Sie sind auf Seite 1von 6

ISSN 1672-9145

Acta Biochimica et Biophysica Sinica 2006, 38(3): 151156

CN 31-1940/Q

Minireview

Human Acyl-CoA:cholesterol Acyltransferase (ACAT) and its Potential as a


Target for Pharmaceutical Intervention against Atherosclerosis
1

Catherine CHANG *, Ruhong DONG , Akira MIYAZAKI , Naomi SAKASHITA , Yi ZHANG , Jay LIU ,
1
4
1
Michael GUO , Bo-Liang LI , and Ta-Yuan CHANG
1

Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire 03755 USA;
2
Department of Biochemistry, Showa University School of Medicine, Tokyo, Japan;
Second Department of Pathology, Kumamoto University School of Medicine, Kumamoto, Japan;
4
State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences,
Chinese Academy of Sciences, Shanghai 200031, China
3

Key words

acyl-CoA:cholesterol acyltransferase; cholesterol; cholesteryl ester; atherosclerosis

Acyl-CoA:cholesterol acyltransferase 1
Two acyl-CoA:cholesterol acyltransferase (ACAT) genes
have been identified in mammals: ACAT1 and ACAT2. The
ACAT1 gene was first identified in 1993 by virtue of its
ability to functionally complement the ACAT deficiency of
a Chinese hamster ovary (CHO) cell mutant [9]. Using
ACAT1-specific antibodies, Miyazaki et al. [10] examined
human atherosclerotic lesions using immunohistochemical
staining. These results showed that in early lesions of the
human aorta, mononuclear cells expressed only limited
ACAT1. In contrast, the same ACAT1 antibodies stained
much more intensely in fatty streak lesions, particularly in
areas that contained macrophages with foamy transformation (Fig. 1). This demonstrates that ACAT1 is a marker
for the early development of human atherosclerosis. To
determine the cell types in this region, in the same study
Received: January 9, 2006
Accepted: February 7, 2006
*Corresponding author: E-mail, Catherine.Chang@dartmouth.edu

Miyazaki et al. used Oil-Red-O to identify the lipid-rich


region, followed by double immunostaining (Fig. 2). The
results demonstrated that monocytes/macrophages were
the major cellular component of the ACAT1-expressing
cells in atherosclerotic lesions [10]. These findings have
potential clinical significance because it is known that plaque
ruptures associated with acute myocardial infarction generally
occur in the plaques shoulder regions that are filled with
lipid-rich, foamy macrophages [11,12].
Human ACAT1 (hACAT1) expressed in CHO cells or in
insect H5 cells has been purified to homogeneity with
retention of catalytic activity [13]. hACAT1 is a homotetrameric integral membrane protein [14] and mainly
resides in the endoplasmic reticulum [15,16]. Unlike many
enzymes involved in cholesterol metabolism, ACAT1 is not
regulated by cholesterol at the transcriptional level. The
main mode of sterol-specific regulation of ACAT1 is at
the post-translational level, involving allosteric activation
by its own substrate, cholesterol [13]. Kinetic analysis
DOI: 10.1111/j.1745-7270.2006.00154.x

Institute of Biochemistry and Cell Biology, SIBS, CAS

Downloaded from http://abbs.oxfordjournals.org/ by guest on February 23, 2015

Abstract
Acyl-CoA:cholesterol acyltransferase (ACAT) catalyzes the formation of cholesteryl esters
from cholesterol and long-chain fatty-acyl-coenzyme A. At the single-cell level, ACAT serves as a regulator
of intracellular cholesterol homeostasis. In addition, ACAT supplies cholesteryl esters for lipoprotein
assembly in the liver and small intestine. Under pathological conditions, the accumulation of cholesteryl
esters produced by ACAT in macrophages contributes to foam cell formation, a hallmark of the early
stage of atherosclerosis. Several reviews addressing various aspects of ACAT and ACAT inhibitors are
available [18]. This review briefly outlines the current knowledge on the biochemical properties of human
ACATs, and then focuses on discussing the merit of ACAT as a drug target for pharmaceutical interventions
against atherosclerosis.

