Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

European Journal of Pharmacology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

European Journal of Pharmacology


journal homepage: www.elsevier.com/locate/ejphar

Full length article

Anti-atherosclerotic effect of hesperidin in LDLr−/− mice and its possible


mechanism

Ye-Zi Suna, , Jian-Fei Chenb, Li-Min Shena, Ji Zhoua, Cui-Fang Wangc
a
Department of Endocrinology, Zhang Jia Gang First People's Hospital, Zhang Jia Gang, People's Republic of China
b
Department of Traditional Chinese Medicine, Zhang Jia Gang First People's Hospital, Zhang Jia Gang, People's Republic of China
c
Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong, People's Republic of China

A R T I C L E I N F O A B S T R A C T

Keywords: Hesperidin, a citrus bioflavonoid, exerts numerous pharmacological activities. However, its protective effect
Hesperidin against atherosclerosis in vivo remains poorly understood. In the present study, we aimed to observe the effects
Atherosclerosis of hesperidin on high fat diet (HFD)-induced atherosclerosis using LDL receptor deficient (LDLr-/-) mice. After 12
Hepatic steatosis weeks of treatment, the animals were sacrificed. The blood samples were collected for further analysis. Mouse
peritoneal macrophages were collected. Hepatic lipid content, quantification of atherosclerosis, assessment of
oxidative stress and inflammation, gene expressions were performed on liver and aorta samples. The data
showed that hesperidin ameliorated HFD-induced weight gain, improved insulin resistance and ameliorated
hyperlipidemia. Hesperidin suppressed HFD-induced hepatic steatosis, atherosclerotic plaque area and macro-
phage foam cell formation. Further study showed that hesperidin down-regulated expressions of acetyl coen-
zyme A carboxylase alpha (ACCα) and fatty acid synthase (FAS) which are two key enzymes in fatty acid and
triglyceride synthesis in liver; and upregulated expression of hepatic ATP-binding cassette transporters G8
(ABCG8), macrophage ATP-binding cassette transporters A1 (ABCA1) and G1 (ABCG1) which are transporters
involved in the process of reverse cholesterol transport. Hesperidin also reduced oxidative stress by normalizing
activities of antioxidant enzymes and inflammation in HFD-fed LDLr−/− mice. These findings suggest that he-
speridin reduced atherosclerosis via its pleiotropic effects, including improvement of insulin resistance, ameli-
oration of lipid profiles, inhibition of macrophage foam cell formation, anti-oxidative effect and anti-in-
flammatory action.

1. Introduction redox status and increased levels of circulating proinflammatory factors


(Xu et al., 2015; Targher et al., 2010). Thus, manipulation of hepatic
Atherosclerosis is a chronic vascular disease process that is char- lipid metabolism, oxidative stress and inflammation is a commonly
acterized by focal narrowing and thickening of the lumen of blood pursued strategy to inhibit the progression of atherosclerosis.
vessels due to the formation of lipid-laden plaques in the luminal sur- Reverse cholesterol transport (RCT) is a cholesterol metabolism
face of arterial wall (Weber et al., 2017). Dyslipidemia, oxidative stress pathway ensuring the effux of cholesterol from lipid-laden macro-
and persistent inflammation are well-known risk factors in the devel- phages in the artery wall and its transport back to the liver for eventual
opment of atherosclerosis (Hurtubise et al., 2016). In addition, ather- excretion (Favari et al., 2015). RCT is believed to be an important de-
osclerosis is usually associated with other metabolic disorders such as fense mechanism in atherogenesis. Cholesterol eflux from the macro-
insulin resistance and fatty liver (Xu et al., 2015; Zeadin et al., 2013). phage is the first step in RCT, which is mediated by various membrane
Insulin has been reported to induce forceful effects on lipid accumula- transporters, such as ATP-binding cassette transporters G1 and A1
tion as well as expressions of genes involved in lipogenesis (Zeadin (ABCG1 and ABCA1) on the membrane of the macrophage. ABCG1
et al., 2013). The liver plays a critical metabolic role in regulating lipid mediates the effux of cholesterol and phospholipids to HDL particles,
homeostasis through many processes including uptake of cholesterol while ABCA1 induces cholesterol effux to lipid-poor apolipoprotein A-I
from the peripheral tissues, synthesis of cholesterol and triglycerides, (apoA-I). After uptaking of HDL/apoA-I-accociated cholesterol or cho-
and excretion of cholesterol into the bile. Hepatic dysfunction can ac- lesterol ester into hepatocytes by hepatic scavenger receptor class B
celerate atherosclerosis via deterioration of dyslipidemia, imbalanced type I (SR-BI), secretion of cholesterol and bile acids into the bile is


Corresponding author.
E-mail address: sunyezi1976@163.com (Y.-Z. Sun).

http://dx.doi.org/10.1016/j.ejphar.2017.09.010
Received 20 June 2017; Received in revised form 30 August 2017; Accepted 8 September 2017
0014-2999/ © 2017 Published by Elsevier B.V.

