Вы находитесь на странице: 1из 16

High Density Lipoproteins and Arteriosclerosis

Role of Cholesterol Efflux and Reverse Cholesterol Transport


Arnold von Eckardstein, Jerzy-Roch Nofer, Gerd Assmann

Abstract—High density lipoprotein (HDL) cholesterol is an important risk factor for coronary heart disease, and HDL
exerts various potentially antiatherogenic properties, including the mediation of reverse transport of cholesterol from
cells of the arterial wall to the liver and steroidogenic organs. Enhancement of cholesterol efflux and of reverse
cholesterol transport (RCT) is considered an important target for antiatherosclerotic drug therapy. Levels and
composition of HDL subclasses in plasma are regulated by many factors, including apolipoproteins, lipolytic enzymes,
lipid transfer proteins, receptors, and cellular transporters. In vitro experiments as well as genetic family and population
studies and investigation of transgenic animal models have revealed that HDL cholesterol plasma levels do not
necessarily reflect the efficacy and antiatherogenicity of RCT. Instead, the concentration of HDL subclasses, the
mobilization of cellular lipids for efflux, and the kinetics of HDL metabolism are important determinants of RCT and
the risk of atherosclerosis. (Arterioscler Thromb Vasc Biol. 2001;21:13-27.)
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

Key Words: apolipoproteins 䡲 lipid transfer proteins 䡲 lipases 䡲 ABC transporter 䡲 Tangier disease

A low level of HDL cholesterol is an important cardiovas-


cular risk factor.1 Moreover, HDLs exert various poten-
tially antiatherogenic properties.2– 4 As a consequence, thera-
than 40% of patients with myocardial infarction have low
HDL-C as a cardiovascular risk factor.7 In the prospective
and multicentric European Concerted Action on Thrombosis
peutic modifications of HDL cholesterol (HDL-C) levels and Disabilities (ECAT) Angina Pectoris Study, we identified
have attracted considerable interest. Drugs increasing HDL-C low HDL-C and low apoA-I as the most important biochem-
are sought for antiatherogenic therapies, and drugs decreasing ical risk factors for coronary events in patients with angio-
HDL-C are suspected to increase cardiovascular risk.4 graphically assessed CHD.8 By convention, the risk threshold
Reverse cholesterol transport (RCT) describes the metab- value of HDL-C has been defined as 35 mg/dL (0.9 mmol/L)
olism and an important antiatherogenic function of HDL, in men and 45 mg/dL (1.15 mmol/L) in women.9 –11 Because
namely, the HDL-mediated efflux of cholesterol from non- of interaction, the strength of the association between HDL-C
hepatic cells and its subsequent delivery to the liver and and cardiovascular risk depends on the presence of additional
steroidogenic organs, in which it is used for the synthesis of risk factors. Therefore, threshold values are higher in men
lipoproteins, bile acids, vitamin D, and steroid hormones.2– 4 with diabetes mellitus or hypercholesterolemia or in the
Approximately 9 mg cholesterol per kilogram body weight is presence of multiple risk factors.4 Low HDL-C has been
synthesized by peripheral tissues every day and must be identified as the most frequent familial dyslipoproteinemia in
moved to the liver for effective catabolism.5 Distortion of patients with premature myocardial infarction.12 Finally, in
RCT can favor the deposition of cholesterol within the the Helsinki Heart Study13 and the High-Density-Lipoprotein
arterial wall and thereby contribute to the development of Cholesterol Intervention Trial of the Department of Veterans
arteriosclerosis. This ⬎30-year-old concept of John Glomset6 Affairs (VA-HIT) study,14 increases of HDL-C on treatment
has been much revived only recently by the identification of with gemfibrozil were correlated with the prevention of CHD
several important players in this metabolic pathway, which is events. Thus, HDL-C has become an important component of
reviewed in the present article. Because of the limitation of algorithms to assess the global cardiovascular risk of patients
space, additional potentially antiatherogenic functions of and also a target for therapeutic intervention and for the
HDL will be reviewed by us in detail elsewhere. definition of treatment goals.9,10 However, in contrast to LDL
cholesterol and smoking, it is as yet unknown whether the
Epidemiological Background epidemiological association between HDL-C and CHD is
Numerous clinical and epidemiological studies have demon- causal.
strated the inverse and independent association between Several arguments against a causal relationship must be
HDL-C and the risk of coronary heart disease (CHD).1 More taken into account: First, the association between HDL-C and

Received May 22, 2000; revision accepted October 18, 2000.


From the Institut für Klinische Chemie und Laboratoriumsmedizin (A.v.E., J.-R.N., G.A.), Zentrallaboratorium, Westfälische Wilhelms-Universität
Münster, and the Institut für Arterioskleroseforschung an der Universität Münster (A.v.E., J.-R.N., G.A.), Münster, Germany.
Correspondence to Dr Arnold von Eckardstein, Institut für Klinische Chemie und Laboratoriumsmedizin, Zentrallaboratorium, Westfälische
Wilhelms-Universität Münster, Albert-Schweitzer-Strasse 33, D-48129 Münster, Germany. E-mail vonecka@uni-muenster.de
© 2001 American Heart Association, Inc.
Arterioscler Thromb Vasc Biol. is available at http://www.atvbaha.org

13
14 Arterioscler Thromb Vasc Biol. January 2001

cardiovascular mortality is U-shaped rather than linear; ie, duced changes in HDL-C may not necessarily lead to ex-
cardiovascular death rates are higher in individuals with very pected changes in clinical outcomes. Despite this skepticism
high levels of HDL-C (eg, fifth quintile) than in individuals toward HDL-C as a suitable intermediary phenotype for the
with intermediate levels of HDL-C (eg, third quintile)15 monitoring of the cardiovascular effects of therapeutic inter-
(Figure I, available online at http://atvb.ahajournals.org). ventions, it is still believed that modulation of HDL metab-
Second, in ecological studies, considerable differences in olism and the RCT system might be an important target for
HDL-C between ethnic populations did not explain the antiatherosclerotic drug therapy.
differences in cardiovascular morbidity and mortality (eg,
differences between Germany and Israel).16 Third, in some HDL Subclasses
but not all populations (eg, in Germans but not in Turks),17 HDL encompasses a heterogeneous class of lipoproteins,
low HDL-C is frequently found as a component of the which have in common a high density (⬎1.063 g/mL) and a
metabolic syndrome, ie, together with hypertriglyceridemia, small size (Stoke’s diameter 5 to 17 nm).25 The majority of
small dense LDL, glucose intolerance or overt diabetes the HDL particles contain apoA-I. Differences in the quanti-
mellitus, hypertension, overweight or obesity, and hyperin- tative and qualitative content of lipids, apolipoproteins, en-
sulinemia. Insulin resistance is considered the common soil of zymes, and lipid transfer proteins (Table 1) result in the
these cardiovascular risk factors. As a consequence, insulin presence of various HDL subclasses, which are characterized
resistance and/or the proatherogenic components of the met- by differences in shape, density, size, charge, and antigenic-
abolic syndrome rather than an antiatherogenic function of ity.25 After agarose gel electrophoresis of plasma and anti–
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

HDL may underlie the inverse association between HDL-C apoA-I immunoblotting, the majority of apoA-I is present in
and cardiovascular risk.18 The coincidence of low HDL-C a fraction, which migrates with ␣-electrophoretic mobility
with many additional risk factors puts patients with the and is designated ␣-LpA-I. This fraction contains eventually
metabolic syndrome at high global risk for coronary events. all of the cholesterol, which is quantified in the routine
This is in contrast to individuals who have low HDL-C as an laboratory as HDL-C. About 5% to 15% of apoA-I in human
isolated risk factor.4,11 Fourth, as a negative acute-phase plasma is associated with particles, which have electro-
reactant, low HDL-C can serve as a surrogate marker for a phoretic pre-␤ mobility and which can be further distin-
systemic inflammation, which causes or aggravates athero- guished by subsequent polyacrylamide gradient gel electro-
sclerosis (eg, smoking or chronic infections), or as a disease phoresis into pre-␤1-LpA-I, pre-␤2-LpA-I, and pre-␤3-LpA-I
marker for local inflammation, especially in unstable (Figure III, available online at http://atvb.ahajournals.org).
plaques.19 In support of the importance of this association, Pre-␤1-LpA-I is the smallest particle, discoidal in shape, and
low HDL-C is a more powerful risk factor in short-term contains apoA-I either as a lipid-free apolipoprotein or in
follow-ups than in long-term follow-ups of prospective epi- association with a few molecules of sphingomyelin and
demiological studies. For example, the relative risk of CHD phosphatidylcholine.26,27 Similar lipid-poor particles contain
events associated with HDL-C ⬍35 mg/dL (⬍0.9 mmol/L) only apoE (␥-LpE) or apoA-IV (LpA-IV) as their only
was 6.1 in a 2-year follow-up of the Prospective Cardiovas- apolipoproteins.28 –30 It is also important to note that relative
cular Münster (PROCAM) study but only 2.1 to 2.7 in longer to the concentration of lipid-rich ␣-HDL, the concentration of
follow-ups (Figure II, available online at http://atvb.ahajour- these lipid-poor particles is increased in extravasal compart-
nals.org). In the ECAT Angina Pectoris Study, HDL choles- ments including the lymph, where RCT is initiated in
terol had no association with coronary events when plasma vivo.27,31,32 ␣-HDL can be further differentiated by ultracen-
levels of C-reactive protein or fibrinogen, which are positive trifugation according to density (HDL2 and HDL3), by non-
acute-phase reactants, were low.8 Also, in the PROCAM denaturing polyacrylamide gradient gel electrophoresis ac-
study, HDL-C had a significant association with cardiovas- cording to size, or by immunoaffinity chromatography
cular and overall mortality in smokers but not in nonsmokers according to apolipoprotein composition (eg, LpA-I/A-II and
(Figure I). Fifth, the elevation of HDL-C with statins, LpA-I).25
fibrates, or estrogens was correlated with the prevention of
CHD events in some trials (ie, Helsinki Heart Study and HDL Metabolism
VA-HIT)13,14 but not in other trials (Bezafibrate Infarction Lipid-rich ␣-HDLs arise from lipid-poor particles or even
Prevention [BIP], Scandinavian Simvastatin Survival Study lipid-free apolipoproteins.25–27,33 These lipid-poor HDL pre-
[4S], Cholesterol and Current Events [CARE], West of cursors are produced either as nascent HDL by hepatocytes34
Scotland Pravastatin Trial [WOSCOP], Air Force/Texas Cor- and the intestinal mucosa,35 or they dissociate from chylomi-
onary Atherosclerosis Prevention Study [AFCAPS/Tex- crons and VLDL during lipoprotein lipase–mediated hydro-
CAPS], and the Heart and Estrogen/Progestin Replacement lysis of triglycerides36 or are generated by the interconversion
Study [HERS]).20 –24 Moreover, most of these interventions of HDL2 and HDL3 by cholesteryl ester transfer protein
have only moderate effects on HDL-C levels, and none of (CETP),37,38 phospholipid transfer protein (PLTP),39,40 and
them is specific for HDL, but they exert a broad scope of hepatic lipase (HL)41 (Figure 1, Table 1). Lipid-free apoli-
mostly antiatherogenic but sometimes even proatherogenic poproteins or lipid-poor particles acquire phospholipids and
effects on lipid metabolism, the hemostatic system, and unesterified cholesterol from hepatic and nonhepatic cells.42
inflammation. It is not known whether this lipidation occurs intracellularly
In summary, the data from observational and interventional or extracellularly or both. On the one hand, lipid-free apoli-
studies demonstrate that the increased coronary risk associ- poproteins were shown to induce phospholipid and choles-
ated with low HDL-C does not necessarily reflect a defective terol efflux from various cells, including hepatocytes and
antiatherogenic repair system and that therapeutically in- macrophages, which suggests extracellular assembly.43,44 On
von Eckardstein et al HDLs and Arteriosclerosis 15

