You are on page 1of 16

Lipids

DOI 10.1007/s11745-016-4145-x

ORIGINAL ARTICLE

Celastrus Orbiculatus Thunb. Reduces Lipid Accumulation


by Promoting Reverse Cholesterol Transport in Hyperlipidemic
Mice
Ying Zhang1 · Yanhong Si1 · Lei Zhai1 · Shoudong Guo1 · Jilong Zhao2 · Hui Sang1 ·
Xiaofei Pang1 · Xue Zhang1 · Anbin Chen1 · Shucun Qin1 

Received: 14 January 2016 / Accepted: 15 March 2016


© AOCS 2016

Abstract  Previously, we found that Celastrus orbiculatus COT-treated mice. An in vitro isotope tracing experiment
Thunb. (COT) decreases athero-susceptibility in lipopro- showed that COT and its bioactive ingredients, such as
teins and the aorta of guinea pigs fed a high-fat diet, and celastrol, ursolic acid, oleanolic acid, and quercetin, sig-
increases high-density lipoprotein (HDL). In the present nificantly increased the efflux of 3H-cholesterol. They also
study, we investigated the effect of COT in reducing lipid increased the expression of SRB1, ABCA1, and ABCG1
accumulation and promoting reverse cholesterol transport significantly in macrophages. Our findings provided a posi-
(RCT) in vivo and vitro. Healthy male mice were treated tive role of COT in reducing lipid accumulation by pro-
with high-fat diet alone, high-fat diet with COT (10.0 g/ moting RCT. These effects may be achieved by activating
kg/d), or general fodder for 6 weeks. Serum levels of total the SRB1 and ABC transporter pathway and promoting
cholesterol (TC), triglyceride (TG), HDL-C, non-HDL-C, cholesterol metabolism via the CYP7A1 pathway in vivo.
and 3H-cholesterol in plasma, liver, bile, and feces were The effective ingredients in vitro are celastrol, ursolic acid,
determined. Pathological changes and the levels of TC oleanolic acid, and quercetin.
and TG in liver were examined. The expression of hepatic
genes and protein associated with RCT were analyzed. Keywords  Celastrus orbiculatus Thunb. · Lipid
COT administration reduced lipid accumulation in the accumulation · Reverse cholesterol transport · Cholesterol
liver, ameliorated the pathological changes, and less- efflux · Isotope tracing
ened liver injury, the levels of TG, TC, and non-HDL-C
in plasma were decreased significantly, and COT led to a Abbreviations
significant increase in plasma HDL-C and apolipopro- COT  Celastrus orbiculatus Thunb
tein A (apoA1). 3H-cholesterol in plasma, liver, bile, and RCT Reverse cholesterol transport
feces was also significantly increased in COT-treated mice HDL-C High-density lipoprotein cholesterol
compared to controls. Both mRNA and protein expression apoA1 Apolipoprotein  A
of SRB1, CYP7A1, LDLR, ATP-binding cassette trans- TC Total cholesterol
porters ABCA1, ABCG5, and LXRα were improved in TG Triglyceride
SR-B1 Scavenger receptor B1
ABCA1 ATP-binding cassette transporter G1
Y. Zhang, Y. Si contributed equally to this study.
ABCG1 ATP-binding cassette transporter A1
CE Cholesterol ester
* Shucun Qin ABCG5 ATP-binding cassette transporters G5
sdyrx@163.com ABCG8 ATP-binding cassette transporters G8
1 CYP7A1 Cholesterol 7-alpha hydroxylase
Key Laboratory of Atherosclerosis in the Universities
of Shandong and Institute of Atherosclerosis, Taishan SR-B1 Scavenger receptor class B type 1
Medical University, Tai’an 271000, Shandong, China LXR Liver X receptor
2
Shandong Agricutural University, Tai’an 271000, Shandong, TCMs Traditional Chinese medicines
China AS Atherosclerosis

13
Lipids

SDS-PAGE Sodium dodecyl sulfate polyacrylamide gel are of interest as potential new drugs for the treatment of
electrophoresis atherosclerosis.
CL Celastrol Celastrus orbiculatus Thunb. (COT), a member of
RT Rutin the Celastraceae family, is used to prevent and cure can-
QC Quercetin cer and inflammatory diseases not only in China but also
KF Kaempferol in the United States and several European countries [9].
OA Oleanolic acid COT is a traditional herb and has been used for thousands
UA Ursolic acid of years in China as a remedy against cancer, inflammatory
RT-PCR Real time PCR diseases, and neurodegenerative diseases [10] due to its
LDL-R Low-density lipoprotein receptor anti-oxidative and anti-inflammatory properties. The com-
LSC Liquid scintillation counter position of COT is complex, including such compounds
as sesquiterpenes, flavonoids, and tripenes. Our previous
study showed that COT decreases athero-susceptibility
Introduction in lipoproteins and the aorta of guinea pigs fed a high-fat
diet by down-regulating the level of non-HDL-C in plasma
Atherosclerotic cardiovascular disease is the leading cause [11]. In this study, the effect of COT on RCT was investi-
of morbidity and mortality in modern societies [1]. It is gated in C57BL/6J mice, and its bioactive ingredients were
well known that elevated levels of plasma total cholesterol screened by an isotopic tracing assay in vitro.
(TC) and triglyceride (TG) are associated with the devel-
opment of atherosclerotic cardiovascular disease. Reverse
cholesterol transport (RCT) is a cholesterol metabolism Materials and Methods
pathway that transports cholesterol from the macrophage
foam cell in atherosclerotic plaque to the liver for excre- Ethics Statement
tion in the bile [2]. Increasing evidence indicates that RCT
is a very important defense mechanism in atherosclerotic This study was approved by the Laboratory Animal Care
diseases. The primary function of high density lipopro- Committee of Taishan Medical University, and all animal
tein (HDL) is to mediate RCT, and cholesterol in HDL is experiments were conducted in accordance with the Guide-
transported into hepatocytes through the scavenger recep- lines of Care and Use of Laboratory Animals at the Taishan
tor B1 (SRB1) [3]. Cholesterol efflux from the macrophage Medical University.
to HDL is mediated by ATP-binding cassette transporters
A1 and G1 (ABCA1 and ABCG1) on the membrane of the
macrophage, and then cholesterol or cholesterol ester (CE) Materials
is transferred to the liver by HDL for bile acid synthesis
or excretion in the feces by ATP-binding cassette transport- Seven-week-old male C57BL/6J mice (SCXK2009-0004)
ers G5 and G8 (ABCG5 and ABCG8) [2]. Another essen- were obtained from Hua Fu Kang Biological Technol-
tial enzyme involving RCT is CYP7A1, the rate-limiting ogy Co., Ltd. (Beijing, China). The mice accessed diet
enzyme of bile acid synthesis. Expression of CYP7A1 is and water ad libitum and were kept in a temperature and
down-regulated by sterol regulatory element binding pro- humidity-controlled room with a 12/12 h light–dark cycle.
teins when plasma cholesterol levels are low and up-regu- Raw 264.7 macrophage was obtained from the Cell Bank
lated by liver X receptor (LXR) when cholesterol levels are of the Chinese Academy of Sciences, Shanghai Institute
high [4, 5]. of Cell Biology (Shanghai, China). Primary antibodies
Various anti-atherosclerosis medications or bioactive of CYP7A1, SRB1, LDLR, LXRα, ABCA1, apoA1, and
components are reported to promote RCT by improving the ABCG1 were obtained from Abcam (Cambridge, MA,
expression of certain transporters or key enzymes involved USA). Antibodies for β-actin were obtained from Santa
[5, 6]. Cholesterol efflux is the initial step of RCT and has Cruz Biotechnology, Inc. (Santa Cruz, CA, USA). Stand-
become one of the therapeutic targets for atherosclerotic ards of rutin, kaempferol, quercetin, celastrol, ursolic acid,
disease [7]. Statins are the preferred drugs for stabilizing and oleanolic acid were purchased from the Drug and Bio-
atherosclerotic plaques because of their lipid-lowering, logical Products Examination Center of China (Beijing,
anti-inflammatory and endothelial-protection activities. China); purity of standards was above 98 % (w/w). The
However, they exhibit side effects and are effective in only primers for PCR were synthesized by Sangon Biotech Co.,
one-third of patients [8]. Some natural products, especially Ltd (Shanghai, China). TC and TG kits were purchased
traditional Chinese medicines, which possess similar lipid- from Applygen Technologies Inc. (Beijing, China). The
lowering, anti-inflammatory, and antioxidant activities, stems and roots of COT were collected from Mountain