152

Acta Biochim Biophys Sin

Vol. 38, No. 3

(A) Diffuse intimal thickening. (B) Fatty streak. Clear immunoreactivity for antiACAT1 antibodies (DM10) is recognized in both endothelial layer and a small
amount of infiltrated mononuclear cells in the early lesion of atherosclerosis (A). In
the fatty streak lesions, prominent ACAT1 expression is found in the infiltrated
macrophages with or without foam cell transformation (B). Bar=100 m. From
Miyazaki et al. [10].

comparing various oxysterols to cholesterol as a substrate


and as an activator revealed that ACAT1 might contain two
types of sterol binding sites. The first site is the activator
site, which strongly prefers cholesterol to oxysterols or
any other sterol analogs examined. Activation of ACAT1
by cholesterol causes the second site, the substrate-binding
site, to accommodate a variety of sterols, including
cholesterol, oxysterols, plant sterols, and various other
sterols as the enzymatic substrate and facilitates enzyme
catalysis [17]. The mode of activation was further analyzed
by using a specific cholesterol analog called enantiomeric
cholesterol. Enantiomeric cholesterol is the mirror image
of cholesterol. In membranes, it possesses identical
biophysical properties as cholesterol [18]. The results
showed that activation of ACAT1 occurs with cholesterol,
but not with enantiomeric cholesterol [19]. These results
indicate that the activation mechanism mainly involves
stereo-specific interaction, rather than biophysical effects
of cholesterol on phospholipid membranes.
In hepatocytes, ACAT1 can provide cholesteryl esters
(CEs) for very low-density lipoprotein (VLDL) assembly
[20,21]. In macrophages and other cell types, ACAT1 is
involved in forming CEs as lipid droplets [10,16]. The VLDL
assembly process occurs within the lumens of the intracellular membrane compartment(s), while the lipid droplets

Fig. 2
Histochemical demonstration of lipid deposition and
immunohistochemical localization of ACAT1 within monocytesmacrophages in the human atherosclerotic plaque
(A) Oil-Red-O staining (red). Numerous lipid droplets have accumulated in the
atheromatous plaque shoulder (the central portion). The necrotic center (the cellular area on the right side) contains a limited number of lipid droplets. (B) Double
immunohistochemical staining with anti-ACAT1 antibodies DM10 (brown) and
anti-monocyte-macrophage antibodies EBM11 (blue). DM10 and EBM11 doublepositive cells are demonstrated in the atheromatous plaque shoulder. (C) Highmagnification view of panel (B). The scale bars in (A) and (B) indicate 200 m; in
(C) the bar indicates 100 m. From Miyazaki et al. [10].

are formed in the cell cytoplasm. For ACAT1 to fulfill its


dual roles, it has been hypothesized that the active site of
ACAT1 may be located within the plane(s) of the ER
membrane, such that the enzyme biosynthesizes CEs within
the ER membrane [22]. This arrangement would enable the
CE formed to leave the cytoplasmic leaflet of the membrane
to form lipid droplets or to be recruited to the lumen of the
membrane by the protein microsomal lipid transfer protein
(MTP) for the VLDL assembly process. The recent

Institute of Biochemistry and Cell Biology, SIBS, CAS

Downloaded from http://abbs.oxfordjournals.org/ by guest on February 23, 2015

Fig. 1
Immunohistochemical detection of ACAT1 in the
human atherosclerotic intima

Mar., 2006

Catherine CHANG et al.: Human Acyl-CoA:cholesterol Acyltransferase (ACAT) and its Potential as a Target

153

evidence reported by Guo et al. [23] demonstrating that


ACAT1 contains 9 transmembrane domains, with the active
site H460 located within transmembrane domain #7,
provided the experimental support for this hypothesis.

