Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Clinica Chimica Acta 424 (2013) 245–252

Contents lists available at ScienceDirect

Clinica Chimica Acta


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

Invited critical review

Foam cells in atherosclerosis


Xiao-Hua Yu a, Yu-Chang Fu c, Da-Wei Zhang d, Kai Yin b,⁎, Chao-Ke Tang a,b,⁎⁎
a
Life Science Research Center, University of South China, Hengyang, Hunan 421001, China
b
Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang, Hunan 421001, China
c
Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294-0012, USA
d
Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta T6G 2S2, Canada

a r t i c l e i n f o a b s t r a c t

Article history: Atherosclerosis is a chronic disease characterized by the deposition of excessive cholesterol in the arterial intima.
Received 25 April 2013 Macrophage foam cells play a critical role in the occurrence and development of atherosclerosis. The generation of
Received in revised form 4 June 2013 these cells is associated with imbalance of cholesterol influx, esterification and efflux. CD36 and scavenger receptor
Accepted 6 June 2013
class A (SR-A) are mainly responsible for uptake of lipoprotein-derived cholesterol by macrophages. Acyl coen-
Available online 16 June 2013
zyme A:cholesterol acyltransferase-1 (ACAT1) and neutral cholesteryl ester hydrolase (nCEH) regulate cholesterol
Keywords:
esterification. ATP-binding cassette transporters A1(ABCA1), ABCG1 and scavenger receptor BI (SR-BI) play crucial
Foam cells roles in macrophage cholesterol export. When inflow and esterification of cholesterol increase and/or its outflow
Atherosclerosis decrease, the macrophages are ultimately transformed into lipid-laden foam cells, the prototypical cells in the
CD36 atherosclerotic plaque. The aim of this review is to describe what is known about the mechanisms of cholesterol
ACAT1 uptake, esterification and release in macrophages. An increased understanding of the process of macrophage
ABCA1 foam cell formation will help to develop novel therapeutic interventions for atherosclerosis.
ABCG1 © 2013 The Authors. Published by Elsevier B.V. Open access under CC BY-NC-ND license.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
2. Cholesterol uptake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
2.1. CD36 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 246
2.2. SR-A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
3. Cholesterol esterification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
3.1. ACAT1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
3.2. nCEH . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 247
4. Cholesterol efflux . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248
4.1. ABCA1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 248
4.2. ABCG1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
4.3. SR-BI . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 249
5. Conclusion and future directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
Disclosure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 250
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 251

Abbreviations: ABCA1, ATP-binding cassette transporter A1; ABCG1, ATP-binding cassette transporter G1; SR-BI, scavenger receptor BI; SR-A, scavenger receptor class A; ACAT1, acyl
coenzyme A:cholesterol acyltransferase-1; nCEH, neutral cholesteryl ester hydrolase; ox-LDL, oxidized low-density lipoprotein; HDL, high-density lipoprotein; apoA-I, apolipoprotein A-I;
LDLR, low-density lipoprotein receptor; apoE, apolipoprotein E; CE, cholesterol ester; FC, free cholesterol; LXR, liver X receptor; RXR, retinoid X receptor; PPAR-γ, peroxisome
proliferator-activated receptor-γ; ERK1/2, extracellular signal-regulated kinases 1 and 2; PPREs, PPAR response elements; PKB, protein kinase B; PKC, protein kinase C; TGF-β,
transforming growth factor-β; MAPK, mitogen-activated protein kinase; RCT, reverse cholesterol transport; PI3K, phosphatidylinositol 3-kinase; AGE, advanced glycation end products.

⁎ Corresponding author. Tel./fax: +86 734 8281852.


⁎⁎ Correspondence to: Institute of Cardiovascular Research, University of South China, Hengyang, Hunan 421001, China. Tel./fax: +86 734 8281853.
E-mail addresses: kaiyinby@yahoo.com.cn (K. Yin), tchaoke@yahoo.com.cn (C.-K. Tang).

0009-8981 © 2013 The Authors. Published by Elsevier B.V. Open access under CC BY-NC-ND license.
http://dx.doi.org/10.1016/j.cca.2013.06.006
246 X.-H. Yu et al. / Clinica Chimica Acta 424 (2013) 245–252

1. Introduction as a high-affinity receptor for ox-LDL. A domain located between


amino acids 155 and 183 of CD36 involves in ox-LDL binding. Other
Atherosclerotic diseases are the major causes of mortality and ox-LDL binding sites have also been reported such as amino acids
morbidity in the world. Formation of macrophage foam cells in the 28–93 and possibly 120–155. Binding of ox-LDL to CD36 leads to
intima is a major hallmark of early stage atherosclerotic lesions. endocytosis through a lipid raft pathway that is distinct from the
Uncontrolled uptake of oxidized low-density lipoprotein (ox-LDL), ex- clathrin-mediated or caveolin internalization pathways. The patho-
cessive cholesterol esterification and/or impaired cholesterol release genic role of ox-LDL in atherosclerosis largely depends on CD36. A
result in accumulation of cholesterol ester (CE) stored as cytoplasmic recent study revealed that plasma soluble CD36 correlates significant-
lipid droplets and subsequently trigger the formation of foam cells. ly with markers of atherosclerosis, insulin resistance and fatty liver in
Scavenger receptors (SRs), CD36 and SR class A (SR-A) are the princi- a nondiabetic healthy population [5]. Patients with acute coronary
pal receptors responsible for the binding and uptake of ox-LDL in mac- syndromes exhibit a significant increase in CD36 expression in circu-
rophages [1]. Acyl coenzyme A:cholesterol acyltransferase-1 (ACAT1) lating monocytes, which can be inhibited by a 6-month treatment of
and neutral cholesteryl ester hydrolase (nCEH) play a critical role in atorvastatin [6]. Small molecules with anti-CD36 activity have been
cholesterol esterification [2]. ATP-binding cassette (ABC) transporter shown to decrease postprandial hyperlipidemia and protect against
A1(ABCA1), ABCG1 and scavenger receptor-BI (SR-BI) mediate cho- atherosclerosis [7]. In addition, the 573A allele of CD36 has a protec-
lesterol export from macrophages [3]. This review focuses on the tive effect against atherosclerosis development while the 591T allele
recent developments in our knowledge of the roles and regulation of is a cardiovascular risk factor [8]. On the other hand, Moore and col-
these receptors, enzymes and transporters in the formation of macro- leagues reported that apoE−/− mice lacking CD36 or SR-A display
phage foam cells. increased aortic sinus atherosclerotic lesion area and abundant mac-
rophage foam cells in the aortic intima despite reductions in peritone-
2. Cholesterol uptake al macrophage CE accumulation in vivo [9]. Moreover, clinical studies
show that patients with CD36 deficiency are associated with severe
Cholesterol uptake is a pathway by which extracellular modified and enhanced atherosclerotic diseases [10]. Therefore, the role of
LDL are ingested by macrophages via receptors-mediated phagocyto- CD36 as a proatherogenic mediator of ox-LDL uptake in vivo needs
sis and pinocytosis. SRs such as SR-A and CD36 have been implicated to be reassessed.
in this process. In vitro studies have shown that CD36 and SR-A ac- CD36 is highly expressed in macrophages and its expression is regu-
count for 75% to 90% of ox-LDL internalization by macrophages, lated by multiple factors (Fig. 1). Recently, Inoue et al. reported that
whereas other SRs cannot compensate for their absence [4]. Thus, astaxanthin (ASX), an oxygenated carotenoid (xanthophyll), signifi-
CD36 and SR-A are the most important receptors responsible for the cantly increases CD36 levels in peritoneal macrophages by stimulating
uptake of modified lipoproteins by macrophages. peroxisome proliferator-activated receptor-γ (PPARγ) [11]. On the
other hand, curcumin, a potent antioxidant extracted from Curcuma
2.1. CD36 longa, induces a PPARγ-independent CD36 overexpression through
upregulating nuclear erythroid-related factor 2(Nrf2) [12]. Additionally,
CD36 is first identified as the platelet glycoprotein III b/IV, an Gao et al. reported that palmitate increases CD36 expression in mono-
88 kDa heavily glycosylated transmembrane protein that belongs to cytes through the regulation of de novo ceramide synthesis [13]. Sup-
SR class B family. It consists of an extracellular domain flanked by plementation of the mushroom Agaricus blazei for 12 weeks markedly
two transmembrane and two cytoplasmic domains. CD36 functions elevates CD36 expression and plaque vulnerability in apoE−/− mice,