13
Lipids

Tai in China and identified by Professor Li Tongde in the methods (TBARS). Ox-LDL was identified using agarose
School of Pharmacy, Taishan Medical University. 3H-cho- gel electrophoresis.
lesterol was obtained from Perkin Elmer (Waltham, MA,
3
USA). Chow diet and high fat diet were obtained from Hua H‑Cholesterol‑Laden Cell Preparation
Fu Kang Biological Technology Co., Ltd. (Beijing, China).
3
H-cholesterol-laden cells were prepared as described pre-
Preparation of COT Extract viously [15]. Briefly, Raw 264.7 cells were grown in RPMI/
HEPES supplemented with 10 % FBS. Cells were cultured
The stems and roots were dried and minced using a grinder, in Teflon flasks and radio-labeled with 5 µCi/mL 3H-cho-
and the powder (1000 g) was soaked with 95 % ethanol lesterol and cholesterol-loaded with 100 µg/mL ox-LDL.
overnight (1:15, w:v). The ethanol extract of COT was fil- At 48 h later, cells were washed with RPMI/HEPES and
tered and concentrated by a rotary evaporator, and 11.34 g equilibrated for 4 h in fresh RPMI/HEPES supplemented
crude extract was obtained. Then the concentrate was with 0.2 % BSA, then harvested for mice intraperitoneal
diluted to prepare solutions of 10.0 g crude drug/mL with injection. The ratio of intracellular 3H-cholesterol and
5 % sodium carboxymethylcellulose (CMC), which were total 3H-cholesterol in the cell suspension was determined
stored at 4 °C. Several important ingredients contained in by liquid scintillation (Beckman Ls 6500, Beckman-Co.,
the stem and root were detected by capillary electrophore- Carlsbad, California, USA).
sis equipment (Beckman Coulter, Inc. Brea, CA, USA) as
shown in Table 1. Animal Management

Preparation of ox‑LDL Thirty-six male mice were divided into three groups
(n  = 12): regular chow diet (CD), high-fat diet (HFD,
Oxidized-low density lipoprotein (ox-LDL) was prepared 15.8 % fat and 1.25 % cholesterol), and HFD with COT
as described in a previous study [12]. Lipoprotein isolation (HFD + COT). They were fed daily by gastric gavage with
was done by sequential ultracentrifugation in an LE-80 K COT (10.0 g COT/kg/day mouse weight, dissolved in 5 %
ultracentrifuge (Beckman Coulter, Inc.) as described pre- sodium CMC at a dose of 10 mL/kg mouse weight for 12
viously [13]. LDL was isolated at a density of 1.063 g/ mice) or 5 % sodium CMC (10 mL/kg mouse weight) only
mL and HDL3 was isolated at a density of 1.125–1.21 g/ for the CD and HFD groups. After 6 weeks, six mice in
mL [14]. The purified lipoproteins were dialyzed against each group were selected and 3H-cholesterol-labeled foam
phosphate-buffered saline (PBS) for 48 h. LDL was incu- cells (6.0 × 106 cells containing 6.1 × 105 counts per min-
bated and dialyzed in PBS (pH 7.2) containing 10 μmol/L ute in 0.5 mL MEM) were injected intraperitoneally into
copper sulfate (CuSO4) for 12 h at 37 °C, and further dia- each mouse. Blood was collected at 0, 12, 24, and 48 h
lyzed in PBS containing 100 mmol/L ethylene diamine after injection. Feces were collected continuously at 24
tetraacetic acid (EDTA) for 24 h at 4 °C. Protein was and 48 h and stored at 4 °C before extraction of cholesterol
determined with a BCA assay kit, and the protein concen- and bile acid. At study termination (48 h after injection),
tration was adjusted to 1 g/L and stored at 4 °C. Malon- mice were exsanguinated and perfused with cold PBS.
dialdehyde (MDA) was determined by thibarbituric acid Liver and intestines were removed and stored at −70 °C for
3
H-cholesterol quantification by liquid scintillation. Plasma
and bile were also analyzed using liquid scintillation. The
Table 1  The contents of bioactive ingredients (OA, UA, CL, RT, KF,
QC) in Celastrus orbiculatus Thunb remaining mice were used for detection of plasma lipids
(TC, TG, HDL-C, non-HDL-C) by an enzymatic method,
Active ingredientsa Contents in sample (mg/g)
and the levels of apolipoprotein A1 (apoA1) in plasma
Root Stem were analyzed by western blot. The levels of TC and TG in
liver were examined by an enzymatic method; histological
OA 0.26 ± 0.045 0.67 ± 0.051
change of liver was detected by HE staining. mRNA and
UA 0.37 ± 0.026 0.16 ± 0.047
protein expression were analyzed by real time polymerase
CL 5.13 ± 0.27 1.51 ± 0.29
chain reaction (RT-PCR) and western blot.
RT – 1.80 ± 0.32
KF 0.16 ± 0.031 2.80 ± 0.24
Lipids Change in Animal Liver and Pathological
QC 0.017 ± 0.009 0.0072 ± 0.15
Examination
a
  OA oleanolic acid, UA ursolic acid, CL celastrol, RT rutin, KF
kaempferol, QC quercetin In order to observe lipids changing in animal liver, the
b
  Data are mean ± SD of at least three independent samples levels of TG and TC in animal liver were detected by an