Please cite this article as: Sun, Y.-Z., European Journal of Pharmacology (2017), http://dx.doi.org/10.1016/j.ejphar.2017.09.010
Y.-Z. Sun et al. European Journal of Pharmacology xxx (xxxx) xxx–xxx

regulated by the respective hepatic transporters ABCG5 and ABCG8 (Folch et al., 1957). Triglyceride and total cholesterol concentrations
(Zhang et al., 2016). were determined using the commercial enzyme kits (Zhongsheng Bio-
Hesperidin, a specific flavonoid glycoside, can be isolated in large tech Co., Ltd, Beijing, China) and were normalized to liver protein
amounts from the rinds of some citrus species. Preclinical studies and content.
clinical trials have demonstrated hesperidin has anti-inflammatory,
anti-oxidant, lipid-lowering and insulin-sensitizing properties, con- 2.4. Quantification of atherosclerosis
tributing to its therapeutical effects in various diseases (Li and
Schluesener, 2017; Mulvihill et al., 2016). But, until now, no study has Atherosclerotic lesion severity was assessed by en face analysis of
reported the direct effect of hesperidin on atherosclerosis using LDL the aorta as previously described (Liu et al., 2017). In brief, the as-
receptor deficient (LDLr-/-) mice, which is a well established animal cending aorta and thoracic aorta were opened longitudinally, and fixed
model with many characteristics of the metabolic syndrome. Thus, the with 4% paraformaldehyde and then stained with Oil Red O (Sigma, St.
aim of the present study was to determine the potential effects of he- Louis, MO). Lesion and total areas were determined using Image-Pro
speridin on atherosclerosis and its associated metabolic disorders in Plus software 6.0.
LDLr-/- mice, and also explore the possible mechanisms involved.
2.5. Isolation of mouse peritoneal macrophages and studies of macrophage
2. Materials and methods foam cell formation

2.1. Animal experiment In a separate study, after 12 weeks, male LDLr−/− mice fed with
standard chow, HFD or HFD with hesperidin as described above were
Male LDLr−/− mice on C57BL/6JNju were purchased from Nanjing injected intraperitoneally with 1 ml of 10% thioglycolate. Three days
Biomedical Research Institute of Nanjing University. All procedures later, Isolation of mouse peritoneal macrophages was performed as
were approved by the animal care and use committee of Nantong described previously (Mo et al., 2014). The cells were resuspended in
University. The mice were housed under standard laboratory conditions RPMI-1640 supplemented with 10% FBS, 100 U/ml penicillin and
with a 12-h light/dark cycle and free access to water and food. Starting 100 mg/ml streptomycin for use in subsequent experiments. Upon
from 8 weeks, the mice were randomly separated into four groups: a fixation with paraformaldehyde (4%), the peritoneal macrophages from
control group, a high fat diet (HFD) model group, 100 mg/kg/day he- mice were stained with 0.5% oil red O and hematoxylin was used as
speridin-treated group and 200 mg/kg/day hesperidin-treated group. counterstaining. After alcohol extraction, density of the total cellular
The dose of hesperidin used in this study was based on previous reports neutral lipid content was determined by a microplate reader (absor-
of its beneficial pharmacological effects in animals with different dis- bance at 540 nm). The intracellular cholesterol was quantified using the
ease (Jung et al., 2004; Mahmoud et al., 2012; Çetin et al., 2016). Amplex Red cholesterol assay kit (Invitrogen, CA, USA) and were nor-
Hesperidin (purity > 98%) was provided by XI’AN natural field bio- malized to cell total protein which was determined by the BCA protein
technioue Co., LTD, China. The control group was fed a standard chow assay (Beyotime Biotech, China).
diet. The HFD group was given a high fat diet containing 21% fat and
0.21% cholesterol (D12079B, Open Source Diets, Research Diets, Inc). 2.6. Cholesterol efflux assay
The two hesperidin treatment groups were given the same high fat diet
and dosed daily via intragastric gavage with 100 and 200 mg/kg/day Peritoneal macrophages from untreated LDLr−/− mice at 8 weeks
hesperidin by weight for 12 weeks. Hesperidin was suspended in 0.5% old were equilibrated with 22-NBD-cholesterol (Life Technologies,
carboxymethyl cellulose (CMC). Mice in control and HFD model group Carlsbad, CA, USA) (1 μg/ml) for 24 h. Then cells were washed with
received the same volume of 0.5% CMC vehicle gastrically. At the end PBS and incubated with 0.1, 1 and 10 µM of hesperidin for an addi-
of the study, animals were fasted overnight and were sacrificed. The tional 18 h. 50 μg/ml HDL (Luwen Biotechnologies, China) or ApoA1
blood samples and tissues were collected for further analysis. (Sigma, USA) was added as the receptor protein to start the efflux ex-
periment for 6 h. The fluorescence-labeled cholesterol released from
2.2. Biochemical measurements cells into the medium was measured using a microplate reader (synergy
H1, BioTek, USA). Cholesterol efflux was expressed as a percentage of
Fasting insulin was determined using a commercial mouse insulin fluorescence in the medium relative to the total amounts of fluores-
elisa kit (Millipore, MA). Fasting glucose were measured with a mul- cence detected in cells and the medium.
tifunctional biochemistry analyzer Olympus AU2700 (Olympus, Tokyo,
Japan). Insulin resistance was evaluated by homeostasis model assess- 2.7. RNA analysis
ment (HOMA) index as described previously for mice (Mulvihill et al.,
2009). Serum high density lipoprotein cholesterol (HDL-c), low density mRNA expressions of targeted genes were quantified by real-time
lipoprotein cholesterol (LDL-c), triglyceride (TG) and total cholesterol PCR with forward and reverse primers (Supplementary Table 1). Total
(TC) were detected using the biochemical kits (Zhongsheng Bio-tech RNA was extracted using Trizol reagent and was reverse-transcribed
Co., Ltd, Beijing, China). Activities of superoxide dismutase (SOD) and into cDNA using the PrimeScript RT Master Mix Kit (Takara, TaKaRa
glutathione peroxidase (GSH-Px) were measured with enzymatic col- Biotechnology, Dalian, China). The real-time PCR were carried out on
orimetric assays using commercial kits (Nanjing Jiancheng Bioengi- StepOne Plus Real-Time PCR System (Applied Biosystems) using SYBR
neering Institute, Nanjing, China). Serum levels of ox-LDL TNF-α and Green PCR master mix (Applied Biosystems) with the resulting cDNAs.
IL-6 were measured using elisa kits (Shanghai Westang Biotechnology Housekeeping gene 18 s was used as a reference. Data were analyzed
Co. Ltd, Shanghai, China). using the cycle threshold (Ct) method.