Fig. 1. Regulation of CD36 and SR-A expression in macrophages. CD36 expression is induced by ASX and inhibited by TSIIA and quercitrin via PPARγ pathway. Palmitate increases
CD36 levels while kaempferol, EM-1, squalene and walnut reduce its levels. Curcumin also upregulates CD36 expression through promoting Nrf-2 nuclear translocation but
decreases SR-A protein expression via UPS. Berberine, Kv1.3 and TNF-α enhance SR-A levels, whereas MLPE and H2S diminish its levels. RXR: retinoid X receptor.
X.-H. Yu et al. / Clinica Chimica Acta 424 (2013) 245–252 247

indicating a proatherogenic property of mushroom A. blazei [14]. In 3. Cholesterol esterification


contrast, a dietary compound quercitrin inhibits CD36 expression in
murine macrophages through interfering with PKC/PPARγ signaling In addition to cholesterol uptake, the balance of free cholesterol
cascades [15]. Tanshinone IIA (TSIIA), a lipophilic diterpene isolated (FC) and CE is also critical for the regulation of intracellular cholester-
from Salvia miltiorrhiza Bunge (Danshen), also decreases CD36 levels ol content in macrophage foam cells. After internalization, lipopro-
in ox-LDL treated macrophages by antagonism of PPARγ [16]. Similarly, teins are delivered to the late endosome/lysosome, where CE is
endomorphin-1(EM-1), a member of the endogenous opioid peptides hydrolyzed into FC by lysosomal acid lipase (LAL). To prevent the
family, and squalene, one of the components of olive oil, downregulate FC-associated cell toxicity, the FC released is re-esterified on the en-
CD36 expression and prevent lipid accumulation in human lipid-laden doplasmic reticulum (ER) by ACAT1 and stored in cytoplasmic lipid
macrophages [17,18]. Treatment of macrophages with kaempferol droplets. If this persistently occurs, excessive CE will accumulate in
inhibits c-Jun/activator protein-1 (AP-1) nuclear translocation and macrophages, thereby resulting in the formation of foam cells.
leads to the downregulation of CD36 expression [19]. Atheroprotective These cells are often referred to as foam cells because of their charac-
effect of dietary intake of walnut that enriches in n-3 polyunsaturated teristic “foamy” appearance. The resulting CE are hydrolyzed by nCEH
fatty acids and antioxidant compounds, is associated with reduced to release FC for transporters-mediated efflux, which is increasingly
CD36 expression in apoE−/− mice [20]. In addition, the combined treat- being recognized as the rate-limiting step in FC outflow [31]. Thus,
ment of interferon γ and tumor necrosis factor α (TNF-α) significantly the cycle of esterification and hydrolysis of CE is one of the key
reduces CD36 levels [21]. Thus, these factors may act as novel modula- steps in maintaining intracellular cholesterol homeostasis, and
tors for macrophage-to-foam cell transformation. ACAT1 and nCEH play a crucial role in the process.

