You are on page 1of 7

Clinica Chimica Acta 428 (2014) 82–88

Contents lists available at ScienceDirect

Clinica Chimica Acta


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

Invited critical review

ABCG5/ABCG8 in cholesterol excretion and atherosclerosis


Xiao-Hua Yu a, Kun Qian c, Na Jiang c, Xi-Long Zheng d, Francisco S. Cayabyab e, 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
The Second Affiliated Hospital, University of South China, Hengyang, Hunan 421001, China
d
Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary,
Alberta T2N 4N1, Canada
e
Department of Physiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada

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

Article history: Cholesterol is essential for the growth and function of all mammalian cells, but abnormally increased blood cho-
Received 19 September 2013 lesterol is a major risk factor for atherosclerotic cardiovascular disease. ATP-binding cassette (ABC) transporters
Received in revised form 7 November 2013 G5 (ABCG5) and G8 (ABCG8) form an obligate heterodimer that limits intestinal absorption and facilitates biliary
Accepted 9 November 2013
secretion of cholesterol and phytosterols. Consistent with their function, ABCG5 and ABCG8 are located on the
Available online 16 November 2013
apical membrane of enterocytes and hepatocytes. Liver X receptor is the major positive regulator of ABCG5
Keywords:
and ABCG8 expression. Mutations in either of the two genes cause sitosterolemia, a condition in which cholester-
ABCG5 ol and plant sterols accumulate in the circulation leading to premature cardiovascular disease. Overexpression of
ABCG8 ABCG5 and ABCG8 in mice retards diet-induced atherosclerosis because of reduced circulating and hepatic cho-
Cholesterol lesterol. In the current review, we summarize recent developments and propose a future framework that
Sitosterolemia provides new perspectives on the regulation of cholesterol metabolism and treatment of atherosclerotic cardio-
Atherosclerosis vascular disease.
© 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
83
2. Structure and characterization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
3. Sterol efflux function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
4. Genetic polymorphisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
5. Expression and regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
6. Sitosterolemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
7. Role in atherosclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
8. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
Disclosure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87

Abbreviations: ABCG5, ATP-binding cassette transporter G5; ABCG8, ATP-binding cassette transporter G8; NPC1L1, Niemann–Pick C1-Like 1; NBD, nucleotide binding domain; ER, en-
doplasmic reticulum; apoA-I, apolipoprotein A-I; HDL, high-density lipoprotein; LDL-C, low-density lipoprotein-cholesterol; VLDL-C, very low-density lipoprotein-cholesterol; LXR, liver X
receptor; TH, thyroid hormone; HNF4α, hepatocyte nuclear factor 4α; GATA4, GATA-binding protein 4; SNP, single nucleotide polymorphisms; HNF4α, hepatocyte nuclear factor 4α;
GATA4, GATA-binding protein 4; ET, endotoxin; LDLR, low-density lipoprotein receptor; PUFAs, polyunsaturated fatty acids.
⁎ Corresponding author at: Institute of Cardiovascular Research, University of South China, Hengyang, Hunan 421001, China. Tel./fax: +86 734 8281853.
E-mail address: tangchaoke@qq.com (C.-K. Tang).

0009-8981/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.cca.2013.11.010
X.-H. Yu et al. / Clinica Chimica Acta 428 (2014) 82–88 83