selective uptake of lipids by scavenger receptor B1 (SR-BI)


and the holoparticle uptake by apoE or apoA-I receptors,
respectively, and 2 indirect pathways, which involve the
action of CETP, HL, and endothelial lipase (EL) (Figure 1,
Table 1).61– 67 (For details see below.)
The removal of lipids from HDL2 by SR-BI, CETP, and
HL, the subsequent conversion of HDL2 into HDL3, and the
conversion of HDL3 into HDL2 by PLTP regenerate pre-␤1-
LpA-I or lipid-free apoA-I.26,33,37– 40 These small apolipopro-
teins or particles can leave the plasma into the extravascular
space,25–27 where they serve as acceptors of cellular lipids and
thus again initiate the generation of HDL. In the kidney, these
small particles are filtered and removed from the plasma. The
reuptake of apoA-I from the proximal tubulus lumen is
Figure 1. Pathways involved in the generation and conversion
of HDL. Mature HDL3 and HDL2 are generated from lipid-free mediated by cubilin68 –70 (Figure 1, Table 1).
apoA-I or lipid-poor pre-␤1-HDL as the precursors. These pre-
cursors are produced as nascent HDL by the liver or intestine or Cholesterol Efflux
are released from lipolysed VLDL and chylomicrons or by inter-
Cellular Cholesterol Trafficking
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

conversion of HDL3 and HDL2. ABC1-mediated lipid efflux from


cells is important for initial lipidation; LCAT-mediated esterifica- The uptake of modified lipoproteins by macrophages of the
tion of cholesterol generates spherical particles that continue to vascular wall plays an important role in the pathogenesis of
grow on ongoing cholesterol esterification and PLTP-mediated
particle fusion and surface remnant transfer. Larger HDL2 parti- atherosclerosis because accumulation of lipids turns them
cles are converted into smaller HDL3 particles on CETP- into activated foam cells, which produce various growth
mediated export of cholesteryl esters from HDL onto apoB- factors, cytokines, and proteases and thereby influence the
containing lipoproteins, on SR-BI–mediated selective uptake of
course of atherosclerosis. Macrophages internalize modified
cholesteryl esters into liver and steroidogenic organs, and on
HL- and EL-mediated hydrolysis of phospholipids. HDL lipids lipoproteins (especially by type A scavenger receptors) and
are catabolized either separately from HDL proteins (ie, by cell debris (by phagocytosis). Neither process is regulated by
selective uptake or via CETP transfer) or together with HDL pro- cholesterol. In consequence and in contrast to other cells
teins (ie, via uptake through as-yet-unknown HDL receptors or
apoE receptors). The conversion of HDL2 into HDL3 and the (which limit their cholesterol content by a finely tuned,
PLTP-mediated conversion of HDL3 into HDL2 liberated lipid-free sterol-regulated interplay of endogenous cholesterol synthesis
or poorly lipidated apoA-I. A part of lipid-free apoA-I undergoes and LDL receptor–mediated uptake of exogenous lipopro-
glomerular filtration in the kidney and tubular readsorption
through cubilin. For further details, see text and Table 1. Blue
teins), macrophages accumulate lipids. Released from cho-
arrows represent lipid transfer processes, and red arrows repre- lesteryl esters by lysosomal acid lipase, unesterified choles-
sent protein transfer processes. TGRL indicates triglyceride-rich terol is transferred to the endoplasmic reticulum (ER) either
lipoproteins. directly or indirectly via the plasma membrane. In the ER,
cholesterol is esterified by acyl coenzyme A:cholesterol
the other hand, macrophages, hepatocytes, and fibroblasts acyltransferase (ACAT) to protect the cell from the cytotox-
were shown to internalize lipid-free apolipoproteins, HDL, icity of excess unesterified cholesterol.71–73 Cholesteryl esters
and chylomicron remnants and to resecrete lipidated apoli- formed by ACAT appear as cytosolic lipid droplets, which
poproteins.45,46 This process has been termed retroendocyto- give lipid-laden macrophages their foamy appearance. Cyto-
sis and was found to be defective in Tangier disease,47 in solic cholesteryl esters can be hydrolyzed by neutral cho-
which mutations in the ATP binding cassette transporter 1 lesteryl ester hydrolase (NCEH), which is activated by a
(ABC1) interfere with cellular lipid efflux and lead to the cAMP-dependent protein kinase A.74 Cholesterol released by
absence of lipid-rich ␣-HDL from plasma.48 –52 Because NCEH is transferred to the cell membrane, where it can be
ABC1 is expressed in many cells, including hepatocytes and transported to the ER for reesterification by ACAT.71–73 This
enterocytes,53,54 this protein probably plays an important role cycle of cholesterol and cholesteryl esters between ACAT
not only in lipid efflux from peripheral cells but also in the and NCEH is interrupted by the presence of extracellular
hepatic and intestinal birth of HDL. cholesterol acceptors, including HDL, which cause cholester-
Lipid-poor HDL precursors become mature, lipid-rich, and ol efflux.42,71–73,75,76
spherical ␣-LpA-I by the acquisition of phospholipids and The plasma membrane, from which cholesterol is ulti-
unesterified cholesterol from either cells or apoB-containing mately released into the extracellular space, accounts for 60%
lipoproteins, by the lecithin:cholesterol acyltransferase to 90% of unesterified cholesterol in mammalian cells, 95%
(LCAT)-mediated esterification of cholesterol, and by the of which is localized in the cytofacial leaflet of the bilayer
association of additional apolipoproteins.33,55–57 The initial membrane.72,73 The lateral distribution of cholesterol within
products are small HDL3 particles, which on esterification of the plasma membrane is organized into microdomains.77
cholesterol through LCAT and fusion with other HDL3 Coated pits, ie, clathrin-stabilized plasma membrane invagi-
particles through PLTP and PLTP-mediated acceptance of nations that contain lipoprotein receptors, have less choles-
surface remnants of triglyceride-rich lipoproteins grow into terol and sphingolipids than the rest of the plasma membrane.
larger HDL2 particles33,58 – 60 (Figure 1, Table 1). In contrast, caveolae, detergent-resistant plasma membrane
Lipids or proteins of ␣-HDL are removed from the circu- invaginations that are characterized by the presence of caveo-
lation by at least 2 direct pathways, which involve the lins but the absence of clathrin, are enriched in cholesterol
16 Arterioscler Thromb Vasc Biol. January 2001

TABLE 1. Characteristics of Pivotal Genes/Proteins Involved in RCT


Regulation

Gene/Protein Locus Structure* Function in RCT Major Sites of Synthesis Up Down


Apolipoproteins
ApoA-I 11q13-qter 243 Major protein component of HDL Liver, small intestine E2, T3, Testosterone,
activation of LCAT, stimulation of glucocorticoids, TGF-␤, cholic
cholesterol efflux, ligand of HDL IL-6, retinoids, acid
binding sites (eg, SR-BI, ABC1) fibrates
ApoA-II 1q21-qter 77 Major protein component of HDL Liver Retinoids, Thyroid hormone,
ligand of HDL binding site, inhibition fibrates TNF-␣
of HL
ApoA-IV 11q13-qter 376 Activation of LCAT, modulation of LPL, Small intestine Unsaturated fatty Leptin
stimulation of cholesterol efflux acids, bile acids,
peptide YY
ApoC-I 19cen-q13.2 57 Activation of LCAT, inhibition of Liver ? ?
hepatic TGRL uptake
ApoC-II 19cen-q13.2 79 Activation of LPL, inhibition of hepatic Liver ? ?
TGRL uptake
ApoC-III 11q13-qter 79 Inhibition of LPL, inhibition of hepatic Liver Retinoids, Insulin, TNF-␣,
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

