroxide and knockout mice (Jawień et al. 2005). It was shown in
superoxide anion, in mRNA and protein levels (Borrás et al. previous reports that genistein can inhibit the activation of
2006; Wei et al. 1993). We further examined the oxidative NF-kB in human cancer cells (prostate, pancreatic, and
indexes of plasma in the present study. Our data demon- breast) (Gong et al. 2003; Li et al. 2004; Raffoul et al.
strated that genistein enhanced TAC and SOD levels in 2006). Bhullar et al. (1998) indicated that genistein inhibited
plasma while reducing MDA level (Fig. 2), suggesting that NF-kB by attenuating shear stress-induced IkB kinase via
the inhibition of the atherosclerotic lesion by genistein may phosphorylation and subsequent degradation of IkB protein
be due to the improvement of endothelial dysfunction via its in bovine aortic endothelial cells (BAECs). Futhermore, it
antioxidative ability. was also shown that genistein could inhibit the expression
NF-kB is one of the first mediators activated in the in- of NF-kB in dendritic cells, macrophages cells, and T lym-
flammatory response that plays a major role in atherogene- phoma cells (Baxa and Yoshimura 2003; Choi et al. 2003;
sis, and its activation has been reported in atherosclerotic Dijsselbloem et al. 2007), which play important roles in in-
# 2008 NRC Canada
782 Can. J. Physiol. Pharmacol. Vol. 86, 2008

Fig. 4. Immunohistochemical staining for VCAM-1 in the aorta of Fig. 5. RT-PCR analysis of VCAM-1 expressed in the aorta of
LDLR–/– mice (A) control, (B) model, and (C) genistein LDLR–/– mice (A) control, (B) model, and (C) genistein. (D) Com-
(magnification  400). (D) Comparison of the VCAM-1 protein ex- parison of the mRNA level of VCAM-1 among groups in LDLR–/–
pression among groups in LDLR–/– mice. *, Significant at p < 0.05 mice. Data are relative intensity versus b-actin mRNA (mean ±
and **, p < 0.01 vs. control; ##, p < 0.01 vs. model, n = 6 per SD). **, Significant at p < 0.01 vs. control; #, p < 0.05 vs. model,
group. n = 6 per group. VCAM, vascular cell adhesion molecule;
LDLR, low-density lipoprotein receptor.

flammatory responses. Taken together, these observations


showed that genistein could inhibit the expression of NF-kB
not only in ECs but also in inflammatory cells in vitro.
Therefore, we further investigated genistein’s influence on
NF-kB levels in vivo. Our data showed that genistein signif-
icantly decreased the protein expression of NF-kB p50, an
index of NF-kB activation, in atherosclerotic aortas (Fig. 3),
suggesting that genistein may suppress the development of
AS via inhibiting NF-kB activation in LDLR–/– mice.
Moreover, we found that genistein treatment led to a sig-
nificant reduction in the adhesion molecule VCAM-1. The
adhesion of circulating leukocytes to vascular ECs plays an
important role in the early stages of AS (Lombardini et al.
1997). The early steps of AS appear to be mediated through
a diverse family of cellular adhesion molecules that are ex-
pressed on the surface of vascular ECs (Price and Loscalzo
1999). Among the identified adhesion molecules involved in
the atherosclerotic process, the expression and biologic
properties of VCAM-1 are well characterized (Blankenberg
# 2008 NRC Canada
Wang et al. 783

et al. 2003). Previous reports showed that plasma soluble ment of estrogen receptors, ERK1/2, and NFkappaB. FASEB J.
VCAM-1 was significantly reduced by genistein administra- 20: 2136–2138. doi:10.1096/fj.05-5522fje. PMID:16966488.
tion in postmenopausal women (Atteritano et al. 2007; Chacko, B.K., Chandler, R.T., Mundhekar, A., Khoo, N., Pruitt, H.M.,
Colacurci et al. 2005). It is also indicated that phenoxodiol, Kucik, D.F., et al. 2005. Revealing anti-inflammatory mechanisms
a synthetic analogue of genistein, inhibited VCAM-1 expres- of soy isoflavones by flow: modulation of leukocyte-endothelial
sion in vivo (Gamble et al. 2006). Furthermore, genistein at cell interactions. Am. J. Physiol. Heart Circ. Physiol. 289: H908–
low concentrations suppressed TNF-a-induced VCAM-1 H915. doi:10.1152/ajpheart.00781.2004. PMID:15805228.