1. Introduction an adipocyte-derived hormone, exhibit decreased abundance of ABCG5


and ABCG8 proteins in the liver, which may occur at the level of the
Cholesterol is an essential structural component of mammalian cell ABCG5/ABCG8 heterodimer assembly within the ER, whereas leptin ad-
membranes and plays a crucial role in normal embryonic development, ministration restores both proteins, suggesting that leptin is required for
cell differentiation, nerve conduction, membrane fluidity and steroid the heterodimer formation [10]. As a lectin-like chaperone, calnexin or
synthesis. Therefore, the normal supply of cholesterol is important for its homolog calreticulin interacts with immature forms of ABCG5 and
human health. Too much cholesterol, however, may lead to pathological ABCG8 to stimulate their productive folding by inhibiting their degrada-
consequences. An uncontrolled buildup of cholesterol in cells is able to tion [11]. Moreover, calnexin has been shown to enhance the ER retention
disrupt membranes and facilitate apoptosis. Accumulation of excessive of ABCG5 and ABCG8; however, calreticulin predominantly promotes the
cholesterol in the blood causes hyperlipidemia and atherosclerotic car- cell surface expression of mature ABCG5 and ABCG8 [11]. These findings
diovascular disease, one of the major causes of death worldwide, and indicate a role for calnexin and calreticulin in the biosynthesis and quality
might contribute to the early onset of Alzheimer's disease [1–3]. control of ABCG5/ABCG8.
Cholesterol homeostasis in the body is maintained mainly by de Lack of expression of either half-transporter leads to the accumula-
novo synthesis, intestinal absorption, and biliary and fecal excretion. tion of the other in the ER as a result of impaired translocation to the
The biosynthesis of cholesterol is a well-defined energy-consuming plasma membrane, further indicating that ABCG5 and ABCG8 are
and feedback-regulated process, which is involved in a variety of enzy- obligate heterodimers for proper trafficking to the cell surface [12].
matic reactions [4]. Niemann–Pick C1-Like 1 (NPC1L1) mediates intesti- N-terminal cytosolic domains of ABCG5 and ABCG8 are also involved
nal cholesterol absorption and biliary cholesterol re-absorption [5]. It is in ER retention of their homodimers [13]. Interestingly, N-linked glyco-
also the target of ezetimibe, an inhibitor of dietary cholesterol uptake sylation of ABCG8 but not ABCG5 is essential for efficient trafficking of
which has been approved for the treatment of hypercholesterolemia. the heterodimer [14]. The two NBDs of the heterodimer are not func-
In direct opposition of NPC1L1, the heterodimer of ATP-binding cassette tionally equivalent, because the integrity of the canonical NBD formed
(ABC) transporters G5 (ABCG5) and G8 (ABCG8) has been shown to in- by the Walker A and B motifs of ABCG5 and the signature motif of
hibit the absorption of cholesterol and plant sterols from the diet by me- ABCG8 is essential for their sterol efflux function [15]. In addition, sub-
diating the efflux of these sterols from enterocytes back into gut lumen, stitutions of the D-loop aspartate and Q-loop glutamine in either NBD
and by promoting efficient secretion of cholesterol and plant sterols do not interfere with sterol transport [16].
from hepatocytes into bile [6]. Thus, the heterodimer constitutes the
body's predominant defense against the accumulation of sterols derived 3. Sterol efflux function
from the diet. This review focuses on the molecular characterization,
physiology, genetic polymorphisms, and regulation of ABCG5/ABCG8, ABCG5 and ABCG8 can promote cholesterol and plant sterol elimina-
and explores their emerging pathogenic significance and therapeutic tion from the body through hepatobiliary secretion (Fig. 2). Disruption
potential in atherosclerosis. of both ABCG5 and ABCG8 in mice leads to extremely low biliary choles-
terol concentrations while biliary phospholipids and bile acid concentra-
2. Structure and characterization tions are not altered, revealing transport selectivity of the heterodimer
[17]. Similar effects are also observed in mice lacking either ABCG5 or
The genes of ABCG5 and ABCG8 map to chromosome 2p21 in a head- ABCG8 [18,19]. Conversely, overexpression of ABCG5 and ABCG8 in
to-head orientation with only 140 bases separating their first exons. mice increases biliary cholesterol levels by more than five fold [20]. It is
Each is composed of 13 exons and 12 introns. The two genes encode worth noting that ABCG5/ABCG8-independent biliary cholesterol excre-
two different proteins, namely ABCG5 (sterolin-1) and ABCG8 (sterolin- tion has been reported under some conditions [21,22], which needs fur-
2). Each protein has an ATP-binding cassette near the N terminus, a trans- ther exploration.
membrane domain containing six helices, and a nucleotide binding do- Some factors have also been identified to influence the effectiveness
main (NBD) containing two conserved peptide motifs (Walkers A and of ABCG5/ABCG8 in secreting cholesterol from the liver. In canine gall-
B) and a signature motif (Fig. 1) [7]. bladder epithelial cells transfected with lentiviral constructs containing
ABCG5 or ABCG8 serves only as a non-functional half-transporter mouse ABCG5 and ABCG8, cholesterol secretion is significantly in-
when expressed alone. Thus, they must form the heterodimer to obtain creased in the presence of bile salts, whereas other cholesterol acceptors
sterol transport functionality. A variety of studies have shown that including apolipoprotein A-I (apoA-I), high-density lipoprotein (HDL)
ABCG5 and ABCG8 heterodimerize in the endoplasmic reticulum (ER), or methyl-β-cyclodextrin fail to elicit this effect, suggesting that bile
transfer together through the Golgi apparatus, and eventually move to salts serve as a cholesterol acceptor for the heterodimer [23]. ABCB4,
apical subdomains in the plasma membrane [8,9]. Mice lacking leptin, also called multidrug resistance protein 2 (MDR2), is a canalicular phos-
phatidylcholine flippase of hepatocytes. Deficiency of ABCB4 in mice
disrupts ABCG5/ABCG8-mediated secretion of cholesterol into the bile,
which indicates that ABCG5 and ABCG8 require ABCB4 for the efficient
biliary export of sterols [24].
Opposing the previously described function for NPC1L1 in the small
intestine, ABCG5 and ABCG8 transport cholesterol and plant sterols
from enterocytes into the gut lumen for fecal disposal (Fig. 2). Mice
overexpressing ABCG5 and ABCG8 exhibit a 50% decrease in the frac-
tional absorption of dietary cholesterol [20]. A recent study by Wang
et al. found that there is a negative correlation between the efficiency
of cholesterol absorption and the abundance of ABCG5 and ABCG8 in
the jejunum and ileum but not the duodenum in inbred mice, suggest-
ing that ABCG5 and ABCG8 in the jejunum and ileum are predominant
determinants of intestinal cholesterol absorption [25]. On the other
hand, ABCG5/ABCG8-deficient mice have a 2- to 3-fold increase in the
Fig. 1. Schematic of ABCG5 and ABCG8 protein structure. ABCG5 and ABCG8 belong to half-
ABC transporters, which consist of a transmembrane domain containing six helices and a
fractional absorption of dietary plant sterols as well as an approximately
NBD region containing two conserved peptide motifs (Walkers A and B) and a signature 30-fold elevation in plasma sitosterol levels, and display many charac-
motif at the N-terminus. teristics similar to those seen in sitosterolemic patients [17].
84 X.-H. Yu et al. / Clinica Chimica Acta 428 (2014) 82–88

Fig. 2. Sterol efflux function of the ABCG5/ABCG8 heterodimer in vivo. The ABCG5/ABCG8 heterodimer locates to the brush border membrane of enterocytes and the canalicular membrane
of hepatocytes. ABCG5 and ABCG8 secrete sterols including phytosterols and cholesterol from hepatocytes into the bile. Biliary sterols account for more than two thirds of the total amount
of sterols in the intestinal lumen. Because the sterols are hydrophobic, they are transferred to the brush border membrane of enterocytes via bile salt micelles. Then, ABCG5 and ABCG8
mediate biliary and dietary sterol efflux from the brush border membrane back into the gut lumen for excretion. NPC1L1 is also present in the apical membrane of enterocytes and hepa-
tocytes, and can facilitate intestinal and biliary cholesterol absorption. Most cholesterol taken up into enterocytes by NPC1L1 is subsequently esterified, packaged into chylomicrons, and
then secreted into the lymph. The chylomicrons are finally delivered to hepatocytes through the circulatory system. This process is called the enterohepatic circulation of sterols. Ezetimibe
specially suppresses NPC1L1-dependent cholesterol absorption in the small intestine and liver.