TGRL uptake cytokines fibrates


ApoD 3p14.2-qter 169 Formation of pre-␤3-LpA-I Liver, spleen, CNS, Testosterone, Estradiol
small intestine, adrenals, glucocorticoids
placenta
ApoE 19cen-q13.2 299 Ligand of apoE receptors, mobilization Liver, macrophages, Cholesterol, E2 Cytokines
of cellular cholesterol in macrophages CNS
and cholesterol efflux
ApoF 12 326 Inhibition of CETP ? ? ?
Enzymes and lipid transfer proteins
LPL 8p22 475 Hydrolysis of triglycerides in TGRL, Muscle, adipose tissue Vitamin D3, Endothelin-1,
generation of HDL precursors and cAMP, insulin, IL-1, IL-11,
surface remnants glucose, ␣1-AR, TNF-␣, IFN-␥,
␤3-AR, FFAs, LPS, PTH, PGE2
fibrates,
thiazolidinediones
HL 15q15-15q22 499 Hydrolysis of triglycerides and Liver Testosterone E2, growth
phospholipids in HDL, cofactor of hormone,
SR-BI in selective uptake, generation dexamethasone
of lipid-free apoA-I fibrates
EL ? 482 Hydrolysis of phospholipids in HDL Endothelium, liver, TNF-␣, IL-1␤,
macrophages shear stress
LCAT 16q22 440 Cholesterol esterification, maturation Liver, testes, CNS IL-6, TGF-␤
of HDL dexamethasone,
FFAs,
corticotropin
CETP 16q12-16q21 476 Exchange of cholesteryl esters and Liver, small intestine, Cholesterol,
triglycerides between HDL and LpB, spleen, macrophages oxysterols,
conversion of HDL, generation of rexinoids
lipid-free apoA-I
PLTP 20q12-q13.1 493 Transfer of phospholipids (surface Liver, endothelium Fibrates, bile
remnants) from TGRL onto HDL, acids, hypoxia
fusion of small HDL, generation of
lipid-free apoA-I
Cellular receptors and transporters
ABC1 9q31 2201 Mediation of cholesterol and Liver, macrophages, Cholesterol, IFN-␥
phospholipid efflux, maturation of HDL intestine, fetal tissues oxysterols,
(ubiquitous) rexinoids, cAMP
SR-BI 12q24.2-qter 509 Mediation of selective uptake and Liver, adrenals, testes, E2 (Kupffer Cholesterol, E2
cholesterol efflux ovary, macrophages cells), cAMP, (hepatocytes),
gonadotropins, glucocorticoids
corticotrophin,
fibrates, PGF2␣
Cubilin 10p12-10p14 3597 Tubular resorption of HDL and apoA-I Renal tubulus cells, Shear stress
intestinum, yolk sac,
placenta
AR indicates adrenergic receptor; CNS, central nervous system; E2, estradiol; FFAs, free fatty acids; IL, interleukin; IFN-␥, interferon-␥; LPS, lipopolysaccharide,
PGE2, prostaglandin E2; PGF2␣, prostaglandin 2␣; PTH, parathyroid hormone; T3, thyroxin; TGRL, triglyceride-rich lipoproteins; TGF-␤, tissue growth factor-␤; and
TNF-␣, tumor necrosis factor-␣.
*Number of amino acids in the mature protein.
von Eckardstein et al HDLs and Arteriosclerosis 17

the content of cytosolic cholesteryl esters.28,42,75,76,84 – 87 Phos-


pholipid efflux appears to precede cholesterol efflux.76,87 As
a result, apolipoprotein-mediated lipid efflux lipidates apoli-
poproteins and hence produces HDL-like lipoproteins with
electrophoretic pre- ␤ and pre- ␣ mobilities. 43,85
Apolipoprotein-mediated lipid efflux involves unspecific de-
sorption of plasma membrane lipids, ie, microsolubilization,
and specific interactions with plasma membrane proteins.76,88
It is cell specific and takes place only in growth-arrested and
cholesterol-enriched cells.51,89 Lipid-free apolipoproteins re-
move phospholipids and cholesterol from normal human skin
fibroblasts, aortic smooth muscle cells, and macro-
phages28,75,76,83– 87,90,91 but not from erythrocytes, rat or por-
Figure 2. Regulation of cholesterol efflux from cells. Aqueous cine smooth muscle cells, or fibroblasts of patients with
diffusion of unesterified cholesterol (UC) from the plasma mem- Tangier disease.84,91–93 Moreover, lipid-free apolipoprotein-
brane onto lipid-rich lipoproteins, albumin, phospholipid vesi- mediated cholesterol efflux is suppressed by low temperature,
cles, or cyclodextrins is slow. Binding of HDL to SR-BI leads to
reorganization of cholesterol within the plasma membrane and by partial proteolysis of cell membranes, or by interference
facilitates cholesterol efflux. ABC1-mediated efflux of UC and with the regular function of the Golgi apparatus and with
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

phospholipids (PL) onto lipid-free apolipoproteins or lipid-poor intracellular vesicular transport (eg, with monensin or brefel-
particles is fast and involves the translocation of cholesterol din A).83,84 It has been suggested that lipid-free apolipopro-
from intracellular compartments to the plasma membrane. This
transport process appears to involve signal transduction pro- teins and pre-␤1-LpA-I specifically release cholesterol that is
cesses, ie, the cAMP-mediated activation of a protein kinase A located in the caveolae.72,94 Activation of protein kinase C
(PKA) and the degradation of phosphatidylcholine by phospho- (PKC) enhances and inhibition of PKC suppresses
lipase C (PC-PLC) and phospholipase D (PC-PLD). The prod-
ucts DAG and phosphatidic acid (PA) act as second messen-
apolipoprotein-mediated cholesterol efflux.90,95,96 cAMP was
gers, which activate intracellular transporters either directly or also found to increase apolipoprotein-induced cholesterol
indirectly by activation of a PKC. efflux from macrophage cell lines.45,97–99 In RAW264 cells,
this was associated with internalization and resecretion of
and sphingolipids. Relatively small amounts of unesterified apoA-I45 as well as with secretion of apoE.98 It has been
cholesterol are found in intracellular organelles except in hypothesized that apoA-I binds to a signal-transducing cell-
those that communicate with the plasma membrane (endo- surface receptor and that this binding facilitates the translo-
somes, lysosomes, and trans-Golgi network [TGN]).72 The cation of cholesterol from intracellular compartments to the
TGN is an acceptor of newly synthesized cholesterol from the plasma membrane.42,75 The nature of this receptor is un-
ER and exogenous cholesterol from endocytic vesicles, lyso- known. Because cholesterol efflux from cells and ABC1 are
somes, and caveolae72,73,78 and subsequently distributes cho- defective in Tangier disease, it is likely that ABC1 plays an
lesterol and phospholipids either in the form of detergent-re- important role in apolipoprotein-mediated efflux.48 –52,84,92,93
sistant and caveolin-containing vesicles (rafts or In support of this possibility, apoA-I–mediated cholesterol
cytolipoproteins) or as secretory detergent-soluble efflux is severely decreased by the inhibition of ABC1 with
vesicles.72,73,79 – 81 either antisense oligonucleotides or pharmacological com-
pounds and is increased by the overexpression of ABC1.51,100
Diverse Cholesterol Efflux Pathways In addition to ABC1, other ABC transporters appear to be
Cholesterol efflux from cells is the result of unspecific and involved in apolipoprotein-mediated lipid efflux. For exam-
passive as well as specific and active processes (Figure ple, inhibition of the sterol-regulated half-transporter ABC8
2).30,42,75,76 (ABCG1) impairs cholesterol efflux from cells.55
Protein-free phospholipid vesicles, synthetic cyclodextrins, Native and reconstituted lipid-rich HDLs induce specific
albumin, and trypsinized HDL mediate a slow and unsatur- and unspecific forms of cholesterol efflux. Partial proteolysis
able cholesterol efflux from all cell types.76,82 This form of of either HDL or cells does not fully prevent HDL-mediated
cholesterol efflux is not prevented by partial proteolysis of cholesterol efflux, which is slow, unsaturable, and bidirec-
plasma membranes or by inhibition of intracellular vesicular tional and thus appears to occur by aqueous diffusion.28,42,75,76
transport.76,81,83 It does not involve specific interactions with Esterification of the released cholesterol by LCAT prevents
cell surface receptors or the specific activation of cellular the rediffusion of cholesterol from HDL back to the plasma
transport processes but simply reflects aqueous diffusion of membrane and thus enhances net cholesterol efflux.101 Ex-
cholesterol out of the plasma membrane onto acceptor mol- pression of SR-BI increases HDL-mediated cholesterol ef-
ecules.76 Although the loss in cholesterol from the plasma flux.102 However, SR-BI–mediated cholesterol efflux does
membrane can be replenished, this form of cholesterol efflux not simply reflect the binding of HDL to cells, inasmuch as
has little effect in depleting cells of intracellularly stored binding of HDL to CD36, which is another HDL binding
cholesteryl esters.76,83 scavenger receptor, does not facilitate cholesterol efflux.103
By contrast, lipid-free apoA-I, apoA-II, apoA-IV, apoC, Therefore, it has been suggested that binding of HDL to
and apoE and also amphipathic synthetic peptides without SR-BI facilitates the bidirectional flux between HDL and
sequence homology to these apolipoproteins cause an efflux plasma membrane by reorganization of lipids within choles-
of phospholipids and cholesterol that is fast, saturable, uni- terol- and caveolae-rich domains within the plasma
directional, independent of LCAT, and efficient in reducing membrane.103
18 Arterioscler Thromb Vasc Biol. January 2001