3.1. ACAT1
2.2. SR-A
ACAT1 is an integral membrane protein that converts FC into the
SR-A, a 77-kDa cell surface glycoprotein, is a member of class A SR storage form of CE. It has already been reported that ACAT1 deficiency
family. The human and murine SR-A genes are located on chromo- increases the synthesis and efflux of FC in mouse peritoneal macro-
some 8 and can be transcribed to produce three (SR-AI/II/III) and phages while its overexpression promotes CE accumulation and
two SR-A splice variants, respectively. SR-A is highly expressed in macrophage-derived foam cell formation [32,33]. However, the role
macrophages and mediates the uptake of ox-LDL into macrophages. of ACAT1 in atherosclerosis is currently under debate. Kusunoki et
Inhibition of SR-A in macrophages significantly ameliorates foam al. revealed that ACAT1 inhibition attenuates atherosclerosis in apoE−/−
cell formation and atherosclerosis in apoE−/− mice [22]. Silencing mice [34]. On the other hand, mice lacking macrophage-derived
of SR-A or CD36 alone reduces atherogenesis in LDL receptor ACAT1 show accelerated atherosclerosis [35,36]. This may be due to
(LDLR)-deficient apolipoprotein B100 mice, whereas simultaneous the cytotoxic effects of increased FC levels, which crystallize in mac-
silencing of both receptors has no beneficial effect, suggesting that rophage foam cells. The formation of cholesterol crystals not only
the compensatory upregulation of the two receptors is sufficient for destroys cholesterol metabolism during the development of athero-
the uptake of modified LDL [23]. Conversely, completely knockout of sclerosis, but also facilitates the secretion of inflammatory mediators
SR-A alone aggravates atherosclerosis despite of reduced peritoneal such as IL-1β and IL-18 [37]. Taken together, the effects of ACAT1 on
macrophage lipid accumulation [9], but combined deletion of SR-A atherosclerosis remain to be further investigated.
and CD36 reduces atherosclerotic lesion complexity without affecting Numerous steps have been implicated in the modulation of ACAT1
foam cell formation in hyperlipidemic mice, indicating that specific expression (Fig. 2). Insulin has been shown to enhance ACAT1 expres-
inhibition of these pathways in vivo may promote plaque stability sion in THP-1 macrophages via MAP kinases (MAPK)/CCAAT/enhanc-
[24]. Thus, further studies are necessary to clarify the role of SR-A in er binding protein α(C/EBPα) signaling pathway [38]. Yang et al.
atherosclerosis. observed that voltage-gated potassium channel Kv1.3 significantly
The expression level of SR-A is upregulated by a variety of agents upregulates ACAT1 expression and increases percentage of CE in
(Fig. 1). Treatment of ox-LDL stimulates SR-A expression and promotes human monocyte-derived macrophages exposed to ox-LDL [27].
the uptake of ox-LDL into macrophages. Li and colleagues reported that Chlamydia pneumoniae (C. pneumoniae) infection is reported to
berberine significantly elevates mRNA and protein levels of SR-A in increase ACAT1 expression and disturb cholesterol homeostasis
mouse RAW264.7 cells through inhibiting PTEN expression and sus- through c-Jun NH(2) terminal kinase (JNK)/PPARγ dependent signal
taining Akt activation [25]. Proinflammatory cytokines such as TNF-α transduction pathways [39]. Leptin, an adipose tissue-derived hor-
also induce SR-A expression but via suppression of nuclear factor-κB mone, accelerates CE accumulation in human monocyte-derived mac-
(NF-κB) signaling [26]. More recently, voltage-gated potassium rophages by increasing ACAT1 expression via Janus-activated kinase 2
channel Kv1.3 has been shown to upregulate SR-A expression in (JAK2) and PI3K [40]. In contrast, treatment of cultured THP-1 macro-
human monocyte-derived macrophages, revealing that specific phages in vitro with an angiotensin receptor blocker, candesartan,
Kv1.3 blockade may represent a novel strategy to modulate choles- leads to a decrease in ACAT1 expression [41]. Incretins such as
terol metabolism in macrophages [27]. Taken together, these find- glucagon-like peptide-1 (GLP-1) and glucose-dependent insulino-
ings indicate that inhibition of these agents may reduce the uptake tropic polypeptide (GIP) are also able to decrease the levels of
of ox-LDL by macrophages and then prevent the formation of foam ACAT1 by cAMP activation [42]. Our recent studies showed that H2S
cells. inhibits macrophage foam cell formation by suppressing ACAT1 ex-
Hydrogen sulfide (H2S), curcumin and mulberry leaf polyphenolic pression via K(ATP)/ERK1/2 pathway [28]. Additionally, ghrelin, an
extracts (MLPE) have been reported to decrease SR-A expression endogenous ligand of the growth hormone secretagogue receptor
(Fig. 1). Our previous studies have shown that H2S reduces SR-A ex- (GHS-R), lowers the expression of ACAT1 in THP-1 macrophages via
pression and foam cell formation in human monocyte-derived macro- pathways involving GHS-R and PPARγ, indicating a protective role
phages via the K(ATP)/ERK1/2 pathway [28]. Curcumin decreases for ghrelin in controlling cellular lipid accumulation [43].
SR-A protein levels in macrophages through the ubiquitin/proteasome
system(UPS)-dependent proteolysis [29]. On the other hand, MLPE 3.2. nCEH
inhibits PPARγ and then reduces macrophage SR-A levels, suggesting
a protective role of MLPE in regulating intracellular lipid homeostasis The esterification of FC into CE by ACAT1 is opposed by nCEH,
within the intima [30]. which hydrolyzes CE to cholesterol for efflux out of the cells. nCEH
248 X.-H. Yu et al. / Clinica Chimica Acta 424 (2013) 245–252

Fig. 2. Regulation of ACAT1 and nCEH expression in macrophages. ACAT1 is upregulated by insulin, Kv1.3, C. pneumoniae and leptin but downregulated by candesartan, GLP-1, GIP,
H2S and ghrelin. PUFA increases nCEH expression while insulin and IL-33 decrease its expression.