ACAT2

Downloaded from http://abbs.oxfordjournals.org/ by guest on February 23, 2015

In 1998 the second ACAT gene, designated as ACAT2,


was simultaneously identified in monkeys, mice and
humans [2426]. ACAT2 shares high homology with
ACAT1 near the C-terminus, but not near the N-terminus.
The kinetic properties of ACAT2 are quite similar to those
of ACAT1 [19,21]. However, membrane topology study
has shown that human ACAT2 contains only two detectable
transmembrane domains [27]. Its active site, H432 (the
equivalent of H460 in hACAT1) may also be located within
the plane of the membrane lipid bilayer [27]. Tissue
distribution studies have shown that the expression of
ACAT1 mRNA and protein is nearly ubiquitous in the human
body, whereas the expression of ACAT2 is more restricted.
Both ACAT1 and ACAT2 are present in human fetal and
adult jejunum samples, with ACAT2 preferentially located
at the apical half of the villi, whereas ACAT1 is uniformly
distributed along the villus-crypt axis. The real-time PCR
analyses showed that in normal human intestines, Acat2
mRNAs are approximate three-fold in abundance over
those of Acat1 [28]. Further experiments using
histochemical staining, Western blot analysis, and
immunodepletion have shown that in the normal (i.e.,
nondiseased) adult livers, ACAT1 is the major isoenzyme
and is present in both Kupffer cells and hepatocytes [16,21].
In contrast, the ACAT2 signal is barely detectable in either
human hepatocytes or Kupffer cells. A representative result
comparing the expression levels of ACAT1 and ACAT2 in
human liver sample is shown in Fig. 3. These findings are
supported by the real-time PCR analyses, which showed
that in normal human livers the abundance of Acat1
mRNAs predominates over those of Acat2 by approximately nine-fold [28]. However, a different study showed
that the immunoreactive ACAT2 protein signals are amply
present in human hepatocytes isolated from patients
affected with gallstone disease [29]. Thus, the relative
functional importance of ACAT1 and ACAT2 in human
hepatocytes is not clear at present and requires further
investigation.
More recently, using Western blot, histochemical staining,
and RT-PCR analyses, Sakashita et al. demonstrated that
immature macrophages expressed only ACAT1, but fully
differentiated macrophages expressed both ACAT1 and

Fig. 3
Immunohistochemical staining using specific antibodies against ACAT1 (A), ACAT2 (B), and Kupffer cells (C) in
the same human liver sample
ACAT1 signal (brown) is most prominent in Kupffer cells, followed by hepatocytes.
In contrast, no positive signal is detected in Kupffer cells or in hepatocytes by
using ACAT2 antibodies. Anti-CD68 stain Kupffer cells (brown) can be visualized
in (C). Nuclei stains are shown in green color. From Sakashita (unpublished data).
Bar=100 m.

ACAT2. In addition, ACAT2 is also present in macrophages


of human atherosclerotic lesions [30]. Thus, the tissue

http://www.abbs.info; www.blackwellpublishing.com/abbs

154

Acta Biochim Biophys Sin

distribution of human ACAT2 is not as restricted as previously


believed. The limited data available suggest that within the
same cell, ACAT1 expression may be constitutive, while
that of ACAT2 may be inducible.

ACAT inhibitors

[37] and Yagyu et al. [38] crossed ACAT1-deficient


(ACAT1-/-) mice with either apoE- (apoE-/-) or LDL
receptor-deficient (LDLR-/-) mice. These double-knockout
mice developed extensive deposition of free cholesterol in
the skin and brain [37,38]. Fazio et al. used LDL receptordeficient (LDLR-/-) mice reconstituted with ACAT1-/donor bone marrow, which led to the accumulation of free
cholesterol in the mouse artery wall. As a result, these mice
developed larger atherosclerotic lesions than the control
LDLR-/- mice did [39]. From these results, the authors
suggest that inhibition of ACAT1 may promote lesion
development. However, data from mouse knockout models
may not always be relevant to human pharmaceutical
applications. ACAT1-/- mice show a complete absence of
ACAT1 enzyme, a condition that does not occur with
pharmaceutical interventions on ACAT1 in animals or in
human patients. Therefore, the mouse knockout model is
probably most appropriate for demonstrating the effects
of drug overdoses that may cause detrimental effects in
animals or in humans. To investigate the efficacy of partial
ACAT inhibition, Kusunoki et al. [40] treated apoEknockout mice with a low-dose ACAT inhibitor, which
resulted in diminution of arterial lesion size, an increase of
smooth muscle cells, and a more stable appearance of the
shoulder area [40]. These results suggest that partial
inhibition of ACAT could be an effective therapeutic
treatment for atherosclerosis.