2.3. Determination of hepatic lipid content 2.8. Western blot analysis

For Oil Red O staining, frozen liver tissues were embedded in Equal amounts of protein were separated and electrophoretically
Tissue-Tek OCT compound, sectioned at 8 µm, and stained with Oil Red transferred to nitrocellulose membranes (Bio-Rad, USA). After blocking,
O (Sigma, St. Louis, USA) to detect lipid deposition. For hepatic lipid the membranes were incubated with the first antibodies (all obtained
content measurement, total lipids were extracted from frozen hepatic from Abcam, USA) overnight. And then the membranes were incubated
tissue (approximately 50 mg) according to the modified Folch method with the appropriate IRDye 680RD secondary antibodies (1:10000) in

2
Y.-Z. Sun et al. European Journal of Pharmacology xxx (xxxx) xxx–xxx

Fig. 1. Effects of hesperidin on HFD-induced metabolic syndrome in LDLr−/− mice. (A) Body weight of LDLr−/− mice fed a chow, HFD or HFD with hesperidin diet for 0 and 12 weeks.
(B) Serum glucose level. (C) Serum insulin level. (D) HOMA index. (E) Serum TC concentrations. (F) Serum TG concentrations. (G) Serum LDL-c concentrations. (H) Serum HDL-c
concentrations. n = 12 per group. Results are presented as the mean ± S.D. #P < 0.05 vs. control group; *P < 0.05 vs. HFD alone group; ΔP < 0.05 vs. HFD+H100 group. H100,
hesperidin 100 mg/kg/day; H200, hesperidin 200 mg/kg/day.

the dark. The blot was imaged and quantitated using the Odyssey in- hesperidin administration (Fig. 2D). The mRNA abundance of genes
frared imaging system (LI-COR Biosciences, Inc.). promoting fatty acid oxidation (peroxisome proliferator-activated re-
ceptor-alpha, PPARα; camitine palmitoyltransferase 1 alpha, CPT-1α)
2.9. Statistical analysis and cholesterol biosynthesis (3-hydroxy-3-methylglutaryl-coenzyme A
reductase, HMGCR; 3-hydroxy-3-methylglutaryl-coenzyme A synthase,
Data were represented as mean ± S.D. Statistical differences were HMGCS) were not significantly altered by hesperidin supplementation
assessed by one-way ANOVA analysis of Tukey's multiple comparison compared with HFD-fed alone group. Among the three hepatic genes
test using Graphpad Prism 5 software, and P < 0.05 was considered ABCG5, ABCG8 and SR-BI which are involved in RCT, hesperidin sig-
statistically significant. nificantly increased ABCG8 mRNA expression but has little effect on
ABCG5 and SR-BI mRNA expression compared with HFD-fed alone
3. Results group (Fig. 2D). The alteration of mRNA expressions in ACCα, FAS and
ABCG8 by hesperidin administration was confirmed at protein levels
3.1. Hesperidin ameliorated HFD-induced metabolic syndrome in LDLr−/− (Fig. 2E-G).
mice
3.3. Hesperidin inhibited atherosclerotic lesion formation in HFD-fed
Compared with standard chow fed mice, LDLr-/- mice fed a HFD for LDLr−/− mice
12weeks had significantly higher body weight, obvious insulin re-
sistance and hyperlipidemia (Fig. 1). Administration with hesperidin at The potential effects of hesperidin in HFD-induced atherosclerosis in
200 mg/kg/day dosage markedly decreased body weight, fasting glu- LDLr−/− mice were then investigated. The percentage of plaque area
cose level, fasting insulin level and HOMA index (Fig. 1A-D), suggesting was significantly higher in HFD-fed LDLr−/− mice, and hesperidin
that insulin resistance caused by HFD had been alleviated. Notably, treatment significantly attenuated HFD-induced atherosclerotic lesion
hesperidin treatment (100 and 200 mg/kg/day) markedly decreased formation (Fig. 3A and B). Furthermore, the expressions of ABCA1 and
the serum levels of TC, TG and LDL-c and increased HDL-c level com- ABCG1 which are responsible for RCT in aorta were measured. The
pared with HFD alone group (Fig. 1E-H). results showed that compared to the HFD alone group, the expressions
of ABCA1 and ABCG1 both at mRNA and protein levels were markedly
3.2. Hesperidin attenuated HFD-induced hepatic steatosis in LDLr−/− mice up-regulated in hesperidin administration group (Fig. 3C-F).