is a single-membrane-spanning type II membrane protein including apolipoprotein A-I (apoA-I). Active transport via transporters includ-
three domains: N-terminal, catalytic, and lipid-binding domains. ing the best characterized ABCA1, ABCG1 and SR-BI is mainly respon-
The N-terminal domain acts as a type II signal anchor sequence to sible for the bulk efflux of cholesterol from macrophages onto
recruit nCEH to the ER with its catalytic domain within the lumen extracellular acceptors.
[44]. N-linked glycosylation at Asn270 is important for its enzymatic
activity. One recent study by Igarashi et al. showed that overex- 4.1. ABCA1
pression of human nCEH promotes the hydrolysis of CE and thereby
stimulates cholesterol mobilization from THP-1 macrophages while ABCA1 is a member of the large superfamily of ABC transporters. It is
knockdown of nCEH markedly accelerates the formation of foam now well established that ABCA1 plays a critical role in the prevention
cells, further conforming the key role for nCEH in maintaining intra- of macrophage foam cell formation and atherosclerosis by mediating
cellular cholesterol equilibrium [45]. Moreover, genetic ablation of the active transport of intracellular cholesterol and phospholipids to
nCEH also facilitates the development of atherosclerosis in apoE−/− apoA-I, the major lipoprotein in HDL. Bochem and colleagues have dem-
mice without affecting their serum lipid profile [46]. Conversely, onstrated that ABCA1 mutation carriers show lower HDL-cholesterol
macrophage-specific overexpression of nCEH leads to a significant (HDL-C) levels and a larger atherosclerotic burden compared with con-
reduction in atherosclerotic lesion area in LDLR−/− mice because of trols [52]. Unexpectedly, mice lacking ABCA1 and SR-BI display severe
enhanced FC efflux and reverse cholesterol transport (RCT) [47]. hypocholesterolemia and foam cell accumulation, but have no athero-
Overall, these results demonstrate the antiatherogenic role of nCEH sclerosis, which is mainly due to the absence of proatherogenic lipopro-
and indicate that this enzyme is a promising target for the treatment teins [53]. ABCA1 overexpression in the liver of LDLR−/− mice results in
of atherosclerosis. accumulation of proatherogenic lipoproteins and enhanced atheroscle-
nCEH is robustly expressed in macrophages as well as in athero- rosis [54]. Thus, in regard to these contradictory observations, a careful
sclerotic lesions. Polyunsaturated fatty acid (PUFA), insulin and re-evaluation of ABCA1 as a potent antiatherogenic agent is necessary.
IL-33 have been described to modulate its expression (Fig. 2). The expression of ABCA1 is highly regulated by multiple processes
Napolitano and colleagues have reported that PUFA derived from (Fig. 3). Lee et al. observed that quercetin stimulates PPARγ/liver X
corn oil can induce nCEH expression in J774 macrophages [48]. On receptor α (LXRα) signaling and then increases ABCA1 expression
the other hand, insulin significantly suppresses the expression of and cholesterol efflux from THP-1 macrophages, indicating that in-
nCEH in THP-1 macrophages, partially accounting for the potential gestion of quercetin or quercetin-rich foods may be an effective way
molecular mechanism of atherogenesis in type 2 diabetes with to lower the risks of atherosclerosis [55]. Studies from our laboratory
hyperinsulinemia [49]. IL-33, a recently discovered member of the showed that apelin-13, a recently discovered adipocytokine, en-
IL-1 cytokine family, also reduces nCEH mRNA levels in primary hances ABCA1 expression in THP-1 macrophage-derived foam cells
human monocyte-derived macrophages by activating ST-2/NF-κB sig- via activating the PKCα pathway and inhibiting calpain activity [56].
naling pathway [50,51]. Variety of other factors that can regulate Toll-like receptor 2 (TLR2) agonist Pam(3)CSK(4) raises ABCA1 levels
nCEH expression will be probably found in later researches. in RAW 264.7 macrophages via activation of PKC-η/phospholipase D2
(PLD2) signaling pathway [57]. S-allylcysteine, the most abundant
4. Cholesterol efflux organosulfur compound in aged garlic extract, also elevates ABCA1
content in human THP-1 macrophages [58]. On the other hand, unsat-
The first step of RCT is cholesterol efflux, namely, accumulated urated fatty acids suppress ABCA1 expression in RAW 264.7 macro-
cholesterol is removed from macrophages in the subintima of the ves- phages by two distinct mechanisms: LXR-dependent transcriptional
sel wall through transporters or other mechanisms such as passive repression possibly through modulating histone acetylation states,
diffusion, and then collected by high-density lipoprotein (HDL) or and LXR-independent posttranslational inhibition [59]. Our previous
X.-H. Yu et al. / Clinica Chimica Acta 424 (2013) 245–252 249

Fig. 3. Regulation of ABCA1, ABCG1 and SR-BI expression in macrophages. ABCA1 expression is induced by apelin-13, Pam(3)CSK(4) and quercetin but inhibited by miR-26 and
IL-18 plus IL-12. Cineole, EVOO and fucosterol upregulate ABCG1 expression by activating LXR, whereas LPS downregulates its expression. In addition, PCA increases ABCG1 levels
via suppression of miR-10b. Res, LA, hydrogen and coffee lead to a significant elevation in the SR-BI levels while PAPP-A treatment reduces its levels. P: phosphorylation, APJ:
G-protein coupled receptor.

studies indicate that IL-18 and IL-12 synergistically decrease ABCA1 [69]. Protocatechuic acid (PCA), a gut microbiota metabolite of
levels in THP-1 macrophage-derived foam cells through the IL-18 re- cyanidin-3 to 0-β-glucoside, exerts an antiatherogenic effect partially
ceptor (IL-18R)/NF-κB/zinc finger protein 202 (ZNF202) signaling through inhibition of miR-10b-mediated downregulation of ABCG1 ex-
pathway [60]. In addition, miR-26 prevents ABCA1 expression and pression, indicating that gut microbiota is a potential novel target for pre-
subsequent cholesterol efflux from macrophages to apoA-I via vention and treatment of atherosclerosis [70]. Conversely, subclinical
targeting LXRα, indicating a novel regulatory role of miR-26 in cellu- low-dose lipopolysaccharide (LPS) potently reduces the expression of
lar lipid accumulation within the intima [61]. Thus, miR-26 inhibitors ABCG1 in bone marrow-derived macrophages through interleukin-1
can be potentially used to treat atherosclerosis. receptor-associated kinase 1(IRAK-1)/glycogen synthase kinase 3β
(GSK3β)/retinoic acid receptor α (RARα) signaling pathway, revealing
a novel intracellular network regulated by low-dose endotoxemia [71].
4.2. ABCG1

ABCG1 mediates cholesterol removal from macrophages to HDL 4.3. SR-BI


particles but not to lipid-free apoA-I. It has been shown that a func-
tional genetic variant in the ABCG1 promoter is associated with an SR-BI promotes cholesterol efflux from macrophages to HDL. A
increased risk of myocardial infarction and ischemic heart disease in genetic variant of the SR-BI in humans leads to a reduction in choles-
the general population [62]. Lack of macrophage ABCG1 has been terol efflux from macrophages but has no significant increase in ath-
reported to cause a modest increase in atherosclerotic lesions [63]. erosclerosis [72]. On the other hand, inactivation of macrophage
However, two other independent groups reported that LDLR−/− SR-BI facilitates atherosclerotic lesion development in apoE−/− mice
mice lacking macrophage ABCG1 show decreased atherosclerotic [73]. However, a recent report suggests that the presence of macro-
lesions [64,65]. Most recently, Meurs and colleagues found that the phage SR-BI inhibits advanced atherosclerotic lesion but promotes
absence of ABCG1 leads to increased lesions in early stage of athero- early lesion development in LDLR−/− mice, indicating a unique dual
sclerosis but causes retarded lesion progression in more advanced role for macrophage SR-BI in the pathogenesis of atherosclerosis
stage of atherosclerosis in LDLR−/− mice, suggesting that the influ- [74]. SR-BI is also highly expressed in the liver. Hepatic SR-BI expres-
ence of ABCG1 deficiency on lesion development depends on the sion is a positive regulator of the rate of RCT from macrophages to the
stage of atherogenesis [66]. Therefore, the impact of ABCG1 on the liver, bile, and feces. Of note, inhibition of hepatic SR-BI using RNA in-
development of atherosclerosis is complex and needs to be further terference technique reduces atherosclerosis in rabbits fed with
investigated. cholesterol-rich diet, indicating that the effect of SR-BI on atherogen-
The regulation of ABCG1 expression has similarities with that of esis in rabbits is different from the one seen in rodents [75]. There-
ABCA1, consistent with a shared role in cholesterol export (Fig. 3). fore, more studies are needed to elucidate the role of SR-BI in
Cineole, a small aroma compound in teas and herbs, induces ABCG1 ex- regulating atherosclerosis.
pression in macrophages through the activation of LXR [67]. Fucosterol, Multiple factors regulate SR-BI expression (Fig. 3). Resveratrol
a sterol that is abundant in marine algae, is also able to increase ABCG1 (Res), a bioactive molecule used in dietary supplements, and
levels in a LXR-dependent mechanism [68]. Consumption of extra- 13-hydroxy linoleic acid (LA), a natural PPAR agonist, induce SR-BI
virgin olive oil (EVOO) for 12 weeks induces a concentration- expression in macrophages through PPARγ/LXRα pathway [76,77].
dependent upregulation of ABCG1 expression in human macrophages Uto-Kondo et al. recently reported that coffee intake significantly
250 X.-H. Yu et al. / Clinica Chimica Acta 424 (2013) 245–252