Fig. 4
Photomicrographs showing the size of rabbit iliac-femoral lesions and the distribution and abundance of esterase-positive
monocyte-macrophages within the lesion in control (A) and the ACAT inhibitor CI976-treated (B) animals
Note the reduced lesion size and relative paucity of monocyte-macrophages within the CI-976-treated animals. Alpha-Naphthol esterase stain. Magnification, 25 . From
Bocan et al. [36].

Institute of Biochemistry and Cell Biology, SIBS, CAS

Downloaded from http://abbs.oxfordjournals.org/ by guest on February 23, 2015

Starting in the 1980s, small molecules that serve as


specific ACAT inhibitors have become available. In cell
culture, ACAT inhibitors have been shown to reduce cellular
cholesteryl ester content [31,32]. In macrophages and
cholesteryl ester-rich hepatoma cell lines, the cholesteryl
ester-reducing effect of ACAT inhibitors is generally attributed not only to direct inhibition of ACAT, but also to their
ability to increase cholesterol efflux to various cholesterol
acceptors present in the growth medium [3335]. In 1991,
Bocan et al. [36] showed that feeding low-dose ACAT
inhibitor CI-976 to cholesterol-fed rabbits reduced the size
of foam cell lesions without affecting plasma cholesterol
levels (Fig. 4). This study suggests that at low dose, the
primary action of the ACAT inhibitor occurs at the foam cell,
not at the liver. It also provides the first in vivo evidence that
ACAT inhibitors may possess an anti-atherosclerosis effect
without significantly affecting total plasma cholesterol level.
However, other laboratories using ACAT1 gene knockout
mouse models obtained disappointing results. Accad et al.

Vol. 38, No. 3

Mar., 2006

Catherine CHANG et al.: Human Acyl-CoA:cholesterol Acyltransferase (ACAT) and its Potential as a Target

tion of drugs concurrently administered. If combination


therapy is to be tested, care must be exercised to avoid
drug-drug interactions.

Acknowledgements
We thank Ellen CHANG and Kathy SAVAGE for critical
reading of this article.

References
1 Chang TY, Chang CC, Cheng D. Acyl-coenzyme A:cholesterol acyltransferase.
Annu Rev Biochem 1997, 66: 613638
2 Sturley SL. Molecular aspects of intracellular sterol esterification: The acyl
coenzyme A:cholesterol acyltransferase reaction. Curr Opin Lipidol 1997, 8:
167173
3 Buhman KF, Accad M, Farese RV. Mammalian acyl-CoA:cholesterol
acyltransferases. Biochim Biophys Acta 2000, 1529: 142154
4 Rudel LL, Lee RG, Cockman TL. Acyl coenzyme A:cholesterol acyltransferase
types 1 and 2: Structure and function in atherosclerosis. Curr Opin Lipidol
2001, 12: 121127
5 Chang TY, Chang CC, Lin S, Yu C, Li BL, Miyazaki A. Roles of acylcoenzyme A:cholesterol acyltransferase-1 and -2. Curr Opin Lipidol 2001, 12:
289296
6 Alegret M, Llaverias G, Silvestre JS. Acyl coenzyme A:cholesterol
acyltransferase inhibitors as hypolipidemic and antiatherosclerotic drugs.
Methods Find Exp Clin Pharmacol 2004, 26: 563586
7 Leon C, Hill JS, Wasan KM. Potential role of acyl-coenzyme A:cholesterol
transferase (ACAT) inhibitors as hypolipidemic and antiatherosclerosis drugs.
Pharm Res 2005, 22: 15781588
8 Li BL, Chang TY, Chen J, Chang CC, Zhao XN. Human ACAT1 gene
expression and its involvement in the development of atherosclerosis. Future
Cardiology 2006, 2: 9399
9 Chang CC, Huh HY, Cadigan KM, Chang TY. Molecular cloning and
functional expression of human acyl-coenzyme A:cholesterol acyltransferase
cDNA in mutant Chinese hamster ovary cells. J Biol Chem 1993, 268:
2074720755
10 Miyazaki A, Sakashita N, Lee O, Takahashi K, Horiuchi S, Hakamata H,
Morganelli PM et al. Expression of ACAT-1 protein in human atherosclerotic
lesions and cultured human monocytes-macrophages. Arterioscler Thromb
Vasc Biol 1998, 18: 15681574
11 Falk E, Shah P, Fuster V. Coronary plaque disruption. Circulation 1995, 92:
657671
12 Libby P, Aikawa M. Stabilization of atherosclerotic plaques: New mechanisms
and clinical targets. Nat Med 2002, 11: 12571262
13 Chang CC, Lee CY, Chang ET, Cruz JC, Levesque MC, Chang TY.
Recombinant human acyl-CoA:cholesterol acyltransferase-1 (ACAT-1) purified
to essential homogeneity utilizes cholesterol in mixed micelles or vesicles in
a highly cooperative manner. J Biol Chem 1998, 273: 3513235141
14 Yu C, Chen J, Lin S, Liu J, Chang CC, Chang TY. Human acyl-CoA:
cholesterol acyltransferase-1 is a homotetrameric enzyme in intact cells and in
vitro. J Biol Chem 1999, 274: 3613936145
15 Chang CC, Chen J, Thomas MA, Cheng D, Del Priore VA, Newton RS, Pape
ME et al. Regulation and immunolocalization of acyl-coenzyme A:cholesterol
acyltransferase in mammalian cells as studied with specific antibodies. J Biol
Chem 1995, 270: 2953229540