13
Lipids

enzymatic method according to the manufacturer’s instruc- dry milk for 2 h at room temperature, the membranes were
tions. In addition, a portion of liver tissue instantly fixed in incubated with antibody and β-actin overnight at 4 °C.
10 % phosphate buffered formalin was processed by rou- After being washed four times with TBS containing 0.1 %
tine histological procedures and then embedded in paraffin. Tween 20, the membranes were incubated with horse-
Tissue Sections (5 μm) were stained with HE for histo- radish peroxidase-conjugated antibodies for 1 h at room
pathological examination. temperature. Protein levels of ABCA1, SRB1, CYP7A1,
apoA1, LXRα, and LDLR were evaluated and referenced
Real Time PCR Experiments with Animal Liver with β-actin by western blot. The content of apoA1 in
plasma was also tested by western blot. Immunoblots were
Total liver mRNA was isolated by TRIZOL reagent (Invit- revealed by enhanced chemiluminescence reaction and
rogen, Carlsbad, CA, USA). The cDNA synthesis was per- visualized using a high-performance chemiluminescence
formed using MuLV Reverse Transcriptase (Applied Bio- film. The integrated optical density value of immunoreac-
systems, Foster, CA, USA). Real-time PCR was performed tive bands were quantified using Image-Pro Plus software
using an RT-PCR automatic analyzer (Rotor-Geneq, Qia- ver.6.0 (Media Cybernetics Corp, Bethesda, MD, USA)
gen, Germany) and a SYBR-green PCR master mix kit and normalized by β-actin.
(TianGen Biotech, Beijing, China). The data was analyzed
by using Rotor-gene Q software ver.1.7 (Qiagen). Relative Cell Culture and Cytotoxicity Assays
mRNA levels were calculated by the method of 2−∆∆Ct
and normalized against GAPDH. Each experiment was To find a suitable concentration for each compound, the
repeated three times. The primers (Sangon Biotech Co. Ltd, classical 3-(4,5)-dimethylthiahiazo (-z-yl)-3,5-diphe-
Shanghai) used for real time PCR are listed in Table 2. nytetrazoliumromide (MTT) method was used to evalu-
ate cell viability of the Raw 264.7 macrophage. Raw
Western Blot Experiments with Animal Liver 264.7 macrophages were cultured in high sugar DMEM
medium with 10 % (v/v) fetal calf serum, antibiotics (100
Fresh livers were harvested and homogenized. Then, the U/mL penicillin A, and 100 μg/mL streptomycin) and
total protein was extracted from liver tissue using a total maintained in a 37 °C humidified incubator containing
protein extraction kit (BestBio, Shanghai, China) accord- 5 % CO2. The effects of active ingredients on the varia-
ing to the manufacturer’s instructions. Equal amounts of tion of cell numbers were determined by the MTT assay.
protein were separated by 8–12 % sodium dodecyl sul- In total, 1 × 104 cells were seeded in 96-well plates. COT
fate polyacrylamide gel electrophoresis (SDS-PAGE) and and the active ingredients, such as rutin (RT), quercetin
transferred onto polyvinylidene fluoride (PVDF) mem- (QC), oleanolic acid (OA), and kaempferol (KF), were
branes by electroblotting. After blocking in Tris-buffered dissolved in DMSO and different concentration solu-
saline (TBS) containing 0.1 % Tween 20 and 10 % nonfat tions (0, 50, 100, 150, 200, and 400 μg/mL) were pre-
pared. Celastrol (CL) concentrations were 0, 0.1, 0.2,
Table 2  Primers used for real time PCR analysis
0.4, 0.6, and 0.8 μg/mL and ursolic acid (UA) concen-
trations were 0, 5, 10, 20, 40, and 60 μg/mL. Exponen-
Gene Primer Sequence (5′–3′) tially growing Raw 264.7 macrophages were incubated
ABCG5 Sense TGCCCTTTCTGAGTCCAGAG with active ingredients at various concentrations for 24 h
Anti-sense GTGCTCTTTCAATGTTCTCCAG before the MTT assay was performed. For this test, sam-
ABCG8 Sense ATGAGCTGGAAGACGGGCTG ples containing the proper concentration of DMSO were
Anti-sense GCCAGTGAGA GCAAGGCTGA used as a control. At the end of the cell growing period,
LDLR Sense AGGCTGTGGGCTCCATAGG
after being incubated with the drugs, Raw 264.7 mac-
Anti-sense TGCGGTCCAGGGTCATCT
rophages were washed twice with PBS, re-supplemented
SR-B1 Sense TCCCCATGAACTG TTCTGTGGA
with 200 μL culture medium containing 10 % MTT dye,
and incubated for 4 h. Cells were washed twice with PBS
Anti-sense TGCCCGATGCCCTTGA
and then suspended in 100 μL DMSO. The plates were
CYP7A1 Sense AGCAACTA AACAACCTGCCAGTACTA
placed in the dark, and continuous gentle shaking was
Anti-sense GTCCGGATATTCAAGGATG CA
applied for 30 min to dissolve the MTT dye thoroughly.
LXRα Sense CGACAGAGCTTCCGTCCACAA
The spectrometric absorbance was measured using a
Anti-sense GCTCGTTCCCCAGCAT TTT
micro-plate reader at 490 nm. In this experiment, the
GAPDH Sense TGGCCTCCAAGGAGTAAGAAAC
maximum tolerated concentration of COT and the active
Anti-sense GGGATAGGGCCTCTCTTGCT
ingredients for cells were detected.

13
Lipids

Cholesterol Efflux Experiments in Macrophages protein extracted from cultured cells was separated by
SDS-PAGE. The proteins were transferred on PVDF mem-
Raw 264.7 macrophages were co-cultured with 1 µCi/mL branes. Protein levels of ABCA1, ABCG1, and SRB1 were
3
H-cholesterol and cholesterol loaded with 100 µg/mL ox- evaluated and referenced with β-actin by western blot. The
LDL for 24 h and divided into five groups for each drug: target bands were quantified using Image-Pro Plus software
control group, HDL3 group, and three drug treatment ver. 6.0 (Media Cybernetics Corp.).
groups. HDL3 (100 μg/mL) and drugs were added to cul-
ture for 24 h. According to the cell cytotoxicity assay, the Statistical Analysis
maximum tolerated concentration of COT and its four com-
ponents (RT, QC, OA and KF) were 150 μg/mL and CL Data were reported as mean ± standard deviation (SD) and
concentrations were 0.4 μg/mL. UA concentrations were subjected to analysis-of-variance analysis. F-test and the
20 μg/mL. For this test, samples containing 3H-cholesterol Student-Neuman-Keuls post-test analyses were performed
and ox-LDL were used as controls; samples containing on these data to analyze the variances and significances
HDL3 were positive groups. At the end of the cell grow- between groups. Probability values less than 0.05 were
ing period, the ratios of 3H-cholesterol and total 3H-cho- considered significant.
lesterol intracellular were determined by liquid scintilla-
tion. For the determination of extracellular 3H-cholesterol,
culture solution was transferred into a borosilicate glass Results
tube, cells were rinsed twice with PBS (1 mL/time), and
the washing fluid was transferred into the borosilicate glass Effects of COT on Body and Liver Weight of Mice
tube. Solution was centrifuged (2000 rpm/min) for 10 min
at 4 °C. Then 200 μL supernatant and 2.5 mL scintillation Body weight was measured at week 0 and 6 and liver
solution were blended together, and the value of extracel- weight was measured at week 6. After 6 weeks treatment,
lular 3H-cholesterol was determined. For the determina- a significant difference in the body weight was observed in
tion of intracellular 3H-cholesterol, cells were added into each group. As shown in Table 3, compared with the CD
a 2 mL intermixture of normal hexane and isopropanol group, body weight was increased by 18.63 % after HFD
(3:2  = v:v) and incubated for 30 min at a room tempera- treatment. COT treatment prevented increases in body
ture, eddied, and mixed for 5 min, then intracellular lipids weight due to the HFD (mice fed vehicle) by 15.33 %.
were extracted. This was done twice. The 200 μL super- Liver weight was increased significantly by 23.15 % in the
natant and 2.5 mL scintillation solution were blended HFD group compared with the CD group. COT prevented
together, and the values of intracellular 3H-cholesterol were increases significantly by 17.29 % in liver weight due to the
determined. The ratio of total cholesterol efflux was calcu- HFD.
lated as the sum of intracellular 3H-cholesterol and extra-
cellular 3H-cholesterol. In this study, more than 95 % of the Effects of COT on Plasma Lipids
3
H-cholesterol was taken up by cultured macrophages.
The levels of TG, non-HDL-C, and TC in plasma were
Experiments with COT and its bioactive Ingredients increased significantly in the HFD group compared to the
for ABCA1, ABCG1 and SRB1 Expression CD group (P < 0.05; P < 0.01), and decreased by 15.26,
in Macrophages
Table 3  Comparison of body weight and liver weight of mice
Raw 264.7 macrophages were cultured in high sugar
Groupa Body weight (g)b Liver weight (g)b
DMEM medium with 10 % (v/v) fetal calf serum, antibiot-
ics (100 U/mL penicillin A and 100 μg/mL streptomycin), 0 week 6 weeks 6 weeks
and maintained in a 37 °C humidified incubator containing
CD 19.36 ± 0.63 25.18 ± 1.23 1.08 ± 0.17
5 % CO2 for 24 h and divided into a control group (CD) and
HFD 20.17 ± 1.28 29.87 ± 1.46c 1.33 ± 0.22
15 drug treatment groups. Each drug was divided into a low
HFD + COT 20.45 ± 1.19 25.29 ± 1.55d 1.10. ± 0.18
dose group (L), a middle dose group (M) and a high dose
group (H). Drugs were added to culture for 12 h. The final Weight difference between week 0 and week 6
concentrations of COT, QC, and OA were 50, 100, 150 μg/ a
  CD, regular chow diet; HFD, high-fat diet; HFD + COT, high-fat
mL. CL concentrations were 0.1, 0.2, and 0.4 μg/mL, and diet + Celastrus orbiculatus Thunb
b
UA concentrations were 5, 10, and 20 μg/mL. Samples   Data (mean ± SD, n = 12) were expressed as grams
c
containing the proper concentration of DMSO were used   P < 0.05 versus control group
d
as a control. At the end of the cell growing period, total   P < 0.05 versus HFD group