HDL-mediated cholesterol efflux also shares some proper- phatidylinositol?) and cholesterol are transferred (“flopped”)
ties of lipid-free apolipoprotein-mediated cholesterol efflux. from the inner leaflet to the outer leaflet of the plasma bilayer
In the presence of HDL, intracellularly stored cholesteryl membrane.100,115 There they may be picked up by lipid-free
esters are removed.75,76 After incubation of lipid-enriched apolipoproteins or lipid-poor particles, which may even bind
cells with brefeldin A or the PKC inhibitor sphingosine, to ABC1.119,120
HDL-mediated cholesterol efflux is reduced by ⬇50%.84,96 HDL and apoA-I have been previously found to be
Native and reconstituted HDL elicit various signal transduc- internalized by macrophages into an endosomal compart-
tion pathways, which may activate intracellular lipid transfer ment, from which they are resecreted together with lip-
processes.95,96,104 –108 HDL induces the hydrolysis of phos- ids.45– 47 Tangier macrophages appear to have a defect in
phatidylcholine and phosphatidylinositol biphosphates by resecretion and erroneously target internalized HDL to lyso-
phospholipases C and D and thereby the generation of somes for degradation.47 For this reason and because of the
diacylglycerol (DAG), phosphatidic acid, and inositol phos- presence of hyperplastic Golgi structures within lipid-laden
phates. DAG activates PKC, which has been shown to Tangier macrophages,121 it has been suggested that ABC1
stimulate the translocation of newly synthesized cholesterol serves as a protein component of vesicles or rafts that target
to the plasma membrane as well as cholesterol ef- lipids and proteins between lipid-rich intracellular organelles
flux.95,96,104,108 By contrast, HDL-mediated breakdown of (lysosomes and TGN) and the plasma membrane. In fact,
phosphatidylinositol biphosphate and the subsequent mobili- several ABC transporters, such as cystic fibrosis transduc-
zation of intracellular calcium106,109 does not stimulate cho- tance receptor, Rim, and MDRs, are important for the
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

lesterol efflux but rather mediates the mitogenic effects of trafficking of proteins and lipids between intracellular or-
HDL.110 In macrophages, HDL has been shown to increase ganelles and the plasma membrane.122,123
the concentration of cAMP, resulting in the activation of In addition to ABC1, there are several other sterol-
protein kinase A, which may activate the hydrolysis of regulated ABC transporters.54,124 One of them, ABC8, also
cytosolic cholesteryl esters by NCEH and the mobilization appears to be involved in cholesterol efflux from macro-
of cholesterol by ABC1.45,74,97–99,111 Another mechanism of phages.54 It is a so-called half-transporter, inasmuch as it
HDL-mediated cholesterol efflux is retroendocytosis, ie, the consists of only 1 ATP binding cassette and 1
uptake of HDL into clathrin-coated endosomes, which is transmembrane-spanning domain. To be operative, this half-
followed by intracellular enrichment with lipids and transporter needs another half-transporter, which has not yet
resecretion.45– 47 been identified.
ABC1 Is a Pivotal Regulator of Cholesterol Efflux Role of Endogenous ApoE for Cholesterol Efflux
As a complete ABC transporter, ABC1 has 2 highly con- From Macrophages
served cytoplasmic ATP binding cassettes and 2 transmem- Macrophages produce apoE, although to a much lesser extent
brane domains, each of which consists of 6 transmembrane- than do hepatocytes.125 Transplantation of wild-type bone
spanning segments.53,112 In analogy with the multidrug marrow126,127 or the selective expression of a human apoE
resistance (MDR) protein 1, whose low-resolution structure is transgene in macrophages127,128 decreased atherosclerosis in
known, it has been suggested that the transmembrane seg- apoE-deficient mice. Conversely, on a western diet, athero-
ments form the wall of an aqueous chamber within the plasma sclerosis was promoted in wild-type mice receiving bone
membrane. This chamber is opened to the extracellular space marrow from apoE-deficient mice.129 Because expression of a
and to the lipid phase of the plasma membrane but not to the human apoE transgene in macrophages of apoE-deficient
cytosol. Substrates of ABC1, ie, cholesterol, phospholipids, mice128 and transplantation of apoE-deficient macrophage
vitamins A, E, and K, anions, and interleukin-1␤, may be stem cells into wild-type mice129 produced little changes in
transported through this channel.113–116 plasma lipid levels, macrophage-derived apoE exerts anti-
ABC1 is expressed in many organs. The highest expression atherogenic properties largely independent of its effects on
was found in fetal tissues, placenta, liver, lung, and adrenal plasma lipoproteins.
glands.53,54 In cell culture, ABC1 was expressed in confluent One possible explanation for the antiatherogenicity of
but not in growing fibroblasts, indicating that ABC1- macrophage-derived apoE is its contribution to cholesterol
mediated cholesterol efflux is important in cell quiescence efflux from these cells.125 In vitro, lipid loading and activa-
and other conditions of low need for cholesterol.51 tion of protein kinase A with 8-bromo-cAMP stimulates the
The promotor of the ABC1 gene has been analyzed.112,117 synthesis and secretion of apoE in macrophages, which in
It contains binding motifs for several transcription factors, turn facilitates cholesterol efflux from these cells.98,125,126 –134
including the sterol regulatory binding protein, the liver X ApoE secretion is also stimulated in the presence of exoge-
receptor/retinoid X receptor, and activator proteins. In agree- nous HDL and apoA-I.130,132,135,136 It was originally suggested
ment with a regulatory role of these transcription factors, that macrophage-derived apoE facilitates cholesterol efflux
ABC1 expression and lipid efflux are upregulated by choles- by improving the cholesterol acceptor properties of
terol,51,53 oxysterols,117 rexinoids,118 and cAMP ana- HDL.125,130 –132 However, at least human monocyte– derived
logues.51,99 The linker segment of ABC1 has several serine macrophages secrete apoE together with cellular lipids also
and threonine residues that can be phosphorylated and independent of exogenous HDL.133,134 The efficacy of this
thereby modulate ABC1 activity posttranslationally.53 pathway depends on the apoE genotype (ie, it is least efficient
In analogy with MDRs, it has been suggested that ABC1 in apoE4 macrophages and most efficient in apoE2 macro-
forms a channel within the plasma membrane through which phages).134 Moreover, in the presence of excess exogenous
phospholipids (sphingomyelin, phosphatidylserine, and phos- apolipoproteins, including apoE cholesterol, efflux from
von Eckardstein et al HDLs and Arteriosclerosis 19

apoE-producing mouse peritoneal macrophages or J774 mac- of rodents like mice, which do not express CETP, and 20% of
rophages transfected with human apoE was more effective cholesteryl ester elimination from HDLs of species that
than cholesterol efflux from apoE-deficient mouse macro- express CETP.61,62 In agreement with the important role of
phages and untransfected J774 macrophages, respective- SR-BI in the regulation of HDL metabolism are the results of
ly.137,138 This suggests that endogenous apoE modulates studies in genetically modified mice. Knockout of the SR-BI
cholesterol efflux and cholesteryl ester hydrolysis in macro- gene resulted in a 2-fold elevation of HDL-C but an increase
phages by the regulation of intracellular transport in atherosclerosis.148 Overexpression of SR-BI enhanced
processes.137,138 selective cholesteryl ester uptake and HDL catabolism and
severely decreased HDL-C levels.149 In LDL receptor knock-
HDL-Mediated Delivery of Cholesterol to the out mice or apoE knockout mice, overexpression of SR-BI
Liver and Steroidogenic Organs also led to the disappearance of atherogenic lipoproteins150,151
Lipids and proteins of mature ␣-HDL are removed from the and inhibited atherosclerosis. Hence, upregulation of SR-BI
circulation by direct pathways, which involve the selective in the liver may be antiatherogenic by increasing the catab-
lipid uptake by SR-BI61,62 and the holoparticle uptake by olism of lipids from HDL and atherogenic lipoproteins.
apoE receptors, such as cubilin and probably also other SR-BI is downregulated by cholesterol and estradiol and
as-yet-unknown HDL receptors,63,139 as well as by indirect upregulated by polyunsaturated fatty acids in parenchymal
pathways that involve the action of CETP,64 HL,65,66 and liver cells, by adrenocorticotropic hormone in adrenocortical
EL.67 cells, by gonadotropins in theca interna and interstitial cells of
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