http://www.abbs.info; www.blackwellpublishing.com/abbs

Downloaded from http://abbs.oxfordjournals.org/ by guest on February 23, 2015

One early concern in developing ACAT inhibitors was


adrenal toxicity. The cause for adrenal toxicity is probably
caused by a certain chemical structure, which is unrelated
to the ability of the compound to inhibit ACAT. Recent
advances in pharmaceutical research, this concern seems to
have been eliminated [41]. Another major concern for using
the ACAT inhibitor is cellular toxicity induced by free
cholesterol loading. By adding ACAT inhibitors to lipidladen mouse peritoneal macrophages maintained in cell
culture with no cholesterol acceptor present, investigators
observed the crystal formation of free cholesterol inside
the cells, which in turn induced cellular toxicity and leads
to cell death [42,43]. The in vivo significance of these
findings is unclear at present. However, this finding does
raise concern for using ACAT inhibitors for treating
atherosclerotic lesions under conditions where cellular
sterol efflux becomes severely hampered.
Coronary heart disease (CHD) is one of the leading
causes of morbidity and mortality in industrialized countries.
Hypercholesterolemia is one of the major risk factors for
progression of atherosclerosis. The development of HMGCoA reductase inhibitors, i.e., statins, has significantly
reduced the mortality of CHD patients by 20%35%,
saving the lives of millions of people each year [44]. On
the other hand, even with the aid of statins, CHD-related
mortality remains high. There is still a need to identify
other compounds that would complement the action of
statins and might further reduce CHD incidence and
mortality. One of these potential novel agents is the ACAT
inhibitor. Different from the effect of statins, which work
mainly by reducing plasma cholesterol, the ACAT inhibitors
may work by directly reducing the size of the lipid-rich
core in the atherosclerotic plaques, thus stabilizing the
lesion against plaque rupture.
Under normal physiological conditions, ACAT1 is expressed more ubiquitously than the more restricted ACAT2
expression. Recent data raise the possibility that ACAT2
may be inducible in different tissues under various pathological conditions [29,30]. Therefore, before the roles of
ACAT1 and ACAT2 in each tissue/cell type are clarified,
developing a general ACAT inhibitor against both ACAT1
and ACAT2 may be advisable. There are currently several
ACAT inhibitors being tested in clinical trials. We look
forward to learning the results from these ongoing trials,
which will help determine whether ACAT inhibition in
patients with CHD provides promise in the treatment of
atherosclerosis. In this regard, it is worthwhile to cite the
recent work of Sahi et al. [45], who showed that the ACAT
inhibitor Avasimibe induces the cytochrome CYP3A4 in
human hepatocytes, thus promoting the hepatic inactiva-