Plant sterols are the non-saponifiable component of plant oils, and hypercholesterolemia cohort indicates that the T400K polymorphism
have a chemical structure similar to cholesterol except for the addition in the ABCG8 gene has an increased risk of cardiovascular disease but
of an extra methyl or ethyl group. Dietary intake of plant sterols can de- is not associated with cardiovascular endpoints [31]. Carriers of the
crease plasma levels of total cholesterol and low-density lipoprotein- minor G allele at the ABCG5_Gln604GluCN G single nucleotide polymor-
cholesterol (LDL-C) in humans because of their ability to displace cho- phism (SNP) show significantly lower very low-density lipoprotein-
lesterol from the mixed micelles in the upper small intestine, thereby cholesterol (VLDL-C) and triglyceride concentrations than homozygous
reducing fractional cholesterol absorption that is determined as the C/C in patients with familial hypercholesterolemia [32]. Additionally, the
ratio of [14C]cholesterol to [5,6-3H]sitostanol by a fecal dual isotope frequency of the CC homozygous genotype for the ABCG5 1950CNG poly-
method to indicate the efficiency of cholesterol absorption [26]. In addi- morphism is significantly increased in hypercholesterolemic patients as
tion to this, other mechanisms have been proposed. Plant sterol feeding compared to healthy controls, whereas no significant difference for the
for 2 weeks results in a dose-dependent decrease of fractional choles- 251AN G polymorphism of the ABCG8 gene is found, revealing a possible
terol absorption (2- to 7-fold reduction) in wild type mice and an 80% link between ABCG5 1950CNG polymorphism and hypercholesterolemia
reduction in mice lacking ABCG5, and stimulates intestinal cholesterol [33].
excretion by 500% and 250% in wild type and ABCG5−/− mice, respec-
tively [27]. These observations indicate that the ABCG5/ABCG8 complex 5. Expression and regulation
is also involved in plant sterol-induced efflux of cholesterol.
ABCG5 and ABCG8 are expressed almost exclusively on the brush
4. Genetic polymorphisms border membrane of enterocytes and the canalicular membrane of he-
patocytes [12]. More recently, Yoon et al. observed that ABCG8 forms
Recently, a variety of studies have focused on the genetic polymor- a diffuse intracellular pattern at the apical pole of human gallbladder ep-
phisms of ABCG5 and ABCG8. It is found that cholesterol absorption is ithelial cells, and that ABCG5 and ABCG8 protein levels are increased in
about 24% lower in individuals carrying the p.D19H allele of ABCG8 human gallbladder with cholesterol-related gallbladder diseases [34].
compared to wild type although there is no significant difference in This suggests a role for the heterodimer in the pathogenesis of human
ABCG5 and ABCG8 expression levels, indicating an important role for cholesterol-related gallbladder diseases.
the ABCG8 19H allele in promoting cholesterol excretion [28]. The inter- The expression of ABCG5 and ABCG8 is primarily regulated at the
action between ABCG8 1199A and the bile acid biosynthetic enzyme transcriptional level (Fig. 3). Liver X receptor α (LXRα), a ligand-
7α-hydroxylase (CYP7A1)-204A allele has been shown to affect lipid- activated nuclear transcription factor involved in the control of lipid me-
lowering response to atorvastatin in a Chinese population [29]. In hy- tabolism, is regarded as the major regulator of ABCG5 and ABCG8 mRNA
percholesterolemic Japanese subjects, serum sitosterol levels and the expression. In support of this concept, both soy protein (SP) diet and
sitosterol/cholesterol ratio are higher in carriers of the ABCG8 M429V LXR agonist T0901317 markedly upregulate ABCG5 and ABCG8 mRNA
variant than non-carriers [30]. A recent report in a heterozygous familial expression in the small intestine and liver of wild type mice, but not
X.-H. Yu et al. / Clinica Chimica Acta 428 (2014) 82–88 85

Fig. 3. Regulation of ABCG5 and ABCG8 expression. Both SP and T0901317 can induce ABCG5/ABCG8 expression by activating LXRα. Their expression is also upregulated by n−3 PUFAs in
a HNF4α-dependent manner. The interaction between HNF4α and GATA4 promotes ABCG5/ABCG8 gene transcription. Fish oil, açaí pulp, TH, CBE and HF/HG significantly increase ABCG5/
ABCG8 levels, whereas atorvastatin and ET decrease their levels. Additionally, binding of miR-200C to ABCG5 leads to a significant decrease in ABCG5 protein.