the preovulatory ovary, in luteinized granulosa cells, and in


Selective Cholesterol Uptake by SR-BI
the corpus luteum as well as in Leydig cells and less so in
SR-BI mediates the selective uptake of cholesteryl esters
Sertoli cells of the testis.61,62 Interestingly, at least in rats,
from HDL and also LDL into hepatocytes and steroid
in contrast to their effects in hepatocytes, cholesterol and
hormone–producing cells without internalizing HDL pro-
estradiol stimulate SR-BI in macrophage-like Kupffer
teins.61,62,140 In contrast to holoparticle receptors (eg, mem-
cells.152
bers of the LDL receptor gene family or class A scavenger
receptors), which reside in clathrin-coated pits of the plasma Cholesteryl Ester Transfer Protein
membrane and which direct internalized lipoproteins via CETP exchanges cholesteryl esters of HDL2 with triglycer-
endosomes to lysosomes for degradation, SR-BI is predomi- ides of VLDL, IDL, and LDL. The HDL-derived cholesteryl
nantly found in caveolae and targets internalized lipids into esters are removed from the circulation via the LDL receptor
nonendosomal and nonlysosomal compartments.141 Epitopes pathway.64,153 Triglycerides in HDL are hydrolyzed by HL.
recognized by SR-BI appear to include phospholipids and The concerted action of CETP and HL converts larger HDL2
apolipoproteins. The mechanism of how SR-BI achieves the into smaller HDL3 and releases lipid-free apoA-I and/or
dissociation of lipids and proteins and the incorporation of pre-␤1-LpA-I.33 The metabolic importance of this pathway in
cholesteryl esters into the plasma membrane is not yet humans is emphasized by the finding of retarded HDL
understood. Binding of HDL appears to be a necessary but catabolism and elevated HDL-C levels in individuals with
not sufficient prerequisite, because binding of HDL to CD36, CETP deficiency.154 CETP also exchanges cholesteryl esters
another member of the SR-B gene family, does not result in between LDL and VLDL. The direction of transfer is mod-
selective lipid uptake.142,143 Thus, it has been suggested that ulated by apoF, which inhibits the transfer between LDL and
SR-BI exerts a lipid transfer activity, eg, by forming a VLDL but activates the transfer from HDL to VLDL.153,155
hydrophobic channel through which cholesterol diffuses from CETP gene expression is upregulated by cholesterol via
the HDL particle into the plasma membrane.144 Alternatively activation of the sterol regulatory binding protein and through
or in addition, selective uptake by SR-BI may depend on the oxysterols via binding to the liver X receptor/retinoid X
presence of cofactors. Thus, selective uptake of lipids by receptor.156 –158 Responsiveness of CETP to sterols is proba-
SR-BI is facilitated in the presence of HL. HL and other bly the basis of reduced cholesteryl ester transfer activity in
phospholipases may hydrolyze phospholipids of surface lay- plasmas of patients treated with statins.159 Cholesteryl ester
ers from HDL and plasma membrane and thereby enable the transfer activity is increased in hypertriglyceridemic individ-
flux of cholesteryl esters from the lipoprotein core into the uals independently of changes in CETP mass. This is consid-
plasma membrane.145,146 Remodeling of HDLs by CETP also ered to be an important mechanism that causes low HDL-C in
makes HDLs more viable for selective uptake.146 Finally, patients with (postprandial) hypertriglyceridemia and thus in
selective uptake is reduced in apoE-deficient mice, suggest- patients with the metabolic syndrome because of insulin
ing that apoE is also involved in the interaction of HDL with resistance.18,160
SR-BI.147 Because CETP enriches LDL with cholesterol and depletes
As discussed before, SR-BI also mediates cholesterol HDL of cholesterol and because of the initial finding of
efflux from cells.102,103 The net movement of cholesterol into CETP deficiency in Japanese families with longevity, CETP
or out of the cell may depend on the relative activities of was originally considered to be a proatherogenic modulator
extracellular lipid transfer enzymes (LCAT, PLTP, and of HDL metabolism. However, the generation of lipid-free
CETP) and intracellular enzymes, which metabolize choles- apoA-I,33,37,38 the enhancement of RCT,158 and the transfer of
terol to cholesteryl esters, bile acids, lipoproteins, or steroid oxidized lipids161 also indicate antiatherogenic properties.
hormones. Data of genetic studies in men and mice are in agreement with
Turnover studies have suggested that selective uptake this more complex scenario.162 Despite high HDL-C, CETP
accounts for 90% of cholesteryl ester elimination from HDLs deficiency in hypertriglyceridemic Japanese has been associ-
20 Arterioscler Thromb Vasc Biol. January 2001

TABLE 2. Genetic Animal Models of HDL Metabolism and Atherosclerosis


Animal Crossed Other
Gene Species Expression With/Expressed in HDL Lipoproteins Arteriosclerosis References
ApoA-I Mouse ApoA-I deficiency A 2 N 194
Mouse ApoA-I deficiency ApoB overexpression 2 1 195
Mouse Human apoA-I A 1 2 198
Rabbit Human apoA-I A 1 2 199
Mouse Human apoA-I ApoE deficiency 1 2 200, 201
Mouse Human apoA-I A 1 2 202
Somatic expression
Mouse Human apoA-I ApoE deficiency 1 2 202
Somatic expression
Mouse Human apoA-I LDL-R deficiency 1 2 203
Somatic expression
ApoA-II Mouse Murine apoA-II A 1 1 213
Mouse Murine apoA-II Murine apoA-I 1 1 214
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

overexpression
Mouse Human apoA-II A 1 215
Mouse Human apoA-II A 1 Postprandial 2 216
remnants
ApoA-IV Mouse Human apoA-IV A 1 2 206, 207
Mouse Human apoA-IV ApoE deficiency 1 2 206
LCAT Mouse LCAT deficiency A 2 N 210
Mouse Human LCAT A 1 TG 1 211
Rabbit Human LCAT A 1 TG 2 212
Mouse Human LCAT Human CETP 1 TG N 172
overexpression
CETP Mouse Simian CETP A 2 VLDL⫹LDL1 1 169
Mouse Human CETP Human apoC-III 2 2 170
Mouse Human CETP Human 2 2 170
apoA-I/Human
apoC-III
Mouse Human CETP ApoE deficiency 2 TG1 1 171
Mouse Human CETP ApoE 1 TG1 2 171
deficiency/Human
apoA-I
overexpression
Mouse Human CETP Human LCAT 1 TG2 N 171
overexpression
Rat Human CETP Dahl/salt-sensitive 2 1 173
hypertension
Mouse Human CETP LDL-R deficiency N 1 171
HL Mouse HL deficiency ApoE deficiency 1 ␤-VLDL1 2 182
Mouse Human HL A 2 2 183
SR-BI Mouse SR-BI deficiency ApoE deficiency 1 VLDL2N 1 148
Mouse Murine SR-BI LDL-R deficiency 2 LDL2 2 150, 151
LDL-R indicates LDL receptor; TG, triglycerides; N, normal.

ated with an increased risk of coronary events.163 Likewise, a human or a simian CETP transgene decreased HDL-C
nonsynonymous alleles of the polymorphic CETP gene in- and resulted in increased atherosclerosis in some but not
creased the risk of CHD events despite being associated with all mouse models (Table 2).169 –173 Thus, whether inhibi-
increased levels of HDL-C.164 –166 In contrast, a Taq1 tion of CETP prevents or enhances atherosclerosis appears
polymorphism of the CETP gene has repeatedly been to depend on the presence or absence of other dyslipid-
associated with increased HDL-C and decreased risk of emias. CETP has been suggested to be proatherogenic in
CHD.167 Immunization of hypercholesterolemic rabbits hypercholesterolemia (especially when the LDL receptor
with CETP decreased plasma CETP activity, increased pathway is ineffective) and antiatherogenic in hypertri-
HDL-C, and reduced atherosclerosis.168 Overexpression of glyceridemia and hyperalphalipoproteinemia.162
von Eckardstein et al HDLs and Arteriosclerosis 21

HL and EL suggested to mediate the uptake of apoA-I into proximal


HL hydrolyzes phospholipids and triglycerides in all lipopro- tubulus cells.68,69 However, cubilin has no transmembrane
tein classes.65,66 In agreement with the broad substrate and domain and can therefore not mediate the internalization of
particle specificity of this enzyme, HL deficiency in men and its ligands without coreceptors.68 –70 Megalin, a member of
knockout of the HL gene in mice result in the accumulation the LDL receptor gene family, has been suggested to play this
of apoB-containing remnant-like lipoproteins and role in the kidney and the yolk sac.68,69,186 After internaliza-
phospholipid- and apoE-rich HDLs.174,175 ApoA-II inhibits tion, the ligands of cubilin (HDL, apoA-I, and the vitamin
HL so that HDLs devoid of apoA-II are the preferred HDL B12–intrinsic factor complex) are targeted to lysosomes for
particle of HL.176,177 The concerted action of CETP-mediated degradation. Thus, it is unlikely that apoA-I of the primary
cholesteryl ester transfer and the HL-mediated hydrolysis of urine taken up by proximal tubulus cells reenters the plasma
triglycerides and phospholipids depletes the core of large compartment and that cubilin is an important determinant of
HDL2 and helps to form smaller HDL3 as well as lipid-free HDL-C plasma concentration.68
apoA-I and/or pre-␤-HDL.33 Independent of its lipolytic ApoE-containing HDLs, which constitute the minority of
activity, HL appears to serve as a cofactor in the selective HDLs, are internalized by hepatic apoE receptors (LDL
uptake of HDL lipids, which is mediated by SR-BI145,146 but receptor and LDL receptor–related protein).63,187 There is
also by an additional putative HDL binding site in considerable evidence of the presence of additional HDL
hepatocytes.178 receptors on liver cells, which mediate the catabolism of
In addition to regulation of SR-BI, regulation of HL is an apoE-free HDL. Ligand blotting studies have identified HDL
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