155

156

Acta Biochim Biophys Sin

Vol. 38, No. 3

16 Sakashita N, Miyazaki A, Takeya M, Horiuchi S, Chang CC, Chang TY,

32 Cadigan KM, Heider JG, Chang TY. Isolation and characterization of

Takahashi K. Localization of human acyl-coenzyme A:cholesterol


acyltransferase-1 (ACAT-1) in macrophages and in various tissues. Am J
Pathol 2000, 156: 227236
17 Zhang Y, Yu C, Liu J, Spencer TA, Chang CC, Chang TY. Cholesterol is

Chinese hamster ovary cell mutants deficient in acyl-coenzyme A:cholesterol


acyltransferase activity. J Biol Chem 1988, 263: 274282
33 Schmitz G, Robenek H, Beuck M, Krause R, Schurek A, Niemann R.

superior to 7-ketocholesterol or 7-alpha-hydroxycholesterol as an allosteric


activator for acyl-coenzyme A:cholesterol acyltransferase 1. J Biol Chem
2003, 278: 1164211647
18 Westover EJ, Covey DF. The enantiomer of cholesterol. J Membrane Biol
2004, 202: 6172
19 Liu J, Chang CC, Westover EJ, Covey DF, Chang TY. Investigating the
allosterism of acyl-CoA:cholesterol acyltransferase (ACAT) by using various
sterols: In vitro and intact cell studies. Biochem J 2005, 391: 389397
20 Spady DK, Willard MN, Meidell RS. Role of acyl-coenzyme A:cholesterol

Immunological quantitation and localization of ACAT-1 and ACAT-2 in


human liver and small intestine. J Biol Chem 2000, 275: 28083 28092
22 Chang TY, Chang CC, Lu X, Lin S. Catalysis of ACAT may be completed
within the plane of the membrane: A working hypothesis. J Lipid Res 2001,
42: 19331938
23 Guo ZY, Lin S, Heinen JA, Chang CC, Chang TY. The active site His460
of human acyl-coenzyme A:cholesterol acyltransferase 1 resides in a hitherto
undisclosed transmembrane domain. J Biol Chem 2005, 280: 3781437826
24 Anderson RA, Joyce C, Davis M, Reagan JW, Clark M, Shelness GS, Rudel
LL. Identification of a form of acyl-CoA:cholesterol acyltransferase specific to
liver and intestine in nonhuman primates. J Biol Chem 1998, 273: 26747
26754
25 Cases S, Novak S, Zheng YW, Myers HM, Lear SR, Sande E, Welch CB et
al. ACAT-2, a second mammalian acyl-CoA:cholesterol acyltransferase. Its
cloning, expression, and characterization. J Biol Chem 1998, 273: 26755
26764
26 Oelkers P, Behari A, Cromley D, Billheimer JT, Sturley SL. Characterization of two human genes encoding acyl coenzyme A:cholesterol acyltransferase
related enzymes. J Biol Chem 1998, 273: 2676526771
27 Lin S, Lu X, Chang CC, Chang TY. Human acyl-coenzyme A:cholesterol
acyltransferase expressed in Chinese hamster ovary cells: Membrane topology
and active site location. Mol Biol Cell 2003, 14: 24472460
28 Smith JL, Rangaraj K, Simpson R, Maclean DJ, Nathanson LK, Stuart KA,
Scott SP et al. Quantitative analysis of the expression of ACAT genes in
human tissues by real-time PCR. J Lipid Res 2004, 45: 686 696.
29 Parini P, Davis M, Lada AT, Erickson SK, Wright TL, Gustafsson U, Sahlin
S et al. ACAT2 is localized to hepatocytes and is the major cholesterolesterifying enzyme in human liver. Circulation 2004, 110: 20172023
30 Sakashita N, Miyazaki A, Chang CC, Chang TY, Kiyota E, Satoh M,
Komohara Y et al. Acyl-coenzyme A:cholesterol acyltransferase 2 (ACAT2) is
induced in monocyte-derived macrophages: In vivo and in vitro studies. Lab
Invest 2003, 83: 15691581
31 Ross AC, Go KJ, Heider JG, Rothblat GH. Selective inhibition of acyl
coenzyme A:cholesterol acyltransferase by compound 58-035. J Biol Chem
1984, 259: 815819