increases SR-BI expression and HDL-mediated cholesterol efflux from have been recently reported to attenuate the formation of macro-
macrophages via its plasma phenolic acids [78]. Hydrogen administra- phage foam cells through downregulation of CD36 expression and
tion also raises macrophage SR-BI levels in apoE−/− mice [79]. On the upregulation of ABCA1 expression [83]. Notably, despite strong
other hand, pregnancy-associated plasma protein-A (PAPP-A), a inverse association of plasma HDL levels with the risks of atheroscle-
newly recognized metalloproteinase in the insulin-like growth factor rotic cardiovascular diseases, HDL-raising therapies are not always
(IGF) system, significantly decreases SR-BI expression and promotes effective. A randomized and placebo-controlled intervention trial has
the formation of macrophage foam cells via IGF-1/PI3K/Akt/LXRα demonstrated that niacin increases HDL-C but does not reduce cardio-
signaling pathway, indicating a novel mechanism for its proatherogenic vascular events [84]. Moreover, a large human genetics study shows
effect [80]. that genetic variants that enhance HDL-C levels do not necessarily as-
sociate with myocardial infarction risk [85]. As a result, additional
studies are needed to evaluate the role of new compounds to raise
5. Conclusion and future directions HDL-C levels or modify HDL composition and functionality. In addition
to these receptors, enzymes and transporters, we should pay attention
The balanced of cholesterol influx, esterification and release is nec- to the role of other molecules in cholesterol trafficking. In summary,
essary to avoid lipid overload within macrophages, and ultimately, further work is required to elucidate the diverse processes that regu-
atheroma development. Under atherogenic conditions, efflux of cho- late the expression and activity of these proteins, and to determine
lesterol is decreased due to reduced expression of ABCA1, ABCG1 their contribution to disease protection in humans. These studies
and SR-BI; however its uptake is increased due to enhanced expres- will provide more insight into their physiological roles and will reveal
sion of CD36 and SR-A as well as excess esterification of cholesterol oc- novel therapeutic strategies for treating atherosclerotic cardiovascular
curs because of higher ACAT1 level and lower nCEH level. This leads to disease.
excessive CE accumulation as lipid droplets in macrophages, thereby
contributing to the formation of foam cells (Fig. 4). Therefore,
targeting these three pathways could be a viable approach to regulate Disclosure
cholesterol metabolism in macrophages and treat atherosclerotic car-
diovascular diseases. K-604 and rimonabant, recently identified as po- The authors have declared no conflict of interest.
tent inhibitors of ACAT1, have been shown to inhibit macrophage
foam cell formation and retard atherosclerosis in apoE−/− mice Acknowledgment
[81,82]. A growing body of evidence indicates that food components
play an important role in prevention of foam cell formation by reduc- The authors gratefully acknowledge the financial support from
ing cholesterol uptake and/or promoting its removal. For instance, the National Natural Sciences Foundation of China (81070220 and
seven phenolic acids, the major bioactive compounds in blueberries, 81170278), and Aid Program for Science and Technology Innovative

Fig. 4. Schematic representation of the mechanism involved in macrophage foam cell formation. Under atherogenic conditions, the increased uptake and esterification of cholesterol
and/or reduced cholesterol efflux lead to excessive CE accumulation in the form of lipid droplets in the cytoplasm, thereby promoting the formation of macrophage foam cells. (+):
activation, (–): inhibition.
X.-H. Yu et al. / Clinica Chimica Acta 424 (2013) 245–252 251