in LXRα knockout mice [35–37]. Moreover, two LXR response elements showing N-linked glycosylation of ABCG5 and ABCG8 as reported by
in the ABCG5 and ABCG8 genes have been identified [38]. Thyroid hor- Graf et al. [12], it is surprising that there are no reports to date regarding
mone (TH), an important regulator of cholesterol metabolism, has been other modes of potential posttranslational modifications of these ABC
shown to raise hepatic gene expression of ABCG5 and ABCG8 in a LXR- transporters. For example, analyses of the intracellular domains of
independent mechanism [39]. Hepatocyte nuclear factor 4α (HNF4α) mouse ABCG5 and ABCG8 revealed numerous consensus sites for ser-
and GATA-binding protein 4 (GATA4) are also transcription factors, ine, threonine and tyrosine phosphorylation. Future investigations in
and they can synergistically stimulate ABCG5 and ABCG8 gene tran- assessing the functional consequences of protein kinase or phosphatase
scription in HepG2 cells [40]. Recently, Kim et al. generated fat-1 trans- modulation of ABC transporters should lead to the development of
genic mice expressing the Caenorhabditis elegans fat-1 gene encoding an other therapeutic options for regulating their function or expression.
n−3 fatty acid desaturase, an enzyme absent in mammals, which con- Many other factors that modulate their expression at the posttranscrip-
verts n − 6 polyunsaturated fatty acids (PUFAs) to n − 3 PUFAs [41]. tional level, such as protein–protein interactions with scaffolding mole-
These fat-1 transgenic mice fed with a high-fat diet exhibit a significant cules, may be discovered in future research.
increase in hepatic mRNA levels of ABCG5 and ABCG8 by activating
HNF4α, suggesting that the endogenously synthesized n−3 PUFAs act 6. Sitosterolemia
as a positive regulator for ABCG5/ABCG8 expression [41]. It has been
suggested that the expression of hepatic ABCG5 and ABCG8 genes in a Sitosterolemia, a rare autosomal recessive disorder, is characterized
rat model of dietary-induced hypercholesterolemia is significantly in- by markedly elevated plasma levels of plant sterols and modest increases
creased in the presence of açaí pulp, a fruit of the Euterpe oleracea in plasma cholesterol, which is attributable to the hyperabsorption of
Martius tree [42]. Fish oil has been shown to strongly induce the these sterols from the small intestine and a reduced excretion into the
mRNA expression of biliary ABCG5 and ABCG8 in rats [43]. On the bile. Due to these abnormal circulating sterol levels, sitosterolemic pa-
other hand, atorvastatin, an inhibitor of 3-hydroxyl-3-methylglutaryl- tients develop atherosclerosis and premature cardiovascular disease
CoA reductase, has been shown to decrease the mRNA levels of both [50]. Other clinical manifestations include tendon xanthomas, hemolytic
ABCG5 and ABCG8 in the duodenum of hyperlipidemic men [44]. Endo- anemia, hypersplenism, macrothrombocytopenia and bleeding. A recent
toxin (ET), the major component of the outer membrane of Gram- report indicates that intercalation of plant sterols into the plasma mem-
negative bacteria, is released during bacterial cell division or lysis [45]. brane induces dysregulation of multiple platelet activation pathways,
It has been reported that ET markedly downregulates ABCG5 and resulting in macrothrombocytopenia and bleeding [51]. This disease is
ABCG8 mRNA expression in the murine liver [46], but the molecular caused by mutations in the genes encoding ABCG5 or ABCG8 [52]. More-
mechanism involved in this response is not known. Taken together, it over, all missense mutations that have been investigated to date either in-
is quite obvious that ABCG5 and ABCG8 are coordinately modulated hibit formation of the ABCG5/ABCG8 heterodimers or impede their
by multiple transcriptional processes. More work in this field may prove efficient transfer from the ER to the plasma membrane [14].
to be key for future ABCG5/ABCG8-based therapeutic intervention. Since increased amounts of cholesterol and phytosterols in the blood
ABCG5 and ABCG8 are also highly modulated posttranscriptionally are deposited on the arterial walls, treatment to reduce plasma sterol
(Fig. 3). It has already been reported that protein levels of ABCG5 and levels may be crucial to prevent life-threatening cardiovascular events
ABCG8 in bile canaliculi of mice are significantly increased by a high- in patients with sitosterolemia. So far, low sterol diets, partial ileal by-
fat/high-sucrose (HF/HG) diet as compared to the standard diet [47]. pass surgery that is defined as shortening of the ileum to decrease the
Black chokeberry (Aronia melanocarpa) is a rich source of polyphenols. total small intestinal length, and drugs such as statins and sitostanol
The polyphenol-rich black chokeberry extract (CBE) significantly en- have been used to cure sitosterolemia; however, these therapies are
hances the levels of ABCG5 and ABCG8 in Caco-2 cells [48]. MicroRNAs only partially effective [53]. Thus, alternative strategies need to be pur-
(miRNAs) are short non-coding RNAs that bind to target mRNAs and sued. Recently, Tang et al. found that genetic inactivation of NPC1L1, a
cause inhibition of translation or initiation of mRNA degradation. Over- cholesterol transporter with the opposite effect of ABCG5/ABCG8, mark-
expression of miR-200c, a member of the miR-200 family, results in a edly protects against sitosterolemia in mice lacking ABCG5/ABCG8 [54].
significant downregulation of ABCG5, indicating a novel regulatory Ezetimibe has been demonstrated to bind directly to NPC1L1 and acts by
role of miR-200c in lipid metabolism [49]. Thus, miR-200c inhibitors blocking sterol-induced internalization of NPC1L1, subsequently sup-
can potentially be used to treat atherosclerosis. Apart from evidence pressing the absorption of sitosterol and cholesterol. Based on this,
86 X.-H. Yu et al. / Clinica Chimica Acta 428 (2014) 82–88