density lipoprotein on esterified cholesterol stores in macrophages and


hepatoma cells. Arteriosclerosis 1990, 10: 135144
35 Rodriguez A, Bachorik PS, Wee SB. Novel effects of the acyl-coenzyme A:
cholesterol acyltransferase inhibitor 58-035 on foam cell development in
primary human monocyte-derived macrophages. Arterioscler Thromb Vasc
Biol 1999, 19: 21992206
36 Bocan TM, Mueller SB, Uhlendorf PD, Newton RS, Krause BR. Comparison
of CI-976, an ACAT inhibitor, and selected lipid-lowering agents for
antiatherosclerotic activity in iliac-femoral and thoracic aortic lesions. A
biochemical, morphological, and morphometric evaluation. Arterioscler
Thromb 1991, 11: 18301843
37 Accad M, Smith SJ, Newland DL, Sanan DA, King LE Jr, Linton MF,
Fazio S et al. Massive xanthomatosis and altered composition of atherosclerotic lesions in hyperlipidemic mice lacking acyl CoA:cholesterol
acyltransferase 1. J Clin Invest 2000, 105: 711719
38 Yagyu H, Kitamine T, Osuga J, Tozawa R, Chen Z, Kaji Y, Oka T et al.
Absence of ACAT-1 attenuates atherosclerosis but causes dry eye and cutaneous
xanthomatosis in mice with congenital hyperlipidemia. J Biol Chem 2000,
275: 2132421330
39 Fazio S, Major AS, Swift LL, Gleaves LA, Accad M, Linton MF, Farese
RV Jr. Increased atherosclerosis in LDL receptor-null mice lacking ACAT1
in macrophages. J Clin Invest 2001, 107: 163 171
40 Kusunoki J, Hansoty DK, Aragane K, Fallon JT, Badimon JJ, Fisher EA.
Acyl-CoA:cholesterol acyltransferase inhibition reduces atherosclerosis in
apolipoprotein E-deficient mice. Circulation 2001, 103: 26042609
41 Junquero D, Oms P, Carilla-Durand E, Autin J, Tarayre J, Degryse A,
Patoiseau J et al. Pharmacological profile of F 12511, (S)-2',3',5'-trimethy4'-hydroxy-alpha-dodecylthioacetanilide, a powerful and systemic acylcoenzyme A:cholesterol acyltransferase inhibitor. Biochem Pharmacol 2001,
61: 97108
42 Warner GJ, Stoudt G, Bamberger M, Johnson WJ, Rothblat GH. Cell
toxicity induced by inhibition of acyl coenzyme A:cholesterol acyltransferase
and accumulation of unesterified cholesterol. J Biol Chem 1995, 270:
57725778
43 Feng B, Yao PM, Li Y, Devlin CM, Zhang D, Harding HP, Sweeney M et
al. The endoplasmic reticulum is the site of cholesterol-induced cytotoxicity in macrophages. Nat Cell Biol 2003, 5: 781 792
44 Scandinavian Simvastatin Survival Study Group. Randomized trial of
cholesterol lowering in 4444 patients with coronary heart disease: The
Scandinavian simvastatin survival study (4S). Lancet 1994, 344: 1383
1389
45 Sahi J, Milad MA, Zheng X, Rose KA, Wang H, Stilgenbauer L, Gilbert
D et al. Avasimibe induces CYP3A4 and multiple drug resistance protein
1 gene expression through activation of the pregnane X receptor. J Pharmacol
Exp Ther 2003, 306: 10271034

Edited by
Ming-Hua XU

Institute of Biochemistry and Cell Biology, SIBS, CAS

Downloaded from http://abbs.oxfordjournals.org/ by guest on February 23, 2015

acyltransferase-1 in the control of hepatic very low density lipoprotein secretion and low density lipoprotein receptor expression in the mouse and hamster.
J Biol Chem 2000 275: 2700527012
21 Chang CC, Sakashita N, Ornvold K, Lee O, Chang ET, Dong R, Lin S et al.

Ca++ antagonists and ACAT inhibitors promote cholesterol efflux from


macrophages by different mechanisms. I. Characterization of cellular lipid
metabolism. Arteriosclerosis 1988, 8: 46 56
34 Bernard DW, Rodriguez A, Rothblat GH, Glick JM. Influence of high

Das könnte Ihnen auch gefallen