13
Lipids

Fig. 1  Plasma triglyceride (TG), non-high density lipoprotein cho- HDL-C (d) were determined by enzyme methods. The apoA1 (e) was
lesterol (non-HDL-C) and total cholesterol (TC) were decreased, analyzed by Western Blot. Data (mean ± SD, n = 6) are expressed as
high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A1 mg per deciliter. *P < 0.05; **P < 0.01 versus CD group; #P < 0.05;
##
(apoA1) were increased significantly in the Celastrus orbiculatus P < 0.01 versus HFD group
Thunb. (COT) group. Plasma TG (a), non-HDL-C (b), TC (c), and

51.56, and 21.70 %, respectively, in the COT group com-


pared to the HFD group (P < 0.05; P < 0.01) (Fig. 1a–c). In
addition, compared with the HFD group, COT significantly
improved the expression of HDL-C and apoA1 in plasma
by 27.41 and 59.12 % (Fig. 1d, e) (P < 0.05, P < 0.01).

Effects of COT on Lipid Accumulation in Liver

The HE staining experiment showed that the CD group


showed a normal lobular architecture with central veins and
radiating hepatic cords. After a high-fat diet for 6 weeks,
a large amount of lipid droplets appeared in hepatocytes
and denaturation and even necrosis was present in parts
of the hepatocytes in the HFD group. The pathological
changes were significantly attenuated by COT (Fig. 2a–c).
Enzymatic experiments also showed that the levels of TC
and TG in liver decreased by 34.36, 13.24 % (P < 0.01,
P < 0.05) in the COT group compared with the HFD group Fig. 2  COT decreased the TC and TG levels in liver and ameliorated
(Fig. 2d). the pathological changes in C57BL/6J mice fed a high-fat diet. The
histological change in liver was shown by HE staining. a–c Were the
CD group, HFD group, and COT group, respectively. The levels of
Effects of COT on RCT TC and TG (d) were determined by enzymatic methods. Data are pre-
sented as mean ± SD (n = 6). e The levels of TG and TC in different
To evaluate the effects of COT on RCT, a classical isotope- groups of mice liver. **P < 0.01 versus CD group; ##P < 0.01 versus
HFD group
labeled cholesterol tracing assay was employed. Compared
with the HFD group, plasma 3H-labeled cholesterol was
increased in the COT group by 37.53 % (P < 0.01) and suggest that COT enhances cholesterol transport from the
27.07 % (P < 0.01) at 12 and 24 h, respectively, but there macrophages to plasma during the first 24 h. 3H-labeled
was no significant difference at 48 h (Fig. 3a). These results cholesterol was increased by 48.15 % in liver after COT

13
Lipids

Fig. 3  Celastrus orbiculatus Thunb. (COT) promoted reverse counts per minute. At the end of the study (48 h after injection), mice
cholesterol transport in C57BL/6J mice. Mice were treated with were exsanguinated and perfused with cold PBS, and portions of liver
either vehicle or high-fat diet alone or high-fat diet with COT for tissue (b), bile (c) and intestine (e) were removed and flash-frozen
6 weeks. 3H-cholesterol-labeled and ox-LDL-loaded Raw264.7 cells for 3H-tracer analysis by liquid scintillation. Feces were collected
(6.0  × 106 cells containing 6.1 × 105 counts per minute in 0.5 mL continuously at 24 h (d) and 48 h and stored at 4 °C before analysis
MEM per mouse) were injected intraperitoneally. Plasma (a) was of 3H-tracer (mean ± SD, n = 6),*P < 0.05, **P < 0.01 versus CD
collected at 0, 12, 24, and 48 h after injection for liquid scintillation group; #P < 0.05, ##P < 0.01 versus HFD group
counting, and the levels were expressed as the percentage of total

treatment (Fig. 3b). As shown in Fig. 3c, compared with were no marked changes at 24–48 h (P > 0.05), the fecal
the HFD group, 3H-cholesterol accumulation in the bile 3
H-tracer levels in the CD, HFD, and COT group were
was increased by 17.06 % (P < 0.05) after COT treatment 4.21, 4.27, and 4.37 %, respectively. The 3H-tracer level
and the fecal 3H-tracer level was increased significantly by was not significantly changed in the intestine (P > 0.05)
46.15 % in the COT group at 0–24 h (Fig. 3d), but there (Fig. 3e).