ezetimibe should have the potential to be a revolutionary treatment sitosterolemic patients are prone to premature atherosclerotic cardio-
agent for sitosterolemia. Indeed, in a multicenter, double-blind, ran- vascular disease.
domized, placebo-controlled study, plasma sitosterol concentrations Considering ABCG5/ABCG8 as cholesterol's way out of the body, ac-
were reduced by 21% in sitosterolemia patients treated with ezetimibe tivation of the heterodimer may provide benefits for regulation of lipid
10 mg/day for 8 weeks as compared to an insignificant 4% rise in metabolism and treatment of atherosclerosis. Indeed, curcuma oil, a li-
those on the placebo, with no serious treatment-related adverse events pophilic component from Curcuma longa L., significantly decreases plas-
or discontinuations due to adverse events [55]. Moreover, in patients ma total cholesterol and LDL-C, but elevates HDL-C, in male golden
with homozygous sitosterolemia, long-term treatment with ezetimibe Syrian hamsters fed with a high-cholesterol and fat-rich diet, primarily
10 mg/day for 2 years was also effective in decreasing plasma sitosterol by upregulation of ABCG5/ABCG8 expression in the liver and jejunum
and LDL-C concentrations with an overall favorable safety and tolerabil- [65]. CM108, a flavone derivative, also improves lipid profiles in hyper-
ity profile [56]. lipidemic rats via enhancing hepatic ABCG5/ABCG8 levels [66]. Similar-
ly, the hypolipidemic effects of n-butanol extract (a LXRα agonist) of
7. Role in atherosclerosis Panax notoginseng root are associated with activation of ABCG5/ABCG8
[67]. It is worth noting that overexpression of human ABCG5 and
Because the ABCG5/ABCG8 heterodimer plays a key role in choles- ABCG8 in the liver, but not intestine, enhances hepatobiliary secretion
terol removal from the body, it seems logical to presume that overex- of cholesterol and plant sterols by 1.5- to 2-fold, but does not affect
pression of ABCG5 and ABCG8 should inhibit atherosclerosis while the development of proximal aortic atherosclerosis in apoE or LDLR
knockout of ABCG5 and ABCG8 should promote atherosclerosis. In knockout mice [68]. This suggests that therapies which simply raise
agreement, high-level expression of ABCG5 and ABCG8 in both the in- biliary sterol output are not always effective, because these sterols can
testine and liver of low-density lipoprotein receptor (LDLR)-deficient be re-absorbed by intestinal NPC1L1. Thus, increased biliary cholesterol
mice leads to a significant reduction in plasma cholesterol levels and secretion must be coupled with decreased intestinal cholesterol absorp-
aortic atherosclerotic lesion area [57]. Treatment of female Fischer rats tion to enhance net sterol loss from the body and inhibit atherosclerosis.
with a hypercholesterolemic diet supplemented with 2% açaí pulp To test this hypothesis, the same authors found in a subsequent study
causes a significant decrease in serum total cholesterol, LDL-C, and ath- that treatment of LDLR knockout mice overexpressing hepatic ABCG5
erogenic index, as well as a significant increase in HDL-cholesterol and and ABCG8 with ezetimibe markedly increases hepatobiliary cholester-
cholesterol excretion in feces through upregulation of hepatic ABCG5 ol secretion rates and reduces intestinal cholesterol absorption, thereby
and ABCG8 expression [42]. Evodiamine, a main bioactive component producing profound protection against atherosclerosis [69]. Therefore,
in the fruit of Evodia rutaecarpa, has been reported to reduce the size dual therapies targeting both ABCG5/ABCG8 and NPC1L1 may be a via-
of atherosclerotic lesions and alleviate the hyperlipidemia in apolipo- ble approach to prevent atherosclerosis in the future.
protein E (apoE)-deficient mice via promotion of ABCG5/ABCG8-
dependent hepatic cholesterol clearance [58]. Li et al. have demonstrat-
ed that the levels of ABCG5 and ABCG8 in the liver are significantly 8. Conclusions
higher in the atherosclerosis-resistant than in the atherosclerosis-
susceptible Japanese quail (Coturnix japonica) strains [59]. In a large The discovery of ABCG5 and ABCG8 is an exciting result in cardiovas-
cohort study of 2012 patients with heterozygous familial hypercholes- cular research. In contrast to NPC1L1, the ABCG5/ABCG8 heterodimer is
terolemia, the ABCG8 rs11887534 variant shows an association with a major route to facilitate excess cholesterol clearance from the body.
higher risk of coronary heart disease [31]. Mice treated with low-dose Defects in ABCG5 and/or ABCG8 are linked to sitosterolemia. Converse-
lipopolysaccharide have reduced bile and fecal counts as well as ly, upregulation of ABCG5 and ABCG8 levels can promote reverse
decreased expression of hepatic ABCG5 and ABCG8, but there is cholesterol transport and reduce atherosclerosis. Since cholesterol ex-
no effect on plasma or liver counts, suggesting that attenuation of cretion is a complex and multistep process that is regulated by multiple
reverse cholesterol transport, particularly cholesterol efflux from genes at the hepatocyte and enterocyte levels, we should also pay atten-
liver to bile and feces, may contribute to atherosclerosis in chronic tion to the effect of other proteins such as NPC1L1 and ABCB4. Besides,
inflammatory states such as obesity, metabolic syndrome, and type there are still many problems left to be solved. How is the heterodimer
2 diabetes [60]. transported from the endoplasmic reticulum to the plasma membrane?
It is generally believed that many sitosterolemic patients suffer from Can these two transporters directly bind and translocate their sub-
premature atherosclerotic cardiovascular disease. However, whether strates? Are there efficient avenues to enhance their function? Can
the atherosclerotic cardiovascular disease can be attributed to an in- a dual therapy of a potential ABCG5/ABCG8 activator plus ezetimibe
creased concentration of plasma plant sterols remains uncertain. Coro- lead to improved protection against atherosclerosis in humans?
nary heart disease still develops in some subjects with sitosterolemia, Understanding these questions will provide insightful knowledge
despite normal plasma cholesterol levels, suggesting that high circulat- regarding the underlying mechanisms of cholesterol output from
ing levels of plant sterols may be atherogenic [61]. In contrast, rapeseed the body and accelerate the future design of novel therapeutic
oil fortified with phytosterols contributes to protection against athero- reagents.
genesis by improving plasma oxidative stress, lipid profiles as well as in-
flammation [62]. Horenstein et al. have demonstrated that although the Disclosure
G574R variant of ABCG8 is associated with moderately elevated plant
sterol levels, carriers of the G574R allele have modestly thinner carotid The authors have declared no conflict of interest.
wall thickness than noncarriers [63]. More recently, Wilund et al. re-
ported that there is no statistically significant difference in aortic ath-
erosclerotic lesion area between ABCG5/ABCG8-deficient mice and Acknowledgments
littermate controls, and that plasma levels of cholesterol, but not sitos-
terol or campesterol, are significantly higher in humans with coronary The authors gratefully acknowledge the financial support from
atherosclerosis, indicating no association between plasma plant sterol the National Natural Sciences Foundation of China (81070220 and
levels and atherosclerosis in mice and humans [64]. In this case, perhaps 81170278), Aid Program for Science and Technology Innovative Re-
increased plasma cholesterol levels are responsible for the progression search Team in Higher Educational Institutions of Hunan Province,
of premature atherosclerotic cardiovascular disease in patients with China (2008-244) and the construct program of the key discipline in
sitosterolemia [64]. Overall, more studies are needed to clarify why Hunan province.
X.-H. Yu et al. / Clinica Chimica Acta 428 (2014) 82–88 87