Research Team in Higher Educational Institutions of Human Province, [27] Yang Y, Wang YF, Yang XF, et al. Specific Kv1.3 blockade modulates key cholesterol-
metabolism-associated molecules in human macrophages exposed to ox-LDL. J Lipid
China (2008-244). Res 2013;54:34–43.
[28] Zhao ZZ, Wang Z, Li GH, et al. Hydrogen sulfide inhibits macrophage-derived foam
cell formation. Exp Biol Med (Maywood) 2011;236:169–76.
References [29] Zhao JF, Ching LC, Huang YC, et al. Molecular mechanism of curcumin on the
suppression of cholesterol accumulation in macrophage foam cells and athero-
[1] Collot-Teixeira S, Martin J, McDermott-Roe C, Poston R, McGregor JL. CD36 and sclerosis. Mol Nutr Food Res 2012;56:691–701.
macrophages in atherosclerosis. Cardiovasc Res 2007;75:468–77. [30] Yang MY, Huang CN, Chan KC, Yang YS, Peng CH, Wang CJ. Mulberry leaf polyphe-
[2] Ghosh S, Zhao B, Bie J, Song J. Macrophage cholesteryl ester mobilization and nols possess antiatherogenesis effect via inhibiting LDL oxidation and foam cell
atherosclerosis. Vascul Pharmacol 2010;52:1–10. formation. J Agric Food Chem 2011;59:1985–95.
[3] Jessup W, Gelissen IC, Gaus K, Kritharides L. Roles of ATP binding cassette trans- [31] Okazaki H, Igarashi M, Nishi M, et al. Identification of neutral cholesterol ester
porters A1 and G1, scavenger receptor BI and membrane lipid domains in choles- hydrolase, a key enzyme removing cholesterol from macrophages. J Biol Chem
terol export from macrophages. Curr Opin Lipidol 2006;17:247–57. 2008;283:33357–64.
[4] Kunjathoor VV, Febbraio M, Podrez EA, et al. Scavenger receptors class A-I/II and [32] Dove DE, Su YR, Swift LL, Linton MF, Fazio S. ACAT1 deficiency increases cholester-
CD36 are the principal receptors responsible for the uptake of modified low den- ol synthesis in mouse peritoneal macrophages. Atherosclerosis 2006;186:267–74.
sity lipoprotein leading to lipid loading in macrophages. J Biol Chem 2002;277: [33] Yang L, Yang JB, Chen J, et al. Enhancement of human ACAT1 gene expression to
49982–8. promote the macrophage-derived foam cell formation by dexamethasone. Cell
[5] Handberg A, Hojlund K, Gastaldelli A, et al. Plasma sCD36 is associated with Res 2004;14:315–23.
markers of atherosclerosis, insulin resistance and fatty liver in a nondiabetic [34] Kusunoki J, Hansoty DK, Aragane K, Fallon JT, Badimon JJ, Fisher EA. Acyl-CoA:
healthy population. J Intern Med 2012;271:294–304. cholesterol acyltransferase inhibition reduces atherosclerosis in apolipoprotein
[6] Piechota M, Banaszewska A, Dudziak J, Slomczynski M, Plewa R. Highly upregulated E-deficient mice. Circulation 2001;103:2604–9.
expression of CD36 and MSR1 in circulating monocytes of patients with acute coro- [35] Fazio S, Major AS, Swift LL, et al. Increased atherosclerosis in LDL receptor-null
nary syndromes. Protein J 2012;31:511–8. mice lacking ACAT1 in macrophages. J Clin Invest 2001;107:163–71.
[7] Geloen A, Helin L, Geeraert B, Malaud E, Holvoet P, Marguerie G. CD36 inhibitors [36] Su YR, Dove DE, Major AS, et al. Reduced ABCA1-mediated cholesterol efflux and
reduce postprandial hypertriglyceridemia and protect against diabetic dyslipidemia accelerated atherosclerosis in apolipoprotein E-deficient mice lacking macrophage-
and atherosclerosis. PLoS One 2012;7:e37633. derived ACAT1. Circulation 2005;111:2373–81.
[8] Rac ME, Safranow K, Rac M, et al. CD36 gene is associated with thickness of ather- [37] Duewell P, Kono H, Rayner KJ, et al. NLRP3 inflammasomes are required for
omatous plaque and ankle-brachial index in patients with early coronary artery atherogenesis and activated by cholesterol crystals. Nature 2010;464:1357–61.
disease. Kardiol Pol 2012;70:918–23. [38] Ge J, Zhai W, Cheng B, et al. Insulin induces human acyl-coenzyme A: cholesterol
[9] Moore KJ, Kunjathoor VV, Koehn SL, et al. Loss of receptor-mediated lipid uptake acyltransferase1 gene expression via MAP kinases and CCAAT/enhancer-binding
via scavenger receptor A or CD36 pathways does not ameliorate atherosclerosis in protein alpha. J Cell Biochem 2013 [Epub ahead of print] [in press].
hyperlipidemic mice. J Clin Invest 2005;115:2192–201. [39] Liu W, He P, Cheng B, Mei CL, Wang YF, Wan JJ. Chlamydia pneumoniae disturbs
[10] Yuasa-Kawase M, Masuda D, Yamashita T, et al. Patients with CD36 deficiency are cholesterol homeostasis in human THP-1 macrophages via JNK-PPARgamma
associated with enhanced atherosclerotic cardiovascular diseases. J Atheroscler dependent signal transduction pathways. Microbes Infect 2010;12:1226–35.
Thromb 2012;19:263–75. [40] Hongo S, Watanabe T, Arita S, et al. Leptin modulates ACAT1 expression and cholesterol
[11] Inoue M, Tanabe H, Matsumoto A, et al. Astaxanthin functions differently as a efflux from human macrophages. Am J Physiol Endocrinol Metab 2009;297:E474–82.
selective peroxisome proliferator-activated receptor gamma modulator in adipo- [41] Hayashi K, Sasamura H, Azegami T, Itoh H. Regression of atherosclerosis in apoli-
cytes and macrophages. Biochem Pharmacol 2012;84:692–700. poprotein E-deficient mice is feasible using high-dose angiotensin receptor
[12] Mimche PN, Thompson E, Taramelli D, Vivas L. Curcumin enhances non-opsonic blocker, candesartan. J Atheroscler Thromb 2012;19:736–46.
phagocytosis of Plasmodium falciparum through up-regulation of CD36 surface ex- [42] Nagashima M, Watanabe T, Terasaki M, et al. Native incretins prevent the devel-
pression on monocytes/macrophages. J Antimicrob Chemother 2012;67:1895–904. opment of atherosclerotic lesions in apolipoprotein E knockout mice. Diabetologia
[13] Gao D, Pararasa C, Dunston CR, Bailey CJ, Griffiths HR. Palmitate promotes mono- 2011;54:2649–59.
cyte atherogenicity via de novo ceramide synthesis. Free Radic Biol Med 2012;53: [43] Cheng B, Wan J, Wang Y, et al. Ghrelin inhibits foam cell formation via simultaneously