13
Lipids

Fig. 4  Celastrus orbiculatus Thunb. (COT) enhanced the mRNA mined by real-time polymerase chain reactions. Data were calculated
expression of scavenger receptor B1 (SRB1) (a), cholesterol 7 alpha- after adjusting for GDAPH using the 2−∆∆Ct method. Expression of
hydroxylase (CYP7A1) (b), liver X receptor (LXRα) (c), low den- mRNA is indicated as fold differences compared with control mice
sity lipoprotein LDL receptor (LDLR) (d), ATP-binding cassette (n = 6). *P < 0.05, versus control group; #P < 0.05, ##P < 0.01 versus
transporters G5 (ABCG5) (e) and G8 (ABCG8) (f) in mouse liver. HFD group. Data are presented as the mean ± SEM of at least three
Effects of COT on mRNA expression of hepatic genes were deter- independent experiments

13
Lipids

Effects of COT on Related RCT Enzymes Compared with the HDL3 group, COT improved the
3
and Transporters H-cholesterol efflux by 21.50 % at the dosage of 150 μg/
mL (P < 0.01). CL exhibited the best effect, followed by
To investigate the potential mechanisms of COT on RCT, UA, OA, and QC. Compared with the HDL3 group, CL
the expression of related enzymes, transporters, and tran- and QC increased 3H-cholesterol significantly by 22.13 and
scription factors in the liver (SRB1, CYP7A1, LXRα, 17.59 %, respectively, at the highest dosage (P < 0.01). OA
LDLR, and ABCG5/g8) were determined by real-time and UA are isomerides and increased the 3H-cholesterol
PCR. As shown in Fig. 4, compared with the HFD group, efflux significantly (P < 0.01) at dosages of 50 and 5 μg/
COT treatment enhanced the mRNA expression of SRB1 mL, respectively. Compared with the HDL3 group, KF
and CYP7A1 by 15.29 and 23.39 %, respectively (P < 0.05) decreased cholesterol efflux (Fig. 7f) in a dose-dependent
(Fig.  4a, b). The mRNA expression of LXRα and LDLR manner and RT promoted cholesterol efflux, but with no
increased by 12.97 and 41.67 %, respectively (P < 0.01) significant difference (Fig. 7e).
(Fig. 4c, d). Moreover, COT treatment enhanced the mRNA
expression of ABCG5 by 17.96 %, while there was no dif- Effects of COT and its Bioactive Ingredients on SRB1,
ference in ABCG8 (Fig. 4e, f). Western blot experiments ABCA1 and ABCG1 Expressions in the Raw 264.7
showed that COT improved the expression of the hepatic Macrophage
gene involved in RCT in the liver. As shown in Fig. 5, there
were no significant differences, such as liver protein levels To further elucidate the effect of COT and its bioactive
of ABCA1 and CYP7A1, between the HFD group and the ingredients on ABCA1, ABCG1, and SRB1, the Raw 264.7
CD group (P > 0.05). Compared with the HFD group, COT macrophage were treated with 50, 100, and 150 μg/mL of
treatment enhanced the protein expression of ABCA1 and COT, QC or OA, 0.1, 0.2, and 0.4 μg/mL of CL, or 5, 10,
CYP7A1 by 123.51 and 192.07 %, respectively (Fig. 5a, and 20 μg/mL of UA. Compared with the control group,
b). Compared with the CD group, liver protein levels of COT and its bioactive ingredients all increased SRB1,
SRB1, LDLR, LXRα, apoA1 were increased significantly ABCA1, and ABCG1 protein levels. Especially, the mid-
in the HFD group (P < 0.01). COT treatment enhanced the dle and high doses of COT improved SRB1, ABCA1, and
protein expression of SRB1, LDLR, LXRα, and apoA1 by ABCG1 protein levels significantly (Fig. 8a, b) (P < 0.01).
42.31, 140.31, 104.02, and 45.65 %, respectively, com- The effects of COT on ABCA1 and SRB1 were similar to
pared to the HFD group (Fig. 5c–f). those in the liver setting (Fig. 5a, c). As shown in Fig. 8c,
d, the effects of CL on SRB1 were significant (P < 0.01);
MTT Experiments both the middle and high doses improved SRB1, ABCG1
and ABCA1. UA improved SRB1, ABCG1, and ABCA1
In general, cell death increases significantly with the drug in a dose-dependent manner and had a significant effect
concentration. This is the first report of COT with RCT (P < 0.01) at the highest dose (Fig. 8e, f). OA also showed
activity, and the effective components are not known. dose-dependent improvement in SRB1, ABCG1, and
Therefore, the previously reported bioactive ingredients of ABCA1, which was significant (P < 0.01) in each dose
COT such as CL, RT, QC, KF, OA, and UA were used to group (Fig. 8g, h). Compared with the control group,
evaluate the RCT effect in vitro. As shown in Fig. 6, there no improvement in ABCG1 expression was observed at
was no significant influence of the compounds on cell death the three doses of QC, but SRB1 expression levels were
under the concentrations screened: COT, QC, RT, OA, and improved significantly at the high dose, and ABCA1
KF (50–150 μg/mL), UA (5–20 μg/mL), and CL (0.1– expression levels were improved significantly at the middle
0.4 μg/mL). Beyond the above range, cell death increased and high doses (P < 0.01), respectively (Fig. 8i, j).
significantly with the drug concentration. The concentra-
tions for the subsequent RCT experiments were partially
based on the MTT results (Fig. 6). Discussion

Effects of COT and its Bioactive Ingredients It is known that the RCT pathway includes a series of
on Cholesterol Efflux in the Raw 264.7 Macrophage steps beginning in peripheral tissues with the efflux of
cholesterol or cholesterol esters (CE) to HDL and ending
To evaluate the effects of COT on RCT in vitro, a classi- in the liver and the gastrointestinal tract with the secre-
cal isotope-labeled cholesterol-tracing assay was employed tion of cholesterol into bile and clearance through feces.
using the Raw 264.7 macrophage. COT and four compo- Among these series of steps, HDL and the related cho-
nents improved 3H-cholesterol efflux in a dose-dependent lesterol transporters play crucial roles. A high level of
manner within the concentrations assayed (Fig. 7a–d, g). HDL-C can reduce the risk of atherosclerosis, and apoA1