References [35] Gonzalez-Granillo M, Steffensen KR, Granados O, et al. Soy protein isoflavones
differentially regulate liver X receptor isoforms to modulate lipid metabolism
[1] Barton M. Cholesterol and atherosclerosis: modulation by oestrogen. Curr Opin and cholesterol transport in the liver and intestine in mice. Diabetologia
Lipidol 2013;24:214–20. 2012;55:2469–78.
[2] Kihara S. Dyslipidemia. Nihon Rinsho 2013;71:275–9. [36] Calpe-Berdiel L, Rotllan N, Fievet C, et al. Liver X receptor-mediated activation of re-
[3] Wellington CL. Cholesterol at the crossroads: Alzheimer's disease and lipid metabo- verse cholesterol transport from macrophages to feces in vivo requires ABCG5/G8. J
lism. Clin Genet 2004;66:1–16. Lipid Res 2008;49:1904–11.
[4] Robichon C, Dugail I. De novo cholesterol synthesis at the crossroads of adaptive re- [37] van der Veen JN, Havinga R, Bloks VW, et al. Cholesterol feeding strongly reduces he-
sponse to extracellular stress through SREBP. Biochimie 2007;89:260–4. patic VLDL-triglyceride production in mice lacking the liver X receptor alpha. J Lipid
[5] Wang LJ, Song BL. Niemann–Pick C1-Like 1 and cholesterol uptake. Biochim Biophys Res 2007;48:337–47.
Acta 1821;2012:964–72. [38] Back SS, Kim J, Choi D, et al. Cooperative transcriptional activation of ATP-binding
[6] Brown JM, Yu L. Opposing gatekeepers of apical sterol transport: Niemann–Pick C1- cassette sterol transporters ABCG5 and ABCG8 genes by nuclear receptors including
Like 1 (NPC1L1) and ATP-binding cassette transporters G5 and G8 (ABCG5/ABCG8). Liver-X-Receptor. BMB Rep 2013;46:322–7.
Immunol Endocr Metab Agents Med Chem 2009;9:18–29. [39] Bonde Y, Plosch T, Kuipers F, et al. Stimulation of murine biliary cholesterol secretion
[7] Brown JM, Yu L. Protein mediators of sterol transport across intestinal brush border by thyroid hormone is dependent on a functional ABCG5/G8 complex. Hepatology
membrane. Subcell Biochem 2010;51:337–80. 2012;56:1828–37.
[8] Calandra S, Tarugi P, Speedy HE, et al. Mechanisms and genetic determinants regu- [40] Sumi K, Tanaka T, Uchida A, et al. Cooperative interaction between hepatocyte nu-
lating sterol absorption, circulating LDL levels, and sterol elimination: implications clear factor 4 alpha and GATA transcription factors regulates ATP-binding cassette
for classification and disease risk. J Lipid Res 2011;52:1885–926. sterol transporters ABCG5 and ABCG8. Mol Cell Biol 2007;27:4248–60.
[9] Dikkers A, Tietge UJ. Biliary cholesterol secretion: more than a simple ABC. World J [41] Kim EH, Bae JS, Hahm KB, et al. Endogenously synthesized n−3 polyunsaturated
Gastroenterol 2010;16:5936–45. fatty acids in fat-1 mice ameliorate high-fat diet-induced non-alcoholic fatty liver
[10] Sabeva NS, Rouse EJ, Graf GA. Defects in the leptin axis reduce abundance of the disease. Biochem Pharmacol 2012;84:1359–65.
ABCG5–ABCG8 sterol transporter in liver. J Biol Chem 2007;282:22397–405. [42] de Souza MO, Souza ESL, de Brito Magalhaes CL, et al. The hypocholesterolemic ac-
[11] Okiyoneda T, Kono T, Niibori A, et al. Calreticulin facilitates the cell surface expres- tivity of acai (Euterpe oleracea Mart.) is mediated by the enhanced expression of
sion of ABCG5/G8. Biochem Biophys Res Commun 2006;347:67–75. the ATP-binding cassette, subfamily G transporters 5 and 8 and low-density lipopro-
[12] Graf GA, Li WP, Gerard RD, et al. Coexpression of ATP-binding cassette proteins tein receptor genes in the rat. Nutr Res 2012;32:976–84.
ABCG5 and ABCG8 permits their transport to the apical surface. J Clin Invest [43] Takahashi Y. Soy protein and fish oil independently decrease serum lipid con-
2002;110:659–69. centrations but interactively reduce hepatic enzymatic activity and gene expres-
[13] Hirata T, Okabe M, Kobayashi A, et al. Molecular mechanisms of subcellular localiza- sion involved in fatty acid synthesis in rats. J Nutr Sci Vitaminol (Tokyo)
tion of ABCG5 and ABCG8. Biosci Biotechnol Biochem 2009;73:619–26. 2011;57:56–64.
[14] Graf GA, Cohen JC, Hobbs HH. Missense mutations in ABCG5 and ABCG8 disrupt [44] Tremblay AJ, Lamarche B, Lemelin V, et al. Atorvastatin increases intestinal expres-