mRNA expression in human umbilical vein endothelial cells Choi, C., Cho, H., Park, J., Cho, C., and Song, Y. 2003. Suppressive
(HUVECs) (Mukherjee et al. 2003). Finally, Simoncini et al. effects of genistein on oxidative stress and NFkappaB activation
(2008) indicated that red clover extracts, particularly genis- in RAW 264.7 macrophages. Biosci. Biotechnol. Biochem. 67:
1916–1922. doi:10.1271/bbb.67.1916. PMID:14519976.
tein and daidzein, inhibited the endothelial expression of
Colacurci, N., Chiàntera, A., Fornaro, F., de Novellis, V.,
VCAM-1 induced by bacterial lipopolysaccharide, acting as
Manzella, D., Arciello, A., et al. 2005. Effects of soy
antiinflammatory and antiatherogenic agents on human ECs.
isoflavones on endothelial function in healthy postmenopausal
Our data also showed that genistein decreased VCAM-1 pro- women. Menopause, 12: 299–307. doi:10.1097/01.GME.0000
tein and VCAM-1 mRNA expression in the aorta of 147017.23173.5B. PMID:15879919.
LDLR–/– mice (Figs. 4 and 5). These results reasonably im- Collins, T., and Cybulsky, M.I. 2001. NF-kB: pivotal mediator or in-
plicate the decreased expression of VCAM-1 protein and nocent bystander in atherogenesis? J. Clin. Invest. 107: 255–264.
mRNA in the aorta in genistein’s inhibition of the develop- doi:10.1172/JCI10373. PMID:11160146.
ment of AS. Dijsselbloem, N., Goriely, S., Albarani, V., Gerlo, S., Francoz, S.,
In conclusion, genistein inhibited the progression of AS in Marine, J.C., et al. 2007. A critical role for p53 in the control of
LDLR–/– mice. The mechanisms may include its antioxida- NF-kappaB-dependent gene expression in TLR4-stimulated den-
tive action and its effect in decreasing NF-kB and VCAM-1 dritic cells exposed to Genistein. J. Immunol. 178: 5048–5057.
expression in the aorta, rather than its direct action on blood PMID:17404287.
lipid concentration. Further studies are needed for under- Dubey, R.K., Gillespie, D.G., Imthurn, B., Rosselii, M., Jackson,
standing other mechanisms of genistein’s antiatherogenic ac- E., and Keller, P. 1999. Phytoestrogens inhibit growth and
tion in detail. MAP kinase activity in human aortic smooth muscle cells. Hy-
pertension, 33: 177–182. PMID:9931101.
Acknowledgements Gamble, J.R., Xia, P., Hahn, C.N., Drew, J.J., Drogemuller, C.J.,
Brown, D., and Vadas, M.A. 2006. Phenoxodiol, an experimental
This work was supported by Key Project of Jiangsu Pro- anticancer drug, shows potent antiangiogenic properties in addi-
vincial Natural Scientific Fund (No. BK2006727). tion to its antitumour effects. Int. J. Cancer, 118: 2412–2420.
doi:10.1002/ijc.21682. PMID:16353157.
References Gong, L., Li, Y., Nedeljkovic-Kurepa, A., and Sarkar, F.H. 2003. In-
Asahi, M., Yanagi, S., Ohta, S., Inazu, T., Sakai, K., Takeuchi, F., et activation of NF-kappaB by genistein is mediated via Akt signaling
al. 1992. Thrombin-induced human platelet aggregation is pathway in breast cancer cells. Oncogene, 22: 4702–4709. doi:10.
inhibited by protein-tyrosine kinase inhibitors, ST638 and genis- 1038/sj.onc.1206583. PMID:12879015.
tein. FEBS Lett. 309: 10–14. doi:10.1016/0014-5793(92)80728-Y. Hansson, G.K., Robertson, A.K., and Söderberg-Nauclér, C. 2006. In-
PMID:1511739. flammation and atherosclerosis. Annu. Rev. Pathol. 1: 297–329.
Atteritano, M., Marini, H., Minutoli, L., Polito, F., Bitto, A., doi:10.1146/annurev.pathol.1.110304.100100. PMID:18039117.