13
Lipids

13
Lipids

◂Fig. 5  Celastrus Orbiculatus Thunb. (COT) enhanced the protein increased cholesterol discharge in the liver pathway, but not
expression of ATP-binding cassette transporters A1 (ABCA1) (a), in the intestinal discharge pathway.
cholesterol 7 alpha-hydroxylase (CYP7A1) (b), scavenger receptor
B1 (SRB1) (c), LDL receptor (LDLR) (d), liver X receptor α (LXRα) Accumulating evidences has shown that RCT is a defen-
(e), and apolipoprotein A1 (apoA1) (f) in mouse liver (n  = 6). sive mechanism to attenuate atherosclerotic lesions [4, 16].
**P < 0.01 versus control group; ##P < 0.01 versus HFD group. Data Various enzymes and cholesterol transporters play crucial
are presented as the mean ± SEM of at least three independent exper- roles in the RCT pathway. Researchers have found that
iments
peripheral ABCA1 and ABCG1 are the main transporters
for transferring cholesterol to plasma HDL, and hepatic
is the major protein constituent of HDL. In this study, we SRB1 plays a critical role in cholesterol uptake from
found that COT increased the plasma levels of HDL-C plasma to the liver [17, 18]. SRB1 mediates the exchange of
and apoA1 (Fig. 1d, e) significantly (P < 0.05, P < 0.01). cholesterol between HDL and cells, and is a crucial factor
This indicates that COT can discharge peripheral choles- in the transport of excessive cellular cholesterol from extra-
terol by RCT and alleviate cholesterol accumulation in hepatic tissues to the liver (“reverse cholesterol transport”)
vivo in locations such as the arterial wall. We also found and also is a factor for cholesterol homeostasis. Hepatic
that the levels of TC, TG, and non-HDL-C were increased SRB1 mediates transfer of HDL-cholesterol to the hepato-
significantly in the HFD group (P < 0.01), and they were cytes where cholesterol may be metabolized to bile acids
decreased significantly (P < 0.05, P < 0.01) in the COT [19]. In the liver cells, SRB1 can identify and combine
group compared to the HFD group. A non-HDL-C low- with the apoA1 in HDL-C. SRB1 can transfer CE in the
ering effect and a HDL-C increasing effect of COT on center of HDL to liver cells. Furthermore, plasma HDL-C
guinea pigs previously reported by us [11] were consistent is closely related to ABCA1 and ABCG1 expression [18].
with our findings in C57BL/6J mice in the present study Our study showed that COT increased the expression of
(Fig. 1b, d). SRB1 and apoA1 in the liver (Figs. 4a, 5c, f). This indi-
Our previous studies reported that COT improved the cates that COT promoted RCT by increasing the expression
plasma lipid profile in guinea pigs fed a high-fat diet [11]. of SRB1. COT increased HDL-C concentration in plasma
Whether or not COT is beneficial to RCT in other animal (Fig. 1d) and eliminate plasma 3H-cholesterol more rapidly
models remains unknown. In this study, we selected male (Fig.  3a) than in the HFD group and the CD group. This
C57BL/6J mice as animal models to confirm the effects of is because COT enhanced significantly the HDL level by
COT on promoting RCT. Our results provide evidence for affecting the expressions of ABCA1 and g1 and moderately
the first time that COT reduces lipid accumulation in the improved hepatic SRB1 expression.
liver (Fig. 2) and promotes RCT from intraperitoneal mac- Liver cholesterol could be affected by de novo synthe-
rophages to feces in the murine model (Fig. 3). After intra- sis, and liver cholesterol excretion. The main pathway of
peritoneal injection of 3H-cholesterol-loaded macrophages, liver cholesterol consumption is bile acid synthesis, and
isotopic contents in plasma, liver, bile, feces, and the intes- the primary rate-limiting enzyme of bile acid synthe-
tinal wall were determined. The results showed that COT sis is CYP7A1 [20]. Our data showed COT significantly

Fig. 6  The effects of Celastrus Orbiculatus Thunb. (COT) and its the drug concentration range. The maximum concentrations of COT,
bioactive ingredients on cell viability were detected by MTT meth- QC, RT, OA, and KF to secure cell viability were 150 μg/mL; UA
ods. Exponentially growing Raw 264.7 macrophages were incubated was 20 μg/mL, and CL was 0.4 μg/mL, respectively. OA oleanolic
with active ingredients at various concentrations for 24 h before the acid, UA ursolic acid, CL celastrol, RT rutin, KF kaempferol, and QC
MTT assay was performed. Each bioactive ingredient corresponds quercetin. Data are presented as the mean ± SEM of at least three
with different cell viability; there was no significant difference within independent experiments

13
Lipids

Fig. 7  Effects of Celastrus orbiculatus Thunb. (COT) and its bioac- (n = 6). a CL, b QC, c OA, d UA, e RT, f KF, g COT. **P < 0.01 ver-
tive ingredients on intracellular cholesterol efflux. 3H-cholesterol- sus control; #P < 0.05, ##P < 0.01 versus HDL3 group. Data are pre-
laden macrophages were harvested and counted by liquid scintillation sented as the mean ± SEM of at least three independent experiments

increased its expression (Figs. 4b, 5b). This suggests that The increase of CYP7A1 expression may be one of the rea-
COT might improve cholesterol uptake of liver and utiliza- sons for reduction of liver cholesterol.
tion of cholesterol by bile acid synthesis, thus leading to the LDLR plays a major role in the regulation of plasma
promotion of RCT. Therefore, the positive effect of COT LDL-C by mediating nearly two-thirds of LDL ingres-
on CYP7A1 expression might be the primary reason that sion into the liver for clearance, and its expression in the
3
H-cholesterol accumulates in bile and in feces (Fig. 3c, d). liver is negatively correlated with the hepatic cholesterol

13
Lipids

level [21]. Our study revealed that the level of LDLR was through a p38-dependent pathway in macrophages [30].
increased in the HFD group compared to the CD group Another flavonoid, KF, has been shown to exert several
(Fig.  5d). This is because increased LDLR expression beneficial effects on cardiovascular and nervous systems,
might be a compensatory effect to maintain cholesterol such as the reduction of hydroxyl radicals [31], and the
homeostasis in the liver and intestinal tract [21]. LDLR inflammatory response in macrophage cells [32]. As a fla-
was compensatorily increased to a decrease of LDL level vonoid, QC has a wide range of biological functions associ-
induced by HFD in order to maintain the LDL steady-state ated with the modulation of oxidative stress and inflamma-
in plasma (Fig. 1b). Study also showed that the mRNA tory processes [33–35]. In addition, QC is able to reduce
and protein levels of LDLR in C57BL/6 J mice were up- plasma cholesterol levels in hyperlipidemia animals [36–
regulated (Figs. 4d, 5d) significantly after COT treatment. 38]. In the present study, we found that QC and COT both
These results indicate that COT could accelerate transport promoted 3H-cholesterol efflux from macrophages (Fig. 7b,
of cholesterol from plasma to liver through the up-regula- g).
tion of LDLR expression. CL, an active quinone methide triterpene extracted
Dietary cholesterol absorption and bile acid secretion from the root bark of the Chinese medicine “Thunder of
are important determinants of cholesterol metabolism. God Vine” (tripterygium wilfordii Hook F.), attenuates
ABCG5 and ABCG8 are two ABC half-transporters, which hypertension-induced inflammation and oxidative stress
form a heterodimer in liver and small intestine. ABCG5/ in vascular smooth muscle cells via hemoxygenase-1
ABCG8 heterodimers mediate the efflux of dietary choles- induction [39], inhibits cholesterol ester accumulation
terol out of the intestinal cells, whereas in hepatocytes, they in macrophages, and decreases vascular smooth muscle
mediate the efflux of sterols into the bile [22]. In this study, cell proliferation [40]. In the present study, CL signifi-
COT improved the expression of ABCG5 in liver sig- cantly promoted 3H-cholesterol efflux from macrophages
nificantly (P < 0.05) compared with the CD and the HFD (Fig. 7a), which indicates that CL might promote RCT via
groups (Fig. 4e). However, there was no difference in the mediating cholesterol efflux to play an anti-atherosclerosis
expression of ABCG8 in the liver (Fig. 4f). This indicates role. Hepato-protective and anti-tumoral activities of OA
that ABCG5 plays a role in excretion of sterols into bile. have long been recognized [41], and some evidence indi-
The effects of ABCG8 and ABCG5 that mediate the efflux cates that it has potential antiatherogenic effects. Chronic
of dietary cholesterol out of the intestine remain uncertain administration of OA to Dahl salt-sensitive rats demon-
and will be investigated in our future study. LXR senses strated antihyperlipidemic, antioxidant, and antihyperten-
intracellular cholesterol content and initiates various adap- sive effects [42]. The present study showed that three doses
tive mechanisms, protecting the cell from imbalance of OA promoted 3H-cholesterol efflux significantly (Fig. 7c).
cholesterol homeostasis [23]. Many genes involved in RCT, It indicated that OA could promote cholesterol efflux to
such as ABCA1/g1, SRB1, CYP7A1, and ABCG5/g8, are alleviate cholesterol accumulation in vivo.
regulated by LXR and our data showed that COT promoted The results of this study provide evidence for the first
LXR expression in liver (Fig. 4c). The expression of SRB1, time that COT reduces lipid accumulation and amelio-
LDLR, and CYP7A1 were increased in keeping with LXR rates the pathological changes of a high-fat diet in the liver
in the COT group. This suggests that COT treatment might (Fig. 2) by promoting RCT in the mice and macrophages.
participate in the expression of LXR subunits. Furthermore, some effects of bioactive components on RCT
COT is a medicinal plant widely distributed in China were proved to occur by 3H-cholesterol efflux experiments.
and is mainly composed of total flavonoids and terpe- COT and its bioactive components not only increased the
noids, which are responsible for attenuating atherosclerotic expression of macrophage SRB1, but also ABCG1 and
lesions due to their antioxidant activity. Some flavonoids ABCA1 (Fig. 8), which might play a role, at least in part,
[24–27] and terpenoids [28, 29] are also bioactive com- in promoting the first step of RCT by COT and its bioac-
pounds with potential lipid-lowering effects. In the present tive components. The study indicated that these compounds
study, we determined that the contents of flavonoids and displace apoA-1 from HDL, thus increasing the efficiency
terpenoids were as follows: CL, UA, KF, and OA- 5.13, of ABCA1-mediated cholesterol efflux, compared to intact
0.37, 0.16, and 0.26 mg/g in the root and 1.51, 0.16, 2.80, HDL.
and 0.67 mg/g in stems, respectively. The contents of QC In addition, we found that administration of COT sig-
are 0.017 mg/g in the root and 0.0072 mg/g in stems. The nificantly prevented increases of the body weight and liver
content of RT is 1.80 mg/g in stems. Results indicate that weight gain in a C57BL/6J mice model of hyperlipidemia
flavonoids and terpenoids are abundant in COT, and that (Table  3), which was consistent with our previous report
they may be the effective components of COT on RCT. [11]. The reason may be that the lipid-lowering activity
An in vitro study found that the flavonoid pigment of COT was associated with the up-regulation of genes
QC enhanced ABCA1 expression and cholesterol efflux involved in lipid metabolism in the liver, including LDLR,