heterodimerization and trafficking. J Biol Chem 2004;279:24881–8. sion of NPC1L1 in hyperlipidemic men. J Lipid Res 2011;52:558–65.
[15] Zhang DW, Graf GA, Gerard RD, et al. Functional asymmetry of nucleotide-binding [45] Lodowska J, Wolny D, Weglarz L. The sugar 3-deoxy-d-manno-oct-2-ulosonic acid
domains in ABCG5 and ABCG8. J Biol Chem 2006;281:4507–16. (Kdo) as a characteristic component of bacterial endotoxin — a review of its biosyn-
[16] Wang J, Grishin N, Kinch L, et al. Sequences in the nonconsensus nucleotide-binding do- thesis, function, and placement in the lipopolysaccharide core. Can J Microbiol
main of ABCG5/ABCG8 required for sterol transport. J Biol Chem 2011;286:7308–14. 2013;59:645–55.
[17] Yu L, Hammer RE, Li-Hawkins J, et al. Disruption of Abcg5 and Abcg8 in mice reveals [46] Khovidhunkit W, Moser AH, Shigenaga JK, et al. Endotoxin down-regulates ABCG5
their crucial role in biliary cholesterol secretion. Proc Natl Acad Sci U S A and ABCG8 in mouse liver and ABCA1 and ABCG1 in J774 murine macrophages: dif-
2002;99:16237–42. ferential role of LXR. J Lipid Res 2003;44:1728–36.
[18] Plosch T, Bloks VW, Terasawa Y, et al. Sitosterolemia in ABC-transporter G5-deficient [47] Yamazaki Y, Hashizume T, Morioka H, et al. Diet-induced lipid accumulation in liver
mice is aggravated on activation of the liver-X receptor. Gastroenterology enhances ATP-binding cassette transporter g5/g8 expression in bile canaliculi. Drug
2004;126:290–300. Metab Pharmacokinet 2011;26:442–50.
[19] Wang HH, Patel SB, Carey MC, et al. Quantifying anomalous intestinal sterol uptake, [48] Kim B, Park Y, Wegner CJ, et al. Polyphenol-rich black chokeberry (Aronia
lymphatic transport, and biliary secretion in Abcg8(−/−) mice. Hepatology melanocarpa) extract regulates the expression of genes critical for intestinal choles-
2007;45:998–1006. terol flux in Caco-2 cells. J Nutr Biochem 2013;24:1564–70.
[20] Yu L, Li-Hawkins J, Hammer RE, et al. Overexpression of ABCG5 and ABCG8 pro- [49] Liu S, Tetzlaff MT, Cui R, et al. miR-200c inhibits melanoma progression
motes biliary cholesterol secretion and reduces fractional absorption of dietary cho- and drug resistance through down-regulation of BMI-1. Am J Pathol
lesterol. J Clin Invest 2002;110:671–80. 2012;181:1823–35.
[21] Coy DJ, Wooton-Kee CR, Yan B, et al. ABCG5/ABCG8-independent biliary cholesterol [50] Salen G, Patel S, Batta AK. Sitosterolemia. Cardiovasc Drug Rev 2002;20:255–70.
excretion in lactating rats. Am J Physiol Gastrointest Liver Physiol 2010;299: [51] Kanaji T, Kanaji S, Montgomery RR, et al. Platelet hyperreactivity explains the bleed-
G228–35. ing abnormality and macrothrombocytopenia in a murine model of sitosterolemia.
[22] Groen A, Kunne C, Jongsma G, et al. Abcg5/8 independent biliary cholesterol excre- Blood 2013;122:2732–42.
tion in Atp8b1-deficient mice. Gastroenterology 2008;134:2091–100. [52] Berge KE, Tian H, Graf GA, et al. Accumulation of dietary cholesterol in
[23] Vrins C, Vink E, Vandenberghe KE, et al. The sterol transporting heterodimer sitosterolemia caused by mutations in adjacent ABC transporters. Science
ABCG5/ABCG8 requires bile salts to mediate cholesterol efflux. FEBS Lett 2000;290:1771–5.
2007;581:4616–20. [53] Tsubakio-Yamamoto K, Nishida M, Nakagawa-Toyama Y, et al. Current therapy for
[24] Langheim S, Yu L, von Bergmann K, et al. ABCG5 and ABCG8 require MDR2 for secre- patients with sitosterolemia—effect of ezetimibe on plant sterol metabolism. J
tion of cholesterol into bile. J Lipid Res 2005;46:1732–8. Atheroscler Thromb 2010;17:891–900.
[25] Duan LP, Wang HH, Wang DQ. Cholesterol absorption is mainly regulated by the je- [54] Tang W, Ma Y, Jia L, et al. Genetic inactivation of NPC1L1 protects against
junal and ileal ATP-binding cassette sterol efflux transporters Abcg5 and Abcg8 in sitosterolemia in mice lacking ABCG5/ABCG8. J Lipid Res 2009;50:293–300.
mice. J Lipid Res 2004;45:1312–23. [55] Salen G, von Bergmann K, Lutjohann D, et al. Ezetimibe effectively reduces plasma
[26] Wu T, Fu J, Yang Y, et al. The effects of phytosterols/stanols on blood lipid plant sterols in patients with sitosterolemia. Circulation 2004;109:966–71.