796–806. down-regulating the expression of acyl-coenzyme A:cholesterol acyltransferase 1 and
[14] Goncalves JL, Roma EH, Gomes-Santos AC, et al. Pro-inflammatory effects of the up-regulating adenosine triphosphate-binding cassette transporter A1. Cardiovasc
mushroom Agaricus blazei and its consequences on atherosclerosis development. Pathol 2010;19:e159–66.
Eur J Nutr 2012;51:927–37. [44] Igarashi M, Osuga J, Isshiki M, et al. Targeting of neutral cholesterol ester hydro-
[15] Choi JS, Bae JY, Kim DS, et al. Dietary compound quercitrin dampens VEGF induc- lase to the endoplasmic reticulum via its N-terminal sequence. J Lipid Res
tion and PPARgamma activation in oxidized LDL-exposed murine macrophages: 2010;51:274–85.
association with scavenger receptor CD36. J Agric Food Chem 2010;58:1333–41. [45] Igarashi M, Osuga J, Uozaki H, et al. The critical role of neutral cholesterol ester
[16] Tang FT, Cao Y, Wang TQ, et al. Tanshinone IIA attenuates atherosclerosis in hydrolase 1 in cholesterol removal from human macrophages. Circ Res 2010;107:
ApoE(−/−) mice through down-regulation of scavenger receptor expression. Eur 1387–95.
J Pharmacol 2011;650:275–84. [46] Sekiya M, Osuga J, Nagashima S, et al. Ablation of neutral cholesterol ester hydro-
[17] Chiurchiu V, Izzi V, D'Aquilio F, et al. Endomorphin-1 prevents lipid accumulation lase 1 accelerates atherosclerosis. Cell Metab 2009;10:219–28.
via CD36 down-regulation and modulates cytokines release from human [47] Zhao B, Song J, Chow WN, St Clair RW, Rudel LL, Ghosh S. Macrophage-specific
lipid-laden macrophages. Peptides 2011;32:80–5. transgenic expression of cholesteryl ester hydrolase significantly reduces athero-
[18] Granados-Principal S, Quiles JL, Ramirez-Tortosa CL, et al. Squalene ameliorates sclerosis and lesion necrosis in Ldlr mice. J Clin Invest 2007;117:2983–92.
atherosclerotic lesions through the reduction of CD36 scavenger receptor expres- [48] Napolitano M, Avella M, Botham KM, Bravo E. Chylomicron remnant induction of
sion in macrophages. Mol Nutr Food Res 2012;56:733–40. lipid accumulation in J774 macrophages is associated with up-regulation of
[19] Li XY, Kong LX, Li J, He HX, Zhou YD. Kaempferol suppresses lipid accumulation in triacylglycerol synthesis which is not dependent on oxidation of the particles.
macrophages through the downregulation of cluster of differentiation 36 and the Biochim Biophys Acta 2003;1631:255–64.
upregulation of scavenger receptor class B type I and ATP-binding cassette trans- [49] Yamashita M, Tamasawa N, Matsuki K, et al. Insulin suppresses HDL-mediated cho-
porters A1 and G1. Int J Mol Med 2013;31:331–8. lesterol efflux from macrophages through inhibition of neutral cholesteryl ester
[20] Nergiz-Unal R, Kuijpers MJ, de Witt SM, et al. Atheroprotective effect of dietary hydrolase and ATP-binding cassette transporter G1 expressions. J Atheroscler Thromb
walnut intake in ApoE-deficient mice: involvement of lipids and coagulation 2010;17:1183–9.
factors. Thromb Res 2013;131:411–7. [50] McLaren JE, Michael DR, Salter RC, et al. IL-33 reduces macrophage foam cell
[21] Chu EM, Tai DC, Beer JL, Hill JS. Macrophage heterogeneity and cholesterol formation. J Immunol 2010;185:1222–9.
homeostasis: classically-activated macrophages are associated with reduced cho- [51] Miller AM, Liew FY. The IL-33/ST2 pathway — a new therapeutic target in cardio-
lesterol accumulation following treatment with oxidized LDL. Biochim Biophys vascular disease. Pharmacol Ther 2011;131:179–86.
Acta 1831;2013:378–86. [52] Bochem AE, van Wijk DF, Holleboom AG, et al. ABCA1 mutation carriers with low
[22] Dai XY, Cai Y, Mao DD, et al. Increased stability of phosphatase and tensin homo- high-density lipoprotein cholesterol are characterized by a larger atherosclerotic
log by intermedin leading to scavenger receptor A inhibition of macrophages burden. Eur Heart J 2013;34:286–91.
reduces atherosclerosis in apolipoprotein E-deficient mice. J Mol Cell Cardiol [53] Zhao Y, Pennings M, Vrins CL, et al. Hypocholesterolemia, foam cell accumulation,
2012;53:509–20. but no atherosclerosis in mice lacking ABC-transporter A1 and scavenger receptor
[23] Makinen PI, Lappalainen JP, Heinonen SE, et al. Silencing of either SR-A or CD36 BI. Atherosclerosis 2011;218:314–22.
reduces atherosclerosis in hyperlipidaemic mice and reveals reciprocal upregulation [54] Joyce CW, Wagner EM, Basso F, et al. ABCA1 overexpression in the liver
of these receptors. Cardiovasc Res 2010;88:530–8. of LDLr-KO mice leads to accumulation of pro-atherogenic lipoproteins and
[24] Manning-Tobin JJ, Moore KJ, Seimon TA, et al. Loss of SR-A and CD36 activity enhanced atherosclerosis. J Biol Chem 2006;281:33053–65.
reduces atherosclerotic lesion complexity without abrogating foam cell formation [55] Lee SM, Moon J, Cho Y, Chung JH, Shin MJ. Quercetin up-regulates expressions of
in hyperlipidemic mice. Arterioscler Thromb Vasc Biol 2009;29:19–26. peroxisome proliferator-activated receptor gamma, liver X receptor alpha, and
[25] Li K, Yao W, Zheng X, Liao K. Berberine promotes the development of atheroscle- ATP binding cassette transporter A1 genes and increases cholesterol efflux in
rosis and foam cell formation by inducing scavenger receptor A expression in human macrophage cell line. Nutr Res 2013;33:136–43.
macrophage. Cell Res 2009;19:1006–17. [56] Liu XY, Lu Q, Ouyang XP, et al. Apelin-13 increases expression of ATP-binding
[26] Hashizume M, Mihara M. Atherogenic effects of TNF-alpha and IL-6 via up-regulation cassette transporter A1 via activating protein kinase C alpha signaling in THP-1
of scavenger receptors. Cytokine 2012;58:424–30. macrophage-derived foam cells. Atherosclerosis 2013;226:398–407.
252 X.-H. Yu et al. / Clinica Chimica Acta 424 (2013) 245–252