13
Lipids

13
Lipids

◂ Fig. 8  Celastrus orbiculatus Thunb. (COT) and its bioactive ingre- and ABCG1, but not SR-BI, promote macrophage reverse cho-
dients stimulated the expression of SRB1, ABCA1, and ABCG1 lesterol transport in vivo. J Clin Invest 117:2216–2244
in Raw264.7 macrophages. Protein levels of ABCA1 and ABCG1 4. Rader DJ, Alexander ET, Weibel GL, Billheimer J, Rothblat
and SRB1 in Raw264.7 macrophages were determined after treat- GH (2009) The role of reverse cholesterol transport in animals
ment with COT, quercetin (QC), oleanolic acid (OA) 0, 50, 100, and and humans and relationship to atherosclerosis. J Lipid Res
150 µg/mL; ursolic acid (UA) 0, 5, 10, and 20 µg/mL; celastrol (CL) 50(Suppl):S189–S194
0, 0.1, 0.2, and 0.4 µg/mL. a, b Effects of COT on ABCA1, ABCG1, 5. Naik SU, Wang X, DaSilva JS, Jaye M, Macphee CH, Reilly MP,
and SRB1 expression. c, d Effects of CL on ABCA1, ABCG1, and Billheimer JT, Rothblat GH, Rader DJ (2006) Pharmacologi-
SRB1 expression. e, f Effects of UA on ABCA1 and ABCG1 and cal activation of liver X receptors promotes reverse cholesterol
SRB1 expression. g, h Effects of OA on ABCA1, ABCG1 and SRB1 transport in vivo. Circulation 113:90–97
expression. i, j Effects of QC on ABCA1, ABCG1, and SRB1 expres- 6. Moon M, Lee MS, Kim CT, Kim Y (2007) Dietary chitosan
sion. CD Control dose group, L low-dose group, M middle-dose enhances hepatic CYP7A1 activity and reduces plasma and liver
group, H high-dose group. **P < 0.01 versus CD group. Data are pre- cholesterol concentrations in diet-induced hypercholesterolemia
sented as the mean ± SEM of six independent experiments in rats. Nutr Res Pract 1:175–179
7. Cavelier C, Lorenzi I, Rohrer L, von Eckardstein A (2006) Lipid
efflux by the ATP-binding cassette transporters ABCA1 and
ABCG1. Biochim Biophys Acta 1761:655–666
CYP7A1, and SRB1, or maybe it is related to the COT-to- 8. Beltowski J, Wojcicka G, Jamroz-wisniewska A (2009) Adverse
gastrointestinal reaction. effects of statins-mechanisma and consequences. Curr Drug Saf
4:209–228
To summarize, the current study revealed that COT
9. Qian YY, Zhang H, Hou Y, Yuan L, Li GQ, Guo SY, Hisamits T,
promotes RCT in C57BL/6J mice by raising HDL-C and Liu YQ (2012) Celastrus orbiculatus extract inhibits tumor angi-
improving the expression of hepatic SRB1, CYP7A1, ogenesis by targeting vascular endothelial growth factor signal-
LXRα, ABCG5, and LDLR. An in vitro study also showed ing pathway and shows potent antitumor activity in hepatocarci-
nomas in vitro and in vivo. Chin J Integr Med 18:752–760
that CL, QC, OA, and UA promoted macrophage cho-
10. Jin HZ, Hwang BY, Kim HS, Lee JH, Kim YH, Lee JJ (2002)
lesterol efflux by increasing the expressions of SRB1, Antiinflammatory constituents of Celastrus orbiculatus inhibit
ABCA1, and ABCG1. This indicates that these substances the NF-kappaB activation and NO production. J Nat Prod
were bioactive components affecting RCT. These find- 65:89–91
11. Zhang Y, Si YH, Yao ST, Yang NN, Song GH, Sang H, Zu DD,
ings suggest that COT is an effective Chinese medicine to
Xu X, Wang J, Qin SC (2013) Celastrus orbiculatus Thunb.
reduce lipid levels and improve RCT and add to the accu- Decreases athero-susceptibility in lipoproteins and the aorta of
mulating evidence for bioactive components of COT in guinea pigs fed high fat diet. Lipids 48:619–631
cholesterol metabolism. To further understand the mecha- 12. Qin L, Qin XP, Wang Z, Zhu BY, Liao DF (2006) Effect of
pravastatin on cholesteryl esters in foam cells and the relation
nisms of COT on RCT, its bioactive components as studied
with caveolin-1. Acta Physiol Sin 58:47–52 (Chinese, English
in vitro in the present study need to be investigated in vivo. abstract)
13. Sherman MP, Aeberhard EE, Wong VZ, Griscavage JM, Ignarro
Acknowledgments  This research was supported by financial dona- LJ (1993) Pyrrolidine dithiocar -bamate inhibits induction of
tions from the Taishan Scholars Foundation of Shandong Province nitric oxide synthase activity in rat alveolar macrophages. Bio-
(200867), University of Science and Technology Plan Projects Fund chem Biophys Res Commun 191:1301–1308
of Shandong Province (J121M01), Taian City Technology Develop- 14. Chapman MJ, Mills GL, Ledford JH (1975) The distribution
ment Plan (201440774-03), Key development projects of Shandong and partial characterization of the serum apolipoproteins in the
province (2015GSF119008), and the Shandong Provincial Natural guinea pig. Biochem J 149:423–436
Science Fund (ZR2013HL063, ZR2013HQ014). 15. Yu Y, Si YH, Song GH, Luo T, Wang JF, Qin SC (2011) Etha-
nolic extract of propolis promotes reverse cholesterol transport
Compliance with Ethical Standards  and the expression of ATP-binding cassette transporter A1 and
G1 in mice. Lipids 46:805–811
Conflict of interest  The authors have declared that no conflict of 16. Zhang Y, Da Silva JR, Reilly M, Billheimer JT, Rothblat GH,
interest exist. Rader DJ (2005) Hepatic expression of scavenger receptor class
B type I (SR-BI) is a positive regulator of macrophage reverse
cholesterol transport in vivo. J Clin Invest 115:2870–2874
17. Van Eck M, Pennings M, Hoekstra M, Out R, Van Berkel TJ
References (2005) Scavenger receptor BI and ATP-binding cassette trans-
porter A1 in reverse cholesterol transport and atherosclerosis.
1. Assmann G, Cullen P, Jossa F, Lewis B, Mancini M (1999) Curr Opin Lipidol 16:307–315
Coronary heart disease: reducing the risk: the scientific back- 18. Yvan-Charvet L, Wang N, Tall AR (2010) Role of HDL,