Altavilla, D., et al. 2007. Effects of the phytoestrogen genistein Helmeste, D.M., and Tang, S.W. 1995. Tyrosine kinase inhibitors
on some predictors of cardiovascular risk in osteopenic, post- regulate serotonin uptake in platelets. Eur. J. Pharmacol. 280:
menopausal women: a two-year randomized, double-blind, R5–R7. doi:10.1016/0014-2999(95)00323-D. PMID:7589171.
placebo-controlled study. J. Clin. Endocrinol. Metab. 92: Ishibashi, S., Goldstein, J.L., Brown, M.S., Herz, J., and Burns,
3068–3075. doi:10.1210/jc.2006-2295. PMID:17682090. D.K. 1994. Massive xanthomatosis and atherosclerosis in
Baxa, D.M., and Yoshimura, F.K. 2003. Genistein reduces NF- cholesterol-fed low density lipoprotein receptor-negative mice.
kappa B in T lymphoma cells via a caspase-mediated cleavage J. Clin. Invest. 93: 1885–1893. doi:10.1172/JCI117179. PMID:
of I kappa B alpha. Biochem. Pharmacol. 66: 1009–1018. 8182121.
doi:10.1016/S0006-2952(03)00415-5. PMID:12963487. Jawień, J., Gajda, M., Mateuszuk, Ł., Olszanecki, R., Jakubowski,
Bhullar, I.S., Li, Y.S., Miao, H., Zandi, E., Kim, M., Shyy, J.Y., A., Szlachcic, A., et al. 2005. Inhibition of nuclear factor-
and Chien, S. 1998. Fluid shear stress activation of IkappaB ki- kappaB attenuates artherosclerosis in apoE/LDLR – double
nase is integrin-dependent. J. Biol. Chem. 273: 30544–30549. knockout mice. J. Physiol. Pharmacol. 56: 483–489. PMID:
doi:10.1074/jbc.273.46.30544. PMID:9804824. 16204769.
Blankenberg, S., Barbaux, S., and Tiret, L. 2003. Adhesion mole- Keys, A., Menotti, A., Karvonen, M.J., Aravanis, C., Blackburn,
cules and atherosclerosis. Atherosclerosis, 170: 191–203. H., Buzina, R., et al. 1986. The diet and 15-year death rate in
doi:10.1016/S0021-9150(03)00097-2. PMID:14612198. the seven countries study. Am. J. Epidemiol. 124: 903–915.
Borrás, C., Gambini, J., Gomez-Cabrera, M.C., Sastre, J., Pallardo, PMID:3776973.
F.V., Mann, G.E., and Vina, J. 2005. 17b-oestradiol upregulates Kirk, E.A., Sutherland, P., Wang, S.A., Chait, A., and LeBoeuf,
longevity-related, antioxidant enzyme expression via the ERK1 R.C. 1998. Dietary isoflavones reduce plasma cholesterol and
and ERK2[MAPK]/NFkappaB cascade. Aging Cell, 4: 113–118. atherosclerosis in C57BL/6 mice but not LDL receptor-deficient
doi:10.1111/j.1474-9726.2005.00151.x. PMID:15924567. mice. J. Nutr. 128: 954–959. PMID:9614153.
Borrás, C., Gambini, J., Gómez-Cabrera, M.C., Sastre, J., Pallardó, Kolonel, L.N. 1988. Variability in diet and its relation to risk in
F.V., Mann, G.E., and Viña, J. 2006. Genistein, a soy iso- ethnic and migration groups. Basic Life Sci. 43: 129–135.
flavone, up-regulates expression of antioxidant genes: involve- PMID:3365214.
# 2008 NRC Canada
784 Can. J. Physiol. Pharmacol. Vol. 86, 2008

Kondo, K., Suzuki, Y., Ikeda, Y., and Umemura, K. 2002. Genis- Nagarajan, S., Burris, R.L., Stewart, B.W., Wilkerson, J.E., and
tein, an isoflavone included in soy, inhibits thrombotic vessel Badger, T.M. 2008. Dietary soy protein isolate ameliorates
occlusion in the mouse femoral artery and in vitro platelet ag- atherosclerotic lesions in apolipoprotein E-deficient mice poten-
gregation. Eur. J. Pharmacol. 455: 53–57. doi:10.1016/S0014- tially by inhibiting monocyte chemoattractant protein-1 expres-
2999(02)02449-4. PMID:12433594. sion. J. Nutr. 138: 332–337. PMID:18203900.