[57] Park DW, Lee HK, Lyu JH, et al. TLR2 stimulates ABCA1 expression via PKC-eta and [71] Maitra U, Li L. Molecular mechanisms responsible for the reduced expression of
PLD2 pathway. Biochem Biophys Res Commun 2013;430:933–7. cholesterol transporters from macrophages by low-dose endotoxin. Arterioscler
[58] Malekpour-Dehkordi Z, Javadi E, Doosti M, et al. S-allylcysteine, a garlic com- Thromb Vasc Biol 2013;33:24–33.
pound, increases ABCA1 expression in human THP-1 macrophages. Phytother [72] Vergeer M, Korporaal SJ, Franssen R, et al. Genetic variant of the scavenger recep-
Res 2013;27:357–61. tor BI in humans. N Engl J Med 2011;364:136–45.
[59] Ku CS, Park Y, Coleman SL, Lee J. Unsaturated fatty acids repress expression of ATP [73] Zhang W, Yancey PG, Su YR, et al. Inactivation of macrophage scavenger receptor
binding cassette transporter A1 and G1 in RAW 264.7 macrophages. J Nutr class B type I promotes atherosclerotic lesion development in apolipoprotein
Biochem 2012;23:1271–6. E-deficient mice. Circulation 2003;108:2258–63.
[60] Yu XH, Jiang HL, Chen WJ, et al. Interleukin-18 and interleukin-12 together down- [74] Van Eck M, Bos IS, Hildebrand RB, Van Rij BT, Van Berkel TJ. Dual role for scavenger
regulate ATP-binding cassette transporter A1 expression through the interleukin- receptor class B, type I on bone marrow-derived cells in atherosclerotic lesion devel-
18R/nuclear factor-kappaB signaling pathway in THP-1 macrophage-derived foam opment. Am J Pathol 2004;165:785–94.
cells. Circ J 2012;76:1780–91. [75] Demetz E, Tancevski I, Duwensee K, et al. Inhibition of hepatic scavenger
[61] Sun D, Zhang J, Xie J, Wei W, Chen M, Zhao X. MiR-26 controls LXR-dependent receptor-class B type I by RNA interference decreases atherosclerosis in rabbits.
cholesterol efflux by targeting ABCA1 and ARL7. FEBS Lett 2012;586:1472–9. Atherosclerosis 2012;222:360–6.
[62] Schou J, Frikke-Schmidt R, Kardassis D, et al. Genetic variation in ABCG1 and risk [76] Voloshyna I, Hai O, Littlefield MJ, Carsons S, Reiss AB. Resveratrol mediates
of myocardial infarction and ischemic heart disease. Arterioscler Thromb Vasc anti-atherogenic effects on cholesterol flux in human macrophages and endothe-
Biol 2012;32:506–15. lium via PPARgamma and adenosine. Eur J Pharmacol 2013;698:299–309.
[63] Out R, Hoekstra M, Hildebrand RB, et al. Macrophage ABCG1 deletion disrupts [77] Kammerer I, Ringseis R, Biemann R, Wen G, Eder K. 13-hydroxy linoleic acid in-
lipid homeostasis in alveolar macrophages and moderately influences atheroscle- creases expression of the cholesterol transporters ABCA1, ABCG1 and SR-BI and
rotic lesion development in LDL receptor-deficient mice. Arterioscler Thromb stimulates apoA-I-dependent cholesterol efflux in RAW264.7 macrophages. Lipids
Vasc Biol 2006;26:2295–300. Health Dis 2011;10:222–9.
[64] Ranalletta M, Wang N, Han S, Yvan-Charvet L, Welch C, Tall AR. Decreased athero- [78] Uto-Kondo H, Ayaori M, Ogura M, et al. Coffee consumption enhances high-density
sclerosis in low-density lipoprotein receptor knockout mice transplanted with lipoprotein-mediated cholesterol efflux in macrophages. Circ Res 2010;106:
Abcg1−/− bone marrow. Arterioscler Thromb Vasc Biol 2006;26:2308–15. 779–87.
[65] Baldan A, Pei L, Lee R, et al. Impaired development of atherosclerosis in hyperlip- [79] Song G, Tian H, Qin S, et al. Hydrogen decreases athero-susceptibility in apolipo-
idemic Ldlr−/− and ApoE−/− mice transplanted with Abcg1−/− bone marrow. protein B-containing lipoproteins and aorta of apolipoprotein E knockout mice.
Arterioscler Thromb Vasc Biol 2006;26:2301–7. Atherosclerosis 2012;221:55–65.
[66] Meurs I, Lammers B, Zhao Y, et al. The effect of ABCG1 deficiency on atherosclerotic [80] Tang SL, Chen WJ, Yin K, et al. PAPP-A negatively regulates ABCA1, ABCG1 and
lesion development in LDL receptor knockout mice depends on the stage of athero- SR-B1 expression by inhibiting LXRalpha through the IGF-I-mediated signaling
genesis. Atherosclerosis 2012;221:41–7. pathway. Atherosclerosis 2012;222:344–54.
[67] Jun HJ, Hoang MH, Yeo SK, Jia Y, Lee SJ. Induction of ABCA1 and ABCG1 expression [81] Yoshinaka Y, Shibata H, Kobayashi H, et al. A selective ACAT-1 inhibitor, K-604,
by the liver X receptor modulator cineole in macrophages. Bioorg Med Chem Lett stimulates collagen production in cultured smooth muscle cells and alters plaque
2013;23:579–83. phenotype in apolipoprotein E-knockout mice. Atherosclerosis 2010;213:85–91.
[68] Hoang MH, Jia Y, Jun HJ, Lee JH, Lee BY, Lee SJ. Fucosterol is a selective liver X [82] Netherland C, Thewke DP. Rimonabant is a dual inhibitor of acyl CoA:cholesterol
receptor modulator that regulates the expression of key genes in cholesterol acyltransferases 1 and 2. Biochem Biophys Res Commun 2010;398:671–6.
homeostasis in macrophages, hepatocytes, and intestinal cells. J Agric Food Chem [83] Xie C, Kang J, Chen JR, Nagarajan S, Badger TM, Wu X. Phenolic acids are in vivo
2012;60:11567–75. atheroprotective compounds appearing in the serum of rats after blueberry con-
[69] Helal O, Berrougui H, Loued S, Khalil A. Extra-virgin olive oil consumption sumption. J Agric Food Chem 2011;59:10381–7.
improves the capacity of HDL to mediate cholesterol efflux and increases ABCA1 [84] Boden WE, Probstfield JL, Anderson T, et al. Niacin in patients with low HDL cho-
and ABCG1 expression in human macrophages. Br J Nutr 2012:1–12. lesterol levels receiving intensive statin therapy. N Engl J Med 2011;365:2255–67.
[70] Wang D, Xia M, Yan X, et al. Gut microbiota metabolism of anthocyanin promotes [85] Voight BF, Peloso GM, Orho-Melander M, et al. Plasma HDL cholesterol and risk of
reverse cholesterol transport in mice via repressing miRNA-10b. Circ Res 2012;111: myocardial infarction: a mendelian randomisation study. Lancet 2012;380:
967–81. 572–80.

You might also like