ground to primary and secondary prevention of coronary heart ABCA1, and ABCG1 transporters in cholesterol efflux and
disease. A worldwide view. International task force for the pre- immune responses. Arterioscler Thromb Vasc Biol 30:139–143
vention of coronary heart disease. Arterioscler Thromb Vasc Biol 19. Trigatti BL, Krieger M, Rigotti A (2003) Influence of the HDL
19:1819–1824 receptor SR-BI on lipoprotein metabolism and atherosclerosis.
2. Lewis GF, Rader DJ (2005) New insights into the regulation of Arterioscler Thromb Vasc Biol 23:1732–1738
HDL metabolism and reverse cholesterol transport. Circ Res 20. Chiang JY (2009) Bile acids: regulation of synthesis. J Lipid Res
96:1221–1232 50:1955–1966
3. Wang X, Collins HL, Ranalletta M, Fuki IV, Billheimer JT, 21. Brown MS, Goldstein JL (1986) A receptor-mediated pathway
Rothblat GH, Tall AR, Rader DJ (2007) Macrophage ABCA1 for cholesterol homeostasis. Science 232:34–47

13
Lipids

22. Schmitz G, Langmann T, Heimerl S (2001) Role of ABCG1 and 33. Bischoff SC (2008) Quercetin: potentials in the prevention and
other ABCG family members in lipid metabolism. J Lipid Res therapy of disease. Curr Opin Clin Nutr Metab Care 11:733–740
42:1513–2018 34. Rivera L, Morón R, Sánchez M, Zarzuelo A, Galisteo M (2008)
23. Malerod L, Juvet L, Hanssen-Bauer A, Eskild W, Berg T (2002) Quercetin ameliorates metabolic syndrome and improves the
Oxysterol-activated LXRalpha/RXR induces SR-BI-promoter inflammatory status in obese Zucker rats. Obesity (Silver Spring)
activity in hepatoma cells and preadipocytes. Biochem Biophys 16:2081–2087
Res Commun 299:916–923 35. Kleemann R, Verschuren L, Morrison M, Zadelaar S, van Erk
24. Bhaskar S, Kumar KS, Krishnan K, Antony H (2013) Querce- MJ, Wielinga PY, Kooistra T (2011) Anti-inflammatory, antipro-
tin alleviates hypercholes-terolemic diet induced inflammation liferative and anti-atherosclerotic effects of quercetin in human
during progression and regression of atherosclerosis in rabbits. in vitro and in vivo models. Atherosclerosis 218:44–52
Nutrition 29:219–229 36. Juźwiak S, Wójcicki J, Mokrzycki K, Marchlewicz M, Białecka
25. Xiao HB, Lu XY, Sun ZL, Zhang HB (2011) Kaempferol
M, Wenda-Rózewicka L, Gawrońska–Szlarz B, Droździk M
regulates OPN-CD44 pathway to inhibit the athero-genesis (2005) Effect of quercetin on experimental hyperlipidemia and
of apolipoprotein E deficient mice. Toxicol Appl Pharmacol atherosclerosis in rabbits. Pharmacol Rep 57:604–609
257:405–411 37. Auger C, Teissedre PL, Gérain P, Lequeux N, Bornet A, Seris-
26. Zhang J, Liang R, Wang L, Yan R, Hou R, Gao S, Yang B (2013) ier S, Besançon P, Caporiccio B, Cristol JP, Rouanet JM (2005)
Effects of an aqueous extract of Crataegus pinnatifida Bge. var. Dietary wine phenolics catechin, quercetin, and resveratrol effi-
major N.E.Br. fruit on experimental atherosclerosis in rats. J Eth- ciently protect hypercholesterolemic hamsters against aortic fatty
nopharmacol 48:63–69 streak accumulation. J Agric Food Chem 53:2015–2021
27. Boots AW, Haenen GR, Bast A (2008) Health effects of
38. Chirumbolo S (2010) The role of quercetin, flavonols and fla-
quercetin: from antioxidant to nutraceutical. Eur J Pharmacol vones in modulating inflammatory cell function. Inflamm
585:325–337 Allergy Drug Targets 9:263–285
28. Sang WK, Min SK, Hyun SK, Yunho K, Daekeun S, Jung Han 39. Li H, Wang QJ, Zhu DN, Yang Y (2008) Reinioside C, a triter-
YP, Young HK (2013) Celastrol attenuates adipokine resistin- pene saponin of Polygala aureocauda Dunn, exerts hypolipi-
associated matrix interaction and migration of vascular smooth demic effect on hyperlipidemic mice. Phytother Res 22:159–164
muscle cells. J Cell Biochem 114:398–408 40. Kannaiyan R, Shanmugam MK, Sethi G (2011) Molecular tar-
29. Buus NH, Hansson NC, Rodriguez-Rodriguez R, Stankevicius gets of celastrol derived from Thunder of God Vine: potential
E, Andersen MR, Simonsen U (2011) Antiatherogenic effects of role in the treatment of inflammatory disorders and cancer. Can-
oleanolic acid in apolipoprotein E knockout mice. Eur J Pharma- cer Lett 303:9–20
col 670:519–526 41. Somova LO, Nadar A, Rammanan P, Shode FO (2003) Cardio-
30. Chang YC, Lee TS, Chiang AN (2012) Quercetin enhances
vascular, antihyperlipidemic and antioxidant effects of oleanolic
ABCA1 expression and cholesterol efflux through a p38-depend- and ursolic acids in experimental hypertension. Phytomedicine
ent pathway in macrophages. J Lipid Res 53:1840–1850 10:115–121
31. Park MY, Ji GE, Sung MK (2012) Dietary kaempferol sup-
42. Liu J (1995) Pharmacology of oleanolic acid and ursolic acid. J
presses inflammation of dextran sulfate sodium-induced colitis Ethnopharmacol 49:57–68
in mice. Dig Dis Sci 57:355–363
32. Calderón-Montaño JM, Burgos-Morón E, Pérez-Guerrero C,

López-Lázaro M (2011) A review on the dietary flavonoid
kaempferol. Mini Rev Med Chem 11:298–344

13

You might also like