Kuiper, G.G., Lemmen, J.G., Carlsson, B., Corton, J.C., Safe, S.H., Nakashima, S., Koike, T., and Nozawa, Y. 1991. Genistein, a pro-
van der Saag, P.T., et al. 1998. Interaction of estrogenic chemicals tein tyrosine kinase inhibitor, inhibits thromboxane A2-mediated
and phytoestrogens with estrogen receptor beta. Endocrinology, human platelet responses. Mol. Pharmacol. 39: 475–480.
139: 4252–4263. doi:10.1210/en.139.10.4252. PMID:9751507. PMID:2017148.
Kuruvilla, A., Putcha, G., Poulos, E., and Shearer, W.T. 1993. Nestel, P.J., Yamashita, T., Sasahara, T., Pomeroy, S., Dart, A.,
Tyrosine phosphorylation of phospholipase C concomitant with Komesaroff, P., et al. 1997. Soy isoflavones improve systemic
its activation by platelet-activating factor in a human B cell arterial compliance but not plasma lipids in menopausal and
line. J. Immunol. 151: 637–648. PMID:8393037. perimenopausal women. Arterioscler. Thromb. Vasc. Biol. 17:
Kutuk, O., and Basaga, H. 2003. Inflammation meets oxidation: 3392–3398. PMID:9437184.
NF-kB as a mediator of initial lesion development in athero- Pan, W., Ikeda, K., Takebe, M., and Yamori, Y. 2001. Genistein,
sclerosis. Trends Mol. Med. 9: 549–557. doi:10.1016/j.molmed. daidzein and glycitein inhibit growth and DNA synthesis of aor-
2003.10.007. PMID:14659470. tic smooth muscle cells from stroke-prone spontaneously hyper-
Lee, C.S., Kwon, S.J., Na, S.Y., Lim, S.P., and Lee, J.H. 2004. tensive rats. J. Nutr. 131: 1154–1158. PMID:11285318.
Genistein supplementation inhibits atherosclerosis with stabiliza- Pearson, T.A., Mensah, G.A., Alexander, R.W., Anderson, J.L.,
tion of the lesions in hypercholesterolemic rabbits. J. Korean Cannon, R.O., Criqui, M., et al. 2003. Markers of inflammation
Med. Sci. 19: 656–661. PMID:15483339. and cardiovascular disease: application to clinical and public
Li, Y., Ellis, K.L., Ali, S., El-Rayes, B.F., Nedeljkovic-Kurepa, A., health practice: a statement for healthcare professionals from
Kucuk, O., et al. 2004. Apoptosis-inducing effect of chemother- the Centers for Disease Control and Prevention and the Ameri-
apeutic agents is potentiated by soy isoflavone genistein, a nat- can Heart Association. Circulation, 107: 499–511. doi:10.1161/
ural inhibitor of NF-kappaB in BxPC-3 pancreatic cancer cell 01.CIR.0000052939.59093.45. PMID:12551878.
line. Pancreas, 28: e90–e95. doi:10.1097/00006676-200405000- Price, D.T., and Loscalzo, J. 1999. Cellular adhesion molecules and
00020. PMID:15097869. atherogenesis. Am. J. Med. 107: 85–97. doi:10.1016/S0002-
Libby, P. 2002. Inflammation in atherosclerosis. Nature, 420: 9343(99)00153-9. PMID:10403357.
868–874. doi:10.1038/nature01323. PMID:12490960. Raffoul, J.J., Wang, Y., Kucuk, O., Forman, J.D., Sarkar, F.H., and
Liu, D., Jiang, H., and Grange, R.W. 2005. Genistein activates the Hillman, G.G. 2006. Genistein inhibits radiation-induced activa-
3’,5’-cyclic adenosine monophosphate signaling pathway in vas- tion of NF-kappaB in prostate cancer cells promoting apoptosis
cular endothelial cells and protects endothelial barrier function. and G2/M cell cycle arrest. BMC Cancer, 6: 107. doi:10.1186/
Endocrinology, 146: 1312–1320. doi:10.1210/en.2004-1221. 1471-2407-6-107. PMID:16640785.
PMID:15591142. Shimokado, K., Yokota, T., Umezawa, K., Sasaguri, T., and Ogata, J.