important mechanism by which sex steroids affect HDL-C binding sites of different size in liver cells, which are
levels. HL activity is suppressed by estradiol and increased by candidates for receptors mediating hepatic HDL holoparticle
testosterone.179,180 Experiences with HL deficiency in men uptake.139,188 Morphological studies provided evidence for
and genetic animal models suggest that HL exerts both the binding and endocytosis of HDL as well. Some authors
proatherogenic and antiatherogenic activities.66,175 Several have demonstrated resecretion of HDL, which was internal-
HL-deficient men have suffered from premature CHD despite ized into liver cells.189,190 Interestingly, hepatic binding,
high HDL-C levels.174 Despite the inverse correlation be- uptake, degradation, and resecretion of HDL is disturbed in
tween HL activity and HDL-C, a low HL activity has been ob/ob mice, which are deficient in leptin.191 Because ob/ob
associated with the presence of atherosclerosis.181 Knockout mice have elevated HDL-C levels because of retarded HDL
of HL led to the occurrence of an atherogenic lipoprotein catabolism,192 Tall and colleagues have suggested the pres-
phenotype but reduced atherosclerosis in apoE-deficient ence of a hepatic leptin-regulated HDL receptor, which
mice.182 Transgenic overexpression of HL in either mice or regulates HDL-C levels by mediating holoparticle uptake into
rabbits decreased HDL-C levels but did not cause atheroscle- liver cells.191
rosis.175,183 The controversial data from genetic animal mod-
els of HL (and SR-BI) demonstrate the difficulty of interpret- Genetic Disturbances of HDL Metabolism in Men
ing the estrogen-induced increase of HDL-C and the and Animal Models
testosterone-induced decrease of HDL-C toward the effects The pivotal role of apoA-I, ABC1, LCAT, PLTP, CETP,
on atherosclerosis. SR-BI, HL, and EL in the regulation of HDL metabolism is
By contrast to lipoprotein lipase and HL, EL uses phos- highlighted by the eventually tremendous changes of HDL-C
pholipids as its exclusive substrate.67,184,185 By hydrolysis of levels in men with inborn errors of HDL metabolism or in
phospholipids in HDL, EL generates free fatty acids, which genetically modified animal models (Tables 2 and 3). How-
are taken up by endothelial cells. In addition, removal of ever, changes in HDL-C were not always associated with the
phospholipids eventually generates smaller HDL particles, expected inverse changes in atherosclerosis, especially when
which thereby may become viable for the passage through the the catabolism of HDL was modified. This indicates that
endothelial layer into the extravascular space.67 Transient HDL-C levels lack both specificity and sensitivity in assess-
overexpression of EL lowers HDL-C.184 Animal models are ing the effect of an intervention on atherosclerosis. Rather,
needed to evaluate proatherogenic or antiatherogenic effects the concentration of various HDL subclasses and thereby the
of EL. function of HDL as well as the kinetics of HDL metabolism
appear to influence the course of atherosclerosis.
Catabolism of HDL Apolipoproteins Many patients with apoA-I null alleles have suffered from
The removal of the protein constituents of HDL from the CHD very early in their life.193 Increased atherosclerosis was
circulation is less well understood than the removal of also found in one but not in another apoA-I– deficient mouse
HDL-associated lipids. Potential mechanisms for the catabo- model.194,195 Plasmas of apoA-I– deficient men and mice have
lism of HDL holoparticles and lipid-free apolipoproteins are half-normal cholesterol efflux capacity.196,197 In mice and
endocytosis into liver and kidney cells as well as into placenta rabbits, transgenic overexpression of the apoA-I gene yielded
and yolk sac during pregnancy.68 the expected result, namely, increased HDL-C levels and
The receptor for the intrinsic factor/vitamin B12, cubilin, reduced atherosclerosis.198 –203 Somatic overexpression of
has recently been identified as an HDL/apoA-I binding site in apoA-I even induces the regression of preexisting le-
epithelial cells of the proximal tubulus of the kidney and of sions.202,203 In line with improved HDL function and accel-
the yolk sac.68 –70 Small HDL particles with a size of ⬍8 nm erated RCT, expression of apoA-I transgenes in mice resulted
and lipid-free apoA-I are filtrated by renal glomeruli into the in an increased cholesterol efflux capacity of plasma.204 In
primary urine. Because cubilin-deficient men or dogs have men, infusion of apoA-I increased fecal sterol excretion.205
increased excretion of apoA-I in their urine, cubilin has been Likewise, overexpression of a human apoA-IV transgene
22 Arterioscler Thromb Vasc Biol. January 2001

TABLE 3. Inborn Errors of HDL Metabolism and Atherosclerosis in Men


Gene Kind of Defect Zygosity HDL Cholesterol CHD Risk References
ApoA-I Null alleles Homozygosity Virtually zero Increased 2, 193
Heterozygosity Half normal Normal 2, 193
Some structural variants Homozygosity Virtually zero Normal/increased 2, 193
Heterozygosity Half normal Normal 2, 193
LCAT LCAT deficiency Homozygosity Virtually zero Normal/increased 193, 208
Heterozygosity Half normal Normal 193, 208
Fisheye disease Homozygosity Virtually zero Normal/increased 193, 208
Heterozygosity Half normal Normal 193, 208
CETP CETP deficiency Homozygosity Strongly elevated Decreased/normal/under some 2, 162, 163, 193
circumstances increased
Heterozygosity Elevated Normal 2, 162, 163, 193
Ile405Val polymorphism Slightly elevated Increased 164, 166
Asp442Gly Elevated Increased 165
polymorphism
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

HL HL deficiency Homozygosity Elevated Increased 65, 174


ABC1 Tangier disease Homozygosity Virtually zero Moderately increased 188, 209
Heterozygosity Half normal Normal/increased? 188, 209

reduced atherosclerosis in mice.206,207 These data suggest that modification of HDL metabolism can influence the metabo-
stimulated production of HDL precursors enhances RCT and lism of atherogenic apoB-containing lipoproteins. Thus,
protects from atherosclerosis. LCAT-deficient men, ABC1-deficient Tangier patients, and
Although they have reduced HDL-C levels, SR-BI trans- SR-BI– overexpressing mice, all of which do not develop
genic mice show features of enhanced RCT, namely, in- atherosclerosis despite HDL deficiency, have low levels of
creased biliary cholesterol excretion, and are protected from apoB and LDL cholesterol.150,151,188,208 Vice versa, athero-
atherosclerosis.150,151 Vice versa, knockout of SR-BI causes genic apoB-containing lipoproteins accumulate in men and
atherosclerosis despite doubling the HDL-C levels in mice.148 mice with HL deficiency and also in LCAT transgenic mice,
Other inborn errors and genetic manipulation of HDL which develop atherosclerosis despite high levels of
metabolism in men and animal models, respectively, showed HDL-C.174,175,211
less conclusive relationships between HDL metabolism and
atherosclerosis. Defective HDL maturation because of struc- Concluding Remarks
tural apoA-I variants, Tangier disease, LCAT deficiency, or Enhancement of RCT has a great potential for antiatheroscle-
fisheye disease caused premature atherosclerosis in some but rotic drug therapy. However, it is not the increase or decrease
not all patients.188,193,208,209 Heterozygotes for the underlying of plasma HDL-C concentration per se but rather the concen-
defects in the genes of apoA-I, LCAT, and ABC1 have tration of various HDL subclasses, the cellular mobilization
half-normal levels of HDL-C, ie, usually below the cutoff of and transport of lipids, and the kinetics of HDL metabolism
35 mg/dL (0.9 mmol/L) but no increased risk of CHD. that determine the efficacy of RCT and thus the risk of
Likewise, knockout of LCAT caused HDL deficiency but not atherosclerosis. This challenging concept is also applicable to
atherosclerosis in mice.210 Overexpression of LCAT in- other antiatherogenic properties of HDL, such as antioxida-
creased HDL-C in mice and rabbits but was antiatherogenic tive, anti-inflammatory, antiadhesive, antiaggregatory, and
in rabbits and proatherogenic in mice.211,212 High HDL-C profibrinolytic effects. Importantly, the interpretation of data
levels in CETP deficiency were originally found to be from recent trials on statins, fibrates, and estrogens should be
associated with reduced CHD risk but were later found to be cautious in view of the diverse functionality of HDL and the
associated with increased CHD risk in hypertriglyceridemic pleiotropic effects of these drugs. Moreover, functional and
subpopulations.163 Likewise, some genetic variants of controlled interventional clinical outcome studies are needed
CETP164 –166 were associated with increases in HDL-C and at an early stage to prove the antiatherogenic effects of
CHD risk, whereas others showed the expected inverse HDL-elevating drugs that are currently being developed by
association between HDL-C and CHD risk (Table 3). In various pharmaceutical companies.
agreement with the controversial data in men, overexpression
of CETP decreased HDL-C but had proatherogenic or anti- Acknowledgment
atherogenic effects in different mouse models.169 –173 Like- Dr Arnold von Eckardstein is supported by a grant from the
wise, human HL deficiency as well as transgenic overexpres- European Union on “The Antiatherogenicity of HDL” (grant No.
sion or knockout of HL or apoA-II in mice unraveled the BIOMED2 BMH4-CT98-3699).
antiatherogenic and proatherogenic effects of HL and apoA-
II, respectively.174,175,182,183,213–216 Thus, whether LCAT,
References
1. Gordon D, Rifkind BM. Current concepts: high density lipoproteins–the
CETP, or HL is proatherogenic or antiatherogenic appears to clinical implications of recent studies. N Engl J Med. 1989;321:
be strongly influenced by additional factors. In addition, 1311–1315.
von Eckardstein et al HDLs and Arteriosclerosis 23

2. Genest J, Marcil M, Denis M, Yu L. High density lipoproteins in health Bezafibrate Infarction Prevention (BIP) Study. Circulation. 2000;102:
and disease. J Invest Med. 1999;47:31– 42. 21–27.
3. Stein O, Stein Y. Atheroprotective mechanisms of HDL. Atherosclero- 24. Hulley S, Grady D, Bush T, Furberg C, Herrington D, Riggs B, Vittinghoff
sis. 1999;144:285–303. E. Randomized trial of estrogen plus progestin for secondary prevention of
4. von Eckardstein A, Assmann G. Prevention of coronary heart disease by coronary heart disease in postmenopausal women: Heart and Estrogen/
raising of high density lipoprotein cholesterol? Curr Opin Lipidol. 2000; Progestin Replacement Study (HERS) Research Group. JAMA. 1998;280:
11:627– 637. 605–613.
5. Dietschy JM, Turley SO, Spady DK. Role of liver in the maintenance of 25. von Eckardstein A, Huang Y, Assmann G. Physiological role and
cholesterol and low density lipoprotein homeostasis in different animal clinical relevance of high-density lipoprotein subclasses. Curr Opin
species including humans. J Lipid Res. 1993;34:1637–1639. Lipidol. 1994;5:404 – 416.
6. Glomset JA. The plasma lecithin:cholesterol acyltransferase reaction. J 26. Barrans A, Jaspard B, Barbaras R, Chap H, Perret B, Collet X. Pre-␤
Lipid Res. 1968, 9:155–167. HDL: structure and metabolism. Biochim Biophys Acta. 1996;1300:
7. Genest JJ, McNamara JR, Salem DN, Schaefer EJ. Prevalence of risk 73– 85.
factors in men with premature coronary artery disease. Am J Cardiol. 27. Fielding C, Fielding PE. Molecular physiology of reverse cholesterol
1991;67:1185–1189. transport. J Lipid Res. 1995;36:211–228.
8. Bolibar I, von Eckardstein A, Assmann G, Thompson S, on behalf of the 28. Huang Y, von Eckardstein A, Wu S, Maeda N, Assmann G. A solely
ECAT Angina Pectoris Study Group. Short-term prognostic value of apolipoprotein E containing plasma lipoprotein with electrophoretic
lipid measurements for coronary events in patients with angina pectoris. gamma-mobility takes up cellular cholesterol. Proc Natl Acad Sci U S A.
Thromb Haemost. 2000;84:955–961. 1994;91:1834 –1838.
9. International Task Force for Prevention of Coronary Heart Disease. 29. Duverger N, Ghalim N, Ailhaud G, Steinmetz A, Fruchart J-C, Castro G.
Coronary heart disease: reducing the risk: the scientific background to Characterization of apoA-IV-containing lipoprotein particles isolated
primary and secondary prevention of coronary heart disease: a from human plasma and interstitial fluid. Arterioscler Thromb. 1993;
worldwide view. Nutr Metab Cardiovasc Dis. 1998;8:205–271. 13:126 –132.
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