Analyse of fat content on liver sections stained with Oil-red-O 3.4. Hesperidin inhibited macrophage foam cell formation in HFD-fed
showed an accumulation of many micro and macro-lipid droplets in the LDLr−/− mice
livers of HFD-fed LDLr-/- mice (Fig. 2A). Consistently, hepatic TC and
TG levels were significantly increased in HFD-fed LDLr-/- mice (Fig. 2B Peritoneal macrophages were collected from mice and analyzed for
and C). However, livers from HFD-fed LDLr-/- mice treated with he- foam-cell formation. The results of Oil-red-O staining and intracellular
speridin displayed a dramatic reduction in the amount of lipid droplets cholesterol content showed that lipid droplet staining and cholesterol
and hepatic TC and TG levels compared with HFD-fed alone group content were significantly increased in peritoneal macrophages from
(Fig. 2A-C). To further elucidate the underlying mechanisms, the mRNA HFD-fed LDLr-/- mice, which indicated that foam cells were formed
expression levels of several key genes related to lipid synthesis and (Fig. 4A-C). But administrated with hesperidin decreased the accumu-
metabolism were determined. The mRNA levels of acetyl coenzyme A lation of cholesterol and foam cell formation in peritoneal macrophages
carboxylase alpha (ACCα) and fatty acid synthase (FAS), two key en- compared with HFD-fed alone group. In addition, the expressions of
zymes in fatty acid synthesis were significantly higher in the livers from ABCA1 and ABCG1 both at mRNA and protein levels in peritoneal
HFD-fed LDLr-/- mice. But these increases were significantly reversed by macrophages were significantly up-regulated in hesperidin

3
Y.-Z. Sun et al. European Journal of Pharmacology xxx (xxxx) xxx–xxx

Fig. 2. Effects of hesperidin on HFD-induced hepatic steatosis in LDLr−/− mice. (A) Representative images of liver sections stained with Oil Red O. (B) Liver TC content (n = 12). (C)
Liver TG content (n = 12). (D) mRNA expression of ACCα, FAS, PPARα, CPT-1α, HMGCR, HMGCS, ABCG5 and ABCG8 in liver (n = 6). (E-G) Protein expression of ACCα (E), FAS (F) and
ABCG8 (G) in liver (n = 6). Results are presented as the mean ± S.D. #P < 0.05 vs. control group; *P < 0.05 vs. HFD alone group; ΔP < 0.05 vs. HFD+H100 group. H100, hesperidin
100 mg/kg/day; H200, hesperidin 200 mg/kg/day.

administration group compared with HFD-fed alone group (Fig. 4D-G). 3.5. Hesperidin reduced oxidative stress in HFD-fed LDLr−/− mice
We subsequently evaluated the effect of hesperidin on cholesterol efflux
from peritoneal macrophages in vitro. As shown in Fig. 4H and I, LDLr−/− mice fed a HFD developed high ox-LDL serum level and
treatment peritoneal macrophages from untreated LDLr−/− mice at 8 low serum levels of SOD and GSH-Px, and these effects were sig-
weeks old with hesperidin (0.1–10 μM) increased NBD-cholesterol ef- nificantly reversed by treatment with hesperidin (Fig. 5A-C). In addi-
flux to both HDL and ApoA1 in a concentration-dependent manner. tion, hesperidin at high dosage evidently increased SOD and GSH-Px

4
Y.-Z. Sun et al. European Journal of Pharmacology xxx (xxxx) xxx–xxx

Fig. 3. Effect of hesperidin on atherosclerotic lesion formation in HFD-fed LDLr−/− mice. (A) Representative images of Oil Red O staining of en face preparations of aortas and (B)
quantification of the atherosclerotic surface area of the entire aorta (n = 6). (C and D) mRNA expression of ABCA1 and ABCG1 in aorta (n = 6). (E and F) Protein expression of ABCA1
and ABCG1 in aorta (n = 6). Results are presented as the mean ± S.D. #P < 0.05 vs. control group; *P < 0.05 vs. HFD alone group. H100, hesperidin 100 mg/kg/day; H200, hesperidin
200 mg/kg/day.