Lombardini, R., Vaudo, G., and Ciuffetti, G. 1997. Leukocyte- 1994. Protein tyrosine kinase inhibitors inhibit chemotaxis of vas-
endothelial cell interactions and vascular injury in the earliest cular smooth muscle cells. Arterioscler. Thromb. 14: 973–981.
preclinical stages of atherosclerosis. Haematologica, 82: 253. PMID:8199189.
PMID:9175334. Simoncini, T., Garibaldi, S., Fu, X.D., Pisaneschi, S., Begliuomini,
Lonn, E., Yusuf, S., Dzavik, V., Doris, C.I., Yi, Q., Smith, S., et S., Baldacci, C., et al. 2008. Effects of phytoestrogens derived
al. 2001. Effects of ramipril and vitamin E on atherosclerosis: from red clover on atherogenic adhesion molecules in human
the study to evaluate carotid ultrasound changes in patients trea- endothelial cells. Menopause, 15: 542–550. doi:10.1097/gme.
ted with ramipril and vitamin E (SECURE). Circulation, 103: 0b013e318156f9d6. PMID:18467954.
919–925. PMID:11181464. Vitolins, M.Z., Anthony, M., and Burke, G.L. 2001. Soy protein iso-
Lucas, A.R., Korol, R., and Pepine, C.J. 2006. Inflammation in flavones, lipids and arterial disease. Curr. Opin. Lipidol. 12: 433–
atherosclerosis: some thoughts about acute coronary syndromes. 437. doi:10.1097/00041433-200108000-00010. PMID:11507329.
Circulation, 113: e728–e732. doi:10.1161/CIRCULATIONAHA. Wei, H., Wei, L., Frenkel, K., Bowen, R., and Barnes, S. 1993. In-
105.601492. PMID:16651475. hibition of tumor promoter-induced hydrogen peroxide forma-
Marotta, F., Mao, G.S., Liu, T., Chui, D.H., Lorenzetti, A., Xiao, Y., tion in vitro and in vivo by genistein. Nutr. Cancer, 20: 1–12.
and Marandola, P. 2006. Anti-inflammatory and neuroprotective PMID:8415125.
effect of a phytoestrogen compound on rat microglia. Ann. N. Y. Wenzel, U., Fuchs, D., and Daniel, H. 2008. Protective effects of
Acad. Sci. 1089: 276–281. doi:10.1196/annals.1386.033. soy-isoflavones in cardiovascular disease. Identification of mole-
PMID:17261775. cular targets. Hamostaseologie, 28: 85–88. PMID:18278168.
Mukherjee, T.K., Nathan, L., Dinh, H., Reddy, S.T., and Wong, W.W., Smith, E.O., Stuff, J.E., Hachey, D.L., Heird, W.C.,
Chaudhuri, G. 2003. 17-epiestriol, an estrogen metabolite, is and Powell, H.J. 1998. Cholesterol-lowering effect of soy pro-
more potent than estradiol in inhibiting vascular cell adhesion tein in normocholesterolemic and hypercholesterolemic men.
molecule 1 (VCAM-1) mRNA expression. J. Biol. Chem. 278: Am. J. Clin. Nutr. 68: 1385S–1389S. PMID:9848504.
11746–11752. doi:10.1074/jbc.M207800200. PMID:12547825. Xu, Y.Y., Yang, C., and Li, S.N. 2006. Effects of genistein on an-
Murphy, C.T., Kellie, S., and Westwick, J. 1993. Tyrosine-kinase giotensin-converting enzyme in rats. Life Sci. 79: 828–837.
activity in rabbit platelets stimulated with platelet-activating fac- doi:10.1016/j.lfs.2006.02.035. PMID:16626761.
tor. The effect of inhibiting tyrosine kinase with genistein on Yamakoshi, J., Piskula, M.K., Izumi, T., Tobe, K., Saito, M.,
platelet-signal-molecule elevation and functional responses. Eur. Kataoka, S., et al. 2000. Isoflavone aglycone-rich extract without
J. Biochem. 216: 639–651. doi:10.1111/j.1432-1033.1993. soy protein attenuates atherosclerosis development in cholesterol-
tb18184.x. PMID:7690703. fed rabbits. J. Nutr. 130: 1887–1893. PMID:10917898.

# 2008 NRC Canada

Você também pode gostar