10. Expert panel on detection, evaluation and treatment of high blood 30. von Eckardstein A, Huang Y, Wu S, Saadat Sarmadi A, Schwarz S,
cholesterol in adults. The second report of the National Cholesterol Steinmetz A, Assmann G. Lipoproteins containing apolipoprotein A-IV
Education Program (NCEP) expert panel on detection, evaluation and but not apolipoprotein A-I take up and esterify cell-derived cholesterol
treatment of high blood cholesterol in adults (Adult Treatment Panel II). in plasma. Arterioscler Thromb Vasc Biol. 1995;15:1755–1763.
Circulation. 1994;89:1329 –1445. 31. Asztalos BF, Sloop CH, Wong L, Roheim PS. Comparison of apo
11. Harper CR, Jacobson TA. New perspectives on the management of low A-I-containing subpopulations of dog plasma and prenodal peripheral
levels of high density lipoprotein cholesterol. Arch Intern Med. 1999; lymph: evidence for alteration in subpopulations in the interstitial space.
159:1049 –1057. Biochim Biophys Acta. 1993;1169:301–304.
12. Genest JJ Jr, Martin-Munleey SS, McNamara JR, Ordovas JM, Jenner J,
32. Nanjee MN, Cooke CJ, Olzewski WL, Miller NE. Concentrations of
Myers RH, Silberman SR, Wilson PW, Salem DN, Schaefer EJ. Familial
electrophoretic and size subclasses of apolipoprotein A-I– containing
lipoprotein disorders in patients with coronary heart disease. Circu-
particles in human peripheral lymph. Arterioscler Thromb Vasc Biol.
lation. 1992;85:2025–2033.
2000;20:2148 –2155.
13. Manninen V, Tenkanen L, Koskinen P, Huttunen JK, Mänttäri M,
33. Rye KA, Clay MA, Barter PJ. Remodeling of high density lipoproteins
Heinonen OP, Frick MH. Joint effects of serum triglycerides and LDL
by plasma factors. Atherosclerosis. 1999;145:227–238.
cholesterol and HDL cholesterol concentrations on coronary heart
34. Castle CK, Pape ME, Marotti KR, Melchior GW. Secretion of pre-beta-
disease risk in the Helsinki Heart Study: implications for treatment.
migrating apo A-I by cynomolgus monkey hepatocytes in culture. J
Circulation. 1992;85:37– 45.
Lipid Res. 1991;32:439 – 447.
14. Rubins HB, Robins SJ, Collins D, Fye CL, Anderson JW, Elam MB,
35. Danielsen EM, Hansen GH, Poulsen MD. Apical secretion of apoli-
Faas FH, Linares E, Schaefer EJ, Schectman G, et al. Gemfibrozil for the
poproteins from enterocytes. J Cell Biol. 1993;120:1347–1356.
secondary prevention of coronary heart disease in men with low levels
36. Musliner TA, Long DL, Forte TM, Nichols AV, Gong EL, Blanche,
of high-density lipoprotein cholesterol. N Engl J Med. 1999;341:
Krauss RM. Dissociation of high density lipoprotein precursors from
410 – 418.
15. De Backer G, de Bacqeur D, Kornitzer M. Epidemiological aspects of apolipoprotein B-containing lipoproteins in the presence of unesterified
high density lipoprotein cholesterol. Atherosclerosis. 1998;137(suppl): fatty acids and a source of apolipoprotein A-I. J Lipid Res. 1991;32:
S1–S6. 917–932.
16. Bobak M, Hense HW, Kark J, Kuch B, Vojtisek P, Sinnreich R, Gostomzyk 37. Liang HQ, Rye KA, Barter PJ. Dissociation of lipid-free apolipoprotein
J, Bui M, von-Eckardstein A, Junker R, et al. An ecological study of A-I from high density lipoproteins. J Lipid Res. 1994;35:1187–1199.
determinants of coronary heart disease rates: a comparison of Czech, 38. Francone OL, Royer L, Haghpassand M. Increased pre-␤-HDL levels,
Bavarian and Israeli men. Int J Epidemiol. 1999;28:437–444. cholesterol efflux, and LCAT mediated esterification in mice expressing
17. Hergenc G, Schulte H, Assmann G, von Eckardstein A. Associations of the human cholesteryl ester transfer protein (CETP) and human apoli-
obesity markers, insulin, and sex hormones with HDL-cholesterol levels poprotein A-I (apoA-I) transgenes. J Lipid Res. 1996;37:1268 –1277.
in Turkish and German individuals Atherosclerosis. 1999;145:147–156. 39. von Eckardstein A, Jauhiainen M, Huang Y, Metso J, Langer C,
18. Després JP, Marette A. Relation of components of insulin resistance to Pussinen P, Wu S, Ehnholm C, Assmann G. Phospholipid transfer
coronary disease risk Curr Opin Lipidol. 1994;5:274 –289. protein mediated conversion of high density lipoproteins (HDL) gen-
19. Erren M, Reinecke H, Junker R, Fobker M, Schulte H, Schurek JO, erates generation of pre␤1-HDL. Biochim Biophys Acta. 1996;1301:
Kropf J, Kerber S, Breithardt G, Assmann G, et al. Systemic inflam- 255–261.
matory parameters in patients with atherosclerosis of the coronary and 40. Jiang X-J, Francone OL, Bruce C, Milne R, Mar J, Walsh A, Breslow
peripheral arteries. Arterioscler Thromb Vasc Biol. 1999;19:2355–2363. JL, Tall AR. Increased pre-␤-high density lipoprotein, apolipoprotein
20. Gould AL, Rossouw JE, Santanello NC, Heyse J, Furberg CD. Choles- A-I, and phospholipid in mice expressing the human phospholipid
terol reduction yields clinical benefit: impact of statin trials. Circulation. transfer protein and human apolipoprotein A-I transgenes. J Clin Invest.
1998;97:946 –952. 1996;98:2373–2380.
21. Gotto AM Jr, Whitney E, Stein EA, Shapiro DR, Clearfield M, Weis S, 41. Barrans A, Collet X, Barbaras R, Jaspard B, Manent J, Vieu C, Chap H,
Jou JY, Langendorfer A, Beere PA, Watson DJ, et al. Relation between Perret B. Hepatic lipase induces the formation of pre-␤-1-high density
baseline and on-treatment lipid parameters and first acute major coro- lipoprotein (HDL) from triacylglycerol-rich HDL2. J Biol Chem. 1994;
nary events in the Air Force/Texas Coronary Atherosclerosis Prevention 269:11572–11577.
Study (AFCAPS/TexCAPS). Circulation. 2000;101:477– 484. 42. Oram JF, Yokoyama S. Apolipoprotein-mediated removal of cellular
22. Ruotolo G, Ericsson CG, Tettamanti C, Karpe F, Grip L, Svane B, cholesterol and phospholipids. J Lipid Res. 1997;37:2473–2491.
Nilsson J, de Faire U, Hamsten A Treatment effects on serum 43. Forte TM, Goth-Goldstein R, Nordhausen RW, McCall MR. Apoli-
lipoprotein lipids, apolipoproteins and low density lipoprotein particle poprotein A-I-cell membrane interaction: extracellular assembly of het-
size and relationships of lipoprotein variables to progression of coronary erogeneous nascent HDL particles. J Lipid Res. 1993;34:317–324.
artery disease in the Bezafibrate Coronary Atherosclerosis Intervention 44. Asztalos B, Zhang W, Roheim PS, Wong L. Role of free apolipoprotein
Trial (BECAIT). J Am Coll Cardiol. 1998;32:1648 –1656. A-I in cholesterol efflux: formation of pre-␤-migrating high density
23. The BIP Study Group. Secondary prevention by raising HDL cholesterol lipoprotein particles. Arterioscler Thromb Vasc Biol. 1997;17:
and reducing triglycerides in patients with coronary artery disease: the 1630 –1636.
24 Arterioscler Thromb Vasc Biol. January 2001