activities in livers compared with HFD-fed alone group (Fig. 5D and E). Hesperidin at high dosage significantly reduced the IL-6 serum level
and hepatic IL-6 mRNA expression compared with HFD-fed alone group
(Fig. 6D-E). In addition, hesperidin had trends in decreasing aortic IL-6
3.6. Hesperidin reduced inflammation in HFD-fed LDLr−/− mice
mRNA expression compared with HFD-fed alone group, yet there was
no statistical significant difference (Fig. 6F).
LDLr-/- mice fed a HFD developed high serum level of pro-in-
flammatory cytokine TNF-α and IL-6 compared with standard chow fed
mice (Fig. 6A and D). Hepatic and aortic mRNA expressions of TNF-α 4. Discussion
and IL-6 were consistent with this finding (Fig. 6B, C, E and F). But
treatment with hesperidin significantly reduced this increase of TNF-α In the present study, LDLr-/- mice were used as an experimental
in serum level, hepatic and aortic mRNA expression (Fig. 6A-C). model of atherosclerosis. LDLr−/− mice fed a diet rich in saturated fats,

5
Y.-Z. Sun et al. European Journal of Pharmacology xxx (xxxx) xxx–xxx

Fig. 4. Effect of hesperidin on macrophage foam cell formation in HFD-fed LDLr−/− mice. (A-G) are results from LDLr−/− mice fed with standard chow diet, HFD or HFD with two
dosages of hesperidin. (A) Representative images of Oil Red O staining of mice peritoneal macrophages and (B) the density of the lipid content by measuring the absorbance at 540 nm (n
= 6). (C) Cholesterol content in mice peritoneal macrophages (n=6). (D and E) mRNA expression of ABCA1 and ABCG1 in peritoneal macrophages (n = 6). (F and G) Protein expression
of ABCA1 and ABCG1 in peritoneal macrophages (n = 6). Results are presented as the mean ± S.D. #P < 0.05 vs. control group; *P < 0.05 vs. HFD alone group. H100, hesperidin
100 mg/kg/day; H200, hesperidin 200 mg/kg/day. (H and I) Cholesterol efflux assay in vitro were performed on peritoneal macrophages from untreated LDLr−/− mice at 8 weeks old
with the indicated concentrations of hesperidin. (H) Cholesterol efflux to ApoA1 (n = 6). (I) Cholesterol efflux to HDL (n = 6). Results are presented as the mean ± S.D. aP < 0.05 vs.
untreated cell group; bP < 0.05 vs. 0.1 µM hesperidin treated cell group; cP < 0.05 vs. 1 µM hesperidin treated cell group.

showing a human ‘‘like’’ lipoprotein profile, develop hepatic steatosis LDLr-/- mice. Dyslipidemia is a significant risk factor leading to ather-
and advanced atherosclerotic lesions combined with high-fat diet-in- osclerosis. Administration of hesperidin has been shown to improve
duced obesity and insulin resistance (Emini et al., 2017; Liang et al., lipid profile in streptozotocin-induced marginal type 1 diabetic rats
2014). This is the first study show that administration of hesperidin can (Akiyama et al., 2010) and high-cholesterol-diet fed rats (Wang et al.,
attenuate hepatic steatosis and atherosclerosis in HFD-fed LDLr-/- mice. 2011). Consistent with these previous findings, this study demonstrated
Contributions of hyperglycemia and insulin resistance in the de- that administration of hesperidin increased level of HDL-c and de-
velopment of atherosclerosis have been identified recently. Insulin re- creased the serum levels of TG, TC and LDL-c in HFD-fed LDLr-/- mice,
sistance is associated with various risk factors for atherosclerosis, in- suggesting that hesperidin might benefit atherosclerosis by reducing
cluding lipid accumulation, inflammation and obesity, which lipid levels. Liver is the organ directly responsible for lipid and lipo-
subsequently promote the development of atherosclerosis (Stöhr and protein metabolism, not only through lipid synthesis and apolipopro-
Federici, 2013; Yoon et al., 2017). In the present study, administration tein production, but also through the final elimination of excess cho-
of hesperidin to HFD-fed LDLr-/- mice prevented the weight gain, in- lesterol from the body. Disturbance of lipid homeostasis in liver by
creases of glucose and insulin serum levels, and HOMA index, sug- overloading this metabolic organ with high levels of fat and cholesterol,
gesting that hesperidin may ameliorate insulin resistance in HFD-fed can lead to the pathology of fatty liver disease and peripheral lipid

6
Y.-Z. Sun et al. European Journal of Pharmacology xxx (xxxx) xxx–xxx

Fig. 5. Effect of hesperidin on oxidative stress in HFD-fed LDLr−/− mice. (A) serum level of ox-LDL (n = 11–12). (B) Serum level of SOD (n = 11–12). (C) Serum level of GSH-Px (n =
11–12). (D) SOD activity in liver (n = 6). (E) GSH-Px activity in liver (n = 6). Results are presented as the mean ± S.D. #P < 0.05 vs. control group; *P < 0.05 vs. HFD alone group;
Δ
P < 0.05 vs. HFD+H100 group. H100, hesperidin 100 mg/kg/day; H200, hesperidin 200 mg/kg/day.