profiles: a systematic review with meta-analysis. Asia Pac J Clin Nutr [56] Lutjohann D, von Bergmann K, Sirah W, et al. Long-term efficacy and safety of
2009;18:179–86. ezetimibe 10 mg in patients with homozygous sitosterolemia: a 2-year, open-label
[27] Brufau G, Kuipers F, Lin Y, et al. A reappraisal of the mechanism by which plant ste- extension study. Int J Clin Pract 2008;62:1499–510.
rols promote neutral sterol loss in mice. PLoS One 2011;6:e21576. [57] Wilund KR, Yu L, Xu F, et al. High-level expression of ABCG5 and ABCG8 attenuates
[28] Renner O, Lutjohann D, Richter D, et al. Role of the ABCG8 19H risk allele in choles- diet-induced hypercholesterolemia and atherosclerosis in Ldlr−/− mice. J Lipid Res
terol absorption and gallstone disease. BMC Gastroenterol 2013;13:1–11. 2004;45:1429–36.
[29] Wei KK, Zhang LR, Zhang Y, et al. Interactions between CYP7A1 A-204C and ABCG8 [58] Wei J, Ching LC, Zhao JF, et al. Essential role of transient receptor potential vanilloid
C1199A polymorphisms on lipid lowering with atorvastatin. J Clin Pharm Ther type 1 in evodiamine-mediated protection against atherosclerosis. Acta Physiol
2011;36:725–33. (Oxf) 2013;207:299–307.
[30] Miwa K, Inazu A, Kobayashi J, et al. ATP-binding cassette transporter G8 M429V [59] Li X, Schulte P, Godin DV, et al. Differential mRNA expression of seven genes in-
polymorphism as a novel genetic marker of higher cholesterol absorption in hyper- volved in cholesterol metabolism and transport in the liver of atherosclerosis-
cholesterolaemic Japanese subjects. Clin Sci (Lond) 2005;109:183–8. susceptible and -resistant Japanese quail strains. Genet Sel Evol 2012;44:20.
[31] Koeijvoets KC, van der Net JB, Dallinga-Thie GM, et al. ABCG8 gene polymorphisms, [60] McGillicuddy FC, de la Llera Moya M, Hinkle CC, et al. Inflammation impairs reverse
plasma cholesterol concentrations, and risk of cardiovascular disease in familial hy- cholesterol transport in vivo. Circulation 2009;119:1135–45.
percholesterolemia. Atherosclerosis 2009;204:453–8. [61] Ikewaki K, Nakada Y, Inoue Y. Sitosterolemia. Nihon Rinsho 2007;65:371–4.
[32] Garcia-Rios A, Perez-Martinez P, Fuentes F, et al. Genetic variations at ABCG5/G8 [62] Xu J, Zhou X, Deng Q, et al. Rapeseed oil fortified with micronutrients reduces
genes modulate plasma lipids concentrations in patients with familial hypercholes- atherosclerosis risk factors in rats fed a high-fat diet. Lipids Health Dis
terolemia. Atherosclerosis 2010;210:486–92. 2011;10:96.
[33] Caamano JM, Pacheco A, Lanas F, et al. Single nucleotide polymorphisms in ABCG5 [63] Horenstein RB, Mitchell BD, Post WS, et al. The ABCG8 G574R variant, serum plant
and ABCG8 genes in Chilean subjects with polygenic hypercholesterolemia and con- sterol levels, and cardiovascular disease risk in the Old Order Amish. Arterioscler
trols. Clin Chem Lab Med 2008;46:1581–5. Thromb Vasc Biol 2013;33:413–9.
[34] Yoon JH, Choi HS, Jun DW, et al. ATP-binding cassette sterol transporters are dif- [64] Wilund KR, Yu L, Xu F, et al. No association between plasma levels of plant ste-
ferentially expressed in normal and diseased human gallbladder. Dig Dis Sci rols and atherosclerosis in mice and men. Arterioscler Thromb Vasc Biol
2013;58:431–9. 2004;24:2326–32.
88 X.-H. Yu et al. / Clinica Chimica Acta 428 (2014) 82–88

[65] Singh V, Jain M, Misra A, et al. Curcuma oil ameliorates hyperlipidaemia and associ- [68] Wu JE, Basso F, Shamburek RD, et al. Hepatic ABCG5 and ABCG8 overexpression in-
ated deleterious effects in golden Syrian hamsters. Br J Nutr 2013;110:437–46. creases hepatobiliary sterol transport but does not alter aortic atherosclerosis in
[66] Ji W, Guo L, Lian J, et al. Hypolipidaemic mechanisms of action of CM108 (a flavone transgenic mice. J Biol Chem 2004;279:22913–25.
derivative) in hyperlipidaemic rats. J Pharm Pharmacol 2008;60:1207–12. [69] Basso F, Freeman LA, Ko C, et al. Hepatic ABCG5/G8 overexpression reduces apoB-
[67] Ji W, Gong BQ. Hypolipidemic effects and mechanisms of Panax notoginseng on lipid lipoproteins and atherosclerosis when cholesterol absorption is inhibited. J Lipid
profile in hyperlipidemic rats. J Ethnopharmacol 2007;113:318–24. Res 2007;48:114–26.

You might also like