accumulative diseases such as atherosclerosis (Xu et al., 2015). It has only prevented aortic fatty streak development but also notably re-
been shown that LDLr-/- mice fed a high-fat-high-cholesterol diet ra- duced lipid accumulation and steatosis in the liver. Consistent with our
pidly develop hepatic steatosis and suggested that in the setting of LDL study, Wang et al. have reported the protective effects of hesperidin
deficiency non-alcoholic fatty liver disease precedes atherosclerosis against hypercholesterolemia and fatty liver in high-cholesterol diet in
(Wouters et al., 2008; Teupser et al., 2003). The present study, for the rats (Wang et al., 2011). Next, the key molecules involved in the lipid
first time, showed that administration of hesperidin to LDLr -/- mice not metabolism pathways such as fatty acid synthesis, fatty acid oxidation,

Fig. 6. Effect of hesperidin on inflammation in HFD-fed LDLr−/− mice. (A) serum level of TNF-α (n = 11–12). mRNA expression levels of TNF-α in liver (B) and aorta (C) (n = 6). (D)
serum level of IL-6 (n = 11–12). mRNA expression levels of IL-6 in liver (E) and aorta (F) (n = 6). Results are presented as the mean ± S.D. #P < 0.05 vs. control group; *P < 0.05 vs.
HFD alone group; ΔP < 0.05 vs. HFD + H100 group. H100, hesperidin 100 mg/kg/day; H200, hesperidin 200 mg/kg/day.

7
Y.-Z. Sun et al. European Journal of Pharmacology xxx (xxxx) xxx–xxx

cholesterol biosynthesis and cholesterol output were assessed to find In summary, the present study showed for the first time that he-
out by which hesperidin regulates lipid metabolism in the liver. Our speridin can inhibit atherosclerosis via its pleiotropic effects, including
data showed that administration of hesperidin significantly decreased improvement of insulin resistance, amelioration of lipid profiles, in-
expression of ACCα and FAS which are two key enzymes in fatty acid hibition of macrophage foam cell formation, anti-oxidative effect and
and triglyceride synthesis in liver (Strable and Ntambi, 2010). Our re- anti-inflammatory action. Therefore, the present study suggested a
sults also demonstrated that hesperidin markedly up-regulated the ex- possible therapeutic application of hesperidin in atherosclerosis and
pression of hepatic ABCG8 which play a critical role in the eflux of atherosclerosis associated with metabolic dysregulation.
cholesterol into the bile (Yu et al., 2013). Hence, the down-regulation of
ACCα and FAS and upregulation of ABCG8 protein expressions in liver Financial support
by hesperidin treatment could partly explain the observed beneficial
effects of hesperidin on lipid profiles, hepatic steatosis and athero- The research was supported by Jiangsu Provincial Maternal and
sclerosis in HFD-fed LDLr-/- mice. Child Health Research Program (F201512).
Both ABCG1 and ABCA1 play an important role in facilitating excess
cholesterol removal from macrophages or foam cells to extracellular Conflict of interest statement
cholesterol acceptor such as HDL and apolipoprotein A-I (apoA-I) in the
initial step of RCT (Chistiakov et al., 2016). The development of foam The authors declare no conflict of interest.
cells, derived mainly from the accumulation of excess cholesterol in
macrophages, leads to the formation of fatty streak, complex athero- Appendix A. Supporting information
sclerotic lesion, and finally plaque rupture (Mo et al., 2014). Our data
showed that hesperidin significantly increased ABCA1 and ABCG1 ex- Supplementary data associated with this article can be found in the
pressions in the aortas with atherosclerotic lesions, suggesting that online version at http://dx.doi.org/10.1016/j.ejphar.2017.09.010.
hesperidin prevented lipid accumulation and atherosclerosis of HFD-fed
LDLr-/- mice partly through upregulating the expression of ABCA1 and References
ABCG1 in the aorta. Since macrophages and macrophage-derived foam
cells are the main cell types that lead to the accumulation of cholesterol Ahotupa, M., 2017. Oxidized lipoprotein lipids and atherosclerosis. Free Radic. Res. 51,
in the arterial wall with atherosclerotic lesions, it is reasonable to 439–447.
Akiyama, S., Katsumata, S., Suzuki, K., Ishimi, Y., Wu, J., Uehara, M., 2010. Dietary
speculate that hesperidin promotes lesional macrophage cholesterol hesperidin exerts hypoglycemic and hypolipidemic effects in streptozotocin-in-
efflux. To test this hypothesis, we isolated and cultured peritoneal ducedmarginal type 1 diabetic rats. J. Clin. Biochem. Nutr. 46, 87–92.
macrophages of mice, which is commonly used substitute for lesional Çetin, A., Çiftçi, O., Otlu, A., 2016. Protective effect of hesperidin on oxidative and his-
tological liver damage following carbontetrachloride administration in Wistar rats.
macrophages (Lin et al., 2015), for further experiments in the present Arch. Med. Sci. 12, 486–493.
study. Our data showed that hesperidin administration resulted in less Chistiakov, D.A., Bobryshev, Y.V., Orekhov, A.N., 2016. Macrophage-mediated choles-
cholesterol content associated with an enhancement of cellular cho- terol handling in atherosclerosis. J. Cell. Mol. Med. 20, 17–28.
Emini, Veseli B., Perrotta, P., De, Meyer, G.R.A., Roth, L., Van, der DoncktC., Martinet,
lesterol efflux capacity. Meanwhile, the expressions of ABCA1 and W., De, Meyer G.R.Y., 2017. Animal models of atherosclerosis. Eur. J. Pharmacol.
ABCG1 in peritoneal macrophages were upregulated in hesperidin- S0014–2999, 30320–30325.
treated mice. In accordance with our finding, Iio et al. reported that Favari, E., Chroni, A., Tietge, U.J., Zanotti, I., Escolà-Gil, J.C., Bernini, F., 2015.
Cholesterol efflux and reverse cholesterol transport. Handb. Exp. Pharmacol. 224,
hesperetin can upregulates ABCA1 expression and promotes cholesterol
181–206.
efflux from THP-1 Macrophages. Besides macrophage transporters, Folch, J., Lees, M., Sloane Stanley, G.H., 1957. Sloane Stanley, A simple method for the
several hepatic transporters are also involved in the process of RCT, isolation and purification of total lipides from animal tissues. J. Biol. Chem. 226,
including SR-BI responsible for the direct selective uptake of HDL-c by 497–509.
Hurtubise, J., McLellan, K., Durr, K., Onasanya, O., Nwabuko, D., Ndisang, J.F., 2016. The
the liver and ABCG5/ABCG8 mediating the secretion of cholesterol into different facets of dyslipidemia and hypertension in atherosclerosis. Curr.
the bile. However, in the present study, the expression of hepatic SR-BI Atheroscler. Rep. 18, 82.
and ABCG5 were not altered in hesperidin-treated mice. These finding Jung, U.J., Lee, M.K., Jeong, K.S., Choi, M.S., 2004. The hypoglycemic effects of he-
speridin and naringin are partly mediated by hepatic glucose-regulating enzymes in
suggest that hesperidin may exert anti-atherosclerotic effects by upre- C57BL/KsJ-db/db mice. J. Nutr. 134, 2499–2503.
gulating the expression of transporters involved in the process of RCT, Li, C., Schluesener, H., 2017. Health-promoting effects of the citrus flavanone hesperidin.
including hepatic ABCG8, macrophage ABCA1 and ABCG1. Crit. Rev. Food Sci. Nutr. 57, 613–631.
Liu, T.T., Zeng, Y., Tang, K., Chen, X., Zhang, W., Xu, X.L., 2017. Dihydromyricetin
Oxidative stress and inflammation play crucial roles in the patho- ameliorates atherosclerosis in LDL receptor deficient mice. Atherosclerosis 262,
genesis of atherosclerosis. Oxidative stress is defined as a state of cel- 39–50.
lular redox imbalance in which pro-oxidants overwhelm antioxidant Liang, W., Menke, A.L., Driessen, A., Koek, G.H., Lindeman, J.H., Stoop, R., Havekes,
L.M., Kleemann, R., van, den Hoek A.M., 2014. Establishment of a general NAFLD
capacity, leading to increased ROS production. Excessive ROS cause
scoring system for rodent models and comparison to humanliver pathology. PLoS One
multiple pathophysiological actions in the vessel wall, including pro- 9, e115922.
moting oxidative modification of LDL to form ox-LDL which aggravates Lin, W., Liu, C., Yang, H., Wang, W., Ling, W., Wang, D., 2015. Chicory, a typical vege-
table in Mediterranean diet, exerts a therapeutic role in establishedatherosclerosis in
subendothelial lipid accumulation, accelerates inflammation and pro-
apolipoprotein E-deficient mice. Mol. Nutr. Food Res. 59, 1803–1813.
motes atherosclerotic lesion formation (Ahotupa, 2017). Hesperidin, as Mahmoud, A.M., Ashour, M.B., Abdel-Moneim, A., Ahmed, O.M., 2012. Hesperidin and
a member of flavonoids, has been proved to possess potent anti-oxi- naringin attenuate hyperglycemia-mediated oxidative stress and proinflammatory
dative and anti-inflammatory properties (Li and Schluesener, 2017). In cytokine production in high fat fed/streptozotocin-induced type 2 diabetic rats. J.
Diabetes Complicat. 26, 483–490.
the present study, hesperidin administration increased antioxidant en- Mo, Z.C., Xiao, J., Tang, S.L., Ouyang, X.P., He, P.P., Lv, Y.C., Long, Z.F., Yao, F., Tan,
zymes SOD and GSH-Px both in serum and liver of HFD-fed LDLr-/- Y.L., Xie, W., Zhang, M., Liu, D., Tian, G.P., Tang, D.P., Zheng, X.L., Zhao, G.J., Tang,
mice, which may strengthen the endogenous defense against oxidative C.K., 2014. Advanced oxidation protein products exacerbates lipid accumulation and
atherosclerosisthrough downregulation of ATP-binding cassette transporter A1 and
stress in vivo. Consistently, an obvious decrease in serum ox-LDL level G1 expression inapolipoprotein E knockout mice. Circ. J. 78, 2760–2770.
in hesperidin-treated mice was observed. In addition, administration of Mulvihill, E.E., Burke, A.C., Huff, M.W., 2016. Citrus Flavonoids as Regulators of
hesperidin showed significant reductions in serum levels of IL-6 and Lipoprotein Metabolism and Atherosclerosis. Annu. Rev. Nutr. 36, 275–299.
Mulvihill, E.E., Allister, E.M., Sutherland, B.G., Telford, D.E., Sawyez, C.G., Edwards, J.Y.,
TNF-α as well as mRNA expression of these two pro-inflammatory cy- Markle, J.M., Hegele, R.A., Huff, M.W., 2009. Naringenin prevents dyslipidemia,
tokines in liver and aorta. Taken together, the present study indicated apolipoprotein B overproduction, and hyperinsulinemia inLDL receptor-null mice
that administration of hesperidin can alleviate oxidative stress and in- with diet-induced insulin resistance. Diabetes 58, 2198–2210.
Stöhr, R., Federici, M., 2013. Insulin resistance and atherosclerosis: convergence between
flammation, which may contribute to its inhibitory effects on athero-
metabolic pathways and inflammatory nodes. Biochem J. 454, 1–11. http://dx.doi.
sclerosis.

8
Y.-Z. Sun et al. European Journal of Pharmacology xxx (xxxx) xxx–xxx

org/10.1042/BJ20130121. A., van, Kruchten R., Maeda, N., Staels, B., van, Bilsen M., Shiri-Sverdlov, R., Hofker,
Strable, M.S., Ntambi, J.M., 2010. Genetic control of de novo lipogenesis: role in diet- M.H., 2008. Dietary cholesterol, rather than liver steatosis, leads to hepatic in-
induced obesity. Crit. Rev. Biochem. Mol. Biol. 45, 199–214. flammation in hyperlipidemic mouse models of nonalcoholic steatohepatitis.
Targher, G., Day, C.P., Bonora, E., 2010. Risk of cardiovascular disease in patients with Hepatology 48, 474–486.
nonalcoholic fatty liver disease. N. Engl. J. Med. 363, 1341–1350. Xu, X., Lu, L., Dong, Q., Li, X., Zhang, N., Xin, Y., Xuan, S., 2015. Research advances in the
Teupser, D., Persky, A.D., Breslow, J.L., 2003. Induction of atherosclerosis by low-fat, relationship between nonalcoholic fatty liver disease and atherosclerosis. Lipids
semisynthetic diets in LDL receptor-deficient C57BL/6J and FVB/NJ mice: compar- Health Dis. 14, 158.
ison of lesions of the aortic root, brachiocephalic artery, and whole aorta (en face Yoon, J.J., Lee, Y.J., Han, B.H., Choi, E.S., Kho, M.C., Park, J.H., Ahn, Y.M., Kim, H.Y.,
measurement), Arterioscler. Thromb. Vasc. Biol. 23, 1907–1913. Kang, D.G., Lee, H.S., 2017. Protective effect of betulinic acid on early atherosclerosis
Wang, X., Hasegawa, J., Kitamura, Y., Wang, Z., Matsuda, A., Shinoda, W., Miura, N., in diabetic apolipoprotein-E gene knockout mice. Eur. J. Pharmacol. 796, 224–232.
Kimura, K., 2011. Effects of hesperidin on the progression of hypercholesterolemia Yu, X.H., Qian, K., Jiang, N., Zheng, X.L., Cayabyab, F.S., Tang, C.K., 2013. ABCG5/
and fatty liver induced byhigh-cholesterol diet in rats. J. Pharmacol. Sci. 117, ABCG8 in cholesterol excretion and atherosclerosis. Clin. Chim. Acta 428, 82–88.
129–138. Zeadin, M.G., Petlura, C.I., Werstuck, G.H., 2013. Molecular mechanisms linking diabetes
Weber, C., Badimon, L., Mach, F., van der Vorst, E.P.C., 2017. Therapeutic strategies for to the accelerated development of atherosclerosis. Can. J. Diabetes 37, 345–350.
atherosclerosis and atherothrombosis: past, present and future. Thromb. Haemost. Zhang, Y., Si, Y., Zhai, L., Guo, S., Zhao, J., Sang, H., Pang, X., Zhang, X., Chen, A., Qin, S.,
117, 1258–1264. 2016. Celastrus orbiculatus thunb. reduces lipid accumulation by promoting reverse
Wouters, K., van, Gorp P.J., Bieghs, V., Gijbels, M.J., Duimel, H., Lütjohann, D., Kerksiek, cholesterol transport in hyperlipidemic mice. Lipids 51, 677–692.

You might also like