45. Takahashi Y, Smith JL. Cholesterol efflux to apolipoprotein A-I 66. Thuren T. Hepatic lipase and HDL metabolism. Curr Opin Lipidol.
involves endocytosis and resecretion in a calcium-dependent pathway. 2000;11:277–284.
Proc Natl Acad Sci U S A. 1999;96:11358 –11363. 67. Rader D, Jaye M. Endothelial lipase: a new member of the triglyceride
46. Heeren J, Weber W, Beisiegel U. Intracellular processing of endo- lipase gene family. Curr Opin Lipidol. 2000;11:141–148.
cytosed triglyceride-rich lipoproteins comprises both recycling and deg- 68. Moestrup SK, Kozyraki R, Cubilin, a high density lipoprotein receptor.
radation. J Cell Sci. 1999;112:349 –359. Curr Opin Lipidol. 2000;11:133–140.
47. Schmitz G, Assmann G, Robenek H, Brennhausen B. Tangier disease: a 69. Kozyraki R, Fyfe J, Kristiansen M, Gerdes C, Jacobsen C, Cui SY,
disorder of intracellular membrane traffic. Proc Natl Acad Sci U S A. Christensen EI, Aminoff M, de la Chapelle A, Krahe R, et al. The
1985;82:6305– 6311. intrinsic factor-vitamin B-12 receptor, cubilin, is a high-affinity apoli-
48. Brooks-Wilson A, Marcil M, Clee SM, Zhang LH, Roomp K, van Dam poprotein A-I receptor facilitating endocytosis of high-density
M, Yu L, Brewer C, Collins JA, Molhuizen HOF, et al. Mutations in lipoprotein. Nat Med. 1999;5:656 – 661.
ABC1 in Tangier disease and familial high density lipoprotein defi- 70. Hammad SM, Steffansson S, Twal WO, Drake CJ, Fleming P, Remaley
ciency. Nat Genet. 1999;22:336 –345. A, Brewer HB Jr, Argraves WS. Cubilin, the endocytic receptor for
49. Bodzioch M, Orso E, Klucken J, Langmann T, Böttcher A, Diedrich W, intrinsic factor-vitamin B(12) complex, mediates high-density
Drobnik W, Barlage S, Büchler C, Porsch-Özcürümez M, et al. The gene lipoprotein holoparticle endocytosis. Proc Natl Acad Sci U S A. 1999;
encoding ATP binding cassette transporter 1 is mutated in Tangier 96:10158 –10163.
disease. Nat Genet. 1999;22:347–351. 71. Chang TY, Chang CY, Cheng D. Acyl coenzyme A: cholesterol acyl-
50. Rust S, Rosier M, Funke H, Real J, Amoura Z, Piette JC, Deleuze JF, transferase. Annu Rev Biochem. 1997;66:613– 638.
Brewer HB, Duverger N, Denefle P, et al. Tangier disease is caused by 72. Fielding CJ, Fielding PE. Intracellular cholesterol transport. J Lipid Res.
mutations in the gene encoding ATP binding cassette transporter 1. Nat 1997;38:1503–1521.
Genet. 1999;22:352–255. 73. Liscum L, Munn NJ. Intracellular cholesterol transport. Biochim
51. Lawn RM, Wade DP, Garvin MR, Wanfg X, Schwarttz K, Porter JG, Biophys Acta. 1999;1438:19 –37.
Seiilhamer JJ, Vaughan AM, Oram JF. The Tangier disease gene prod- 74. Bernard D, Rodriguez A, Rothblat GH, Glick JM. cAMP stimulates
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

uct controls the cellular apolipoprotein-mediated lipid removal pathway. cholesteryl ester clearance to high density lipoproteins in J774 macro-
J Clin Invest. 1999;104:R25–R31. phages. J Biol Chem. 1991;266:710 –716.
52. McNeish J, Aiello RJ, Guyot D, Turi T, Gabel C, Aldinger C, Hoppe 75. von Eckardstein A. Cholesterol efflux from macrophages and other
KL; Roach ML, Royer LJ, de Wet J, et al. High density lipoprotein cells. Curr Opin Lipidol. 1996;7:308 –319.
deficiency and foam cell accumulation in mice with targeted disruption 76. Rothblat GH, de la Llera Moya M, Atger V, Kellner-Weibel G, Williams
of ATP-binding cassette transporter-1. Proc Natl Acad Sci U S A. 2000; DL, Phillips MC. Cell cholesterol efflux: integration of old and new
97:4245– 4250. observations provides new insights. J Lipid Res. 1999;40:781–796.
53. Langmann T, Klucken J, Reil M, Liebisch G, Luciani M-F, Chimini G, 77. Kurzchiella TV, Parton RG. Membrane microdomains and caveolae.
Curr Opin Cell Biol. 1999;11:424 – 431.
Kaminski WE, Schmitz G. Molecular cloning of the human ATP
78. Bretscher MS, Munro S. Cholesterol and the Golgi apparatus. Science.
binding cassette transporter 1 (hABC1): evidence for sterol-dependent
1993;261:1280 –1281.
regulation in macrophages. Biochem Biophys Res Commun. 1999;257:
79. Simons K, Ikonen E. Functional rafts in cell membranes. Nature. 1997;
29 –33.
387:569 –572.
54. Klucken J, Büchler C, Orso E, Kaminski WE, Porsch-Özcürümez M,
80. Rothman JE, Wieland FT. Protein sorting by transport vesicles. Science.
Liebisch G, Kapinsky M, Diedrich W, Drobnik W, Dean M, et al.
1996;272:227–234.
ABCG1 (ABC8), the human homolog of the Drosophila white gene, is
81. Mendez AJ. Monensin and brefeldin A inhibit high density lipoprotein
a regulator of macrophage cholesterol and phospholipid transport. Proc
mediated cholesterol efflux from cholesterol-enriched cells: implications
Natl Acad Sci U S A. 2000;97:817– 822.
for intracellular cholesterol trafficking. J Biol Chem. 1995;270:
55. Miida T, Kawano M, Fielding CJ, Fielding PE. Regulation of the
5891–900.
concentration of pre high density lipoprotein in normal plasma by cell
82. Zhao Y, Marcel YL. Serum albumin is a significant intermediate in
membranes and lecithin:cholesterol acyltransferase. Biochemistry. 1992;
cholesterol transfer between cells and lipoproteins. Biochemistry. 1996;
31:11112–11120.
35:7174 –7180.
56. Liang HQ, Rye KA, Barter PJ. Remodelling of reconstituted high 83. Mendez AJ. Cholesterol efflux mediated by apolipoproteins is an active
density lipoproteins by lecithin:cholesterol acyltransferase. J Lipid Res. cellular process distinct from efflux mediated by passive diffusion. J
1996;37:1962–1970. Lipid Res. 1997;38:1807–1821.
57. von Eckardstein A, Huang Y, Kastelein JJP, Geisel J, Réal J, Miccoli R, 84. Remaley AT, Schumacher K, Stonik JA, Farsi BD, Nazih H, Brewer HB
Noseda G, Kuivenhoven JA, Assmann G. Lipid-free apolipoprotein Jr. Decreased reverse cholesterol transport from Tangier disease fibro-
(apo) A-I is converted into alpha-migrating high density lipoproteins by blasts: acceptor specificity and effect of brefeldin on lipid efflux. Arte-
lipoprotein depleted plasma of normolipidemic donors and apoA-I- rioscler Thromb Vasc Biol. 1997;17:1813–1821.
deficient patients but not of Tangier disease patients. Atherosclerosis. 85. Hara H, Yokoyama S. Interaction of free apolipoproteins with macro-
1998;138:25–34. phages: formation of high density lipoprotein-like lipoproteins and
58. Dieplinger H, Zechner R, Kostner GM. The in vitro formation of HDL2 reduction of cellular cholesterol. J Biol Chem. 1991;266:3080 –3086.
during the action of LCAT: the role of triglyceride-rich lipoproteins. J 86. Mendez AJ, Anantharamaiah GM, Segrest JP, Oram JF. Synthetic
Lipid Res. 1985;26:273–279. amphipathic helical peptides that mimic apolipoprotein A-I in clearing
59. Lusa S, Jauhiainen M, Metso J, Somerharju P, Ehnholm C. The cellular cholesterol. J Clin Invest. 1994;94:1698 –1705.
mechanism of human plasma phospholipid transfer protein-induced 87. Yancey PG, Bielicki JK, Johnson WJ, Lund-Katz S, Palgunachari MN,
enlargement of high-density lipoprotein particles: evidence for particle Anantharamaiah AM, Segrest JP, Phillips MC, Rothblat GH. Efflux of
fusion. Biochem J. 1996;313:275–282. cellular cholesterol and phospholipid to lipid-free apolipoproteins and
60. Jiang XC, Bruce C, Mar J, Lin M, Ji Y, Francone OL, Tall AR. Targeted class A amphipathic peptides. Biochemistry. 1995;34:7955–7965.
mutation of plasma phospholipid transfer protein markedly reduces high 88. Gillotte KL, Zaiou M, Lund-Katz S, Anantaraqmaiah GM, Holvoet P,
density lipoprotein levels. J Clin Invest. 1999;103:907–914. Dhoest A, Palgunachari MN, Segrest JP, Weisgraber KH, Rothblat GH,
61. Krieger M. Charting the fate of the good cholesterol: identification and et al. Apolipoprotein-mediated plasma membrane microsolubilization:
characterization of the high density lipoprotein receptor SR-BI. Annu role of lipid affinity and membrane penetration in the efflux of cellular
Rev Biochem. 1999;68:523–558. cholesterol and phospholipid. J Biol Chem. 1999;274:2021–2028.
62. Trigatti B, Rigotti A, Krieger M. The role of high-density lipoprotein 89. Fielding CJ, Bist A, Fielding PE. Intracellular cholesterol transport in
receptor SR-BI in cholesterol metabolism. Curr Opin Lipidol. 2000;11: synchronized human skin fibroblasts. Biochemistry. 1999;38:
123–132. 2506 –2513.
63. Curtiss LK, Boisvert WA. Apolipoprotein E and atherosclerosis. Curr 90. Francis GA, Tsujita M, Terry TL. Apolipoprotein A-I efficiently binds
Opin Lipidol. 2000;11:243–251. to and mediates cholesterol and phospholipid efflux from human but not
64. Tall AR, Jiang XC, Luo Y, Silver D. George Lyman Duff Memorial from rat aortic smooth muscle cells. Biochemistry. 1999;38:
Lecture: lipid transfer proteins, HDL metabolism and atherogenesis. 16315–16322.
Arterioscler Thromb Vasc Biol. 2000;1999:20:1185–1188. 91. Li Q, Yokoyama S. Independent regulation of cholesterol incorporation
65. Cohen J, Vega GL, Grundy SM. Hepatic lipase: new insights from into free apolipoprotein-mediated cellular lipid efflux in rat vascular
genetic and metabolic studies. Curr Opin Lipidol. 1999;10:259 —268. smooth muscle cells. J Biol Chem. 1995;269:26216 –26223.
von Eckardstein et al HDLs and Arteriosclerosis 25

92. Francis GA, Knopp RH, Oram JF. Defective removal of cellular cho- 113. Scott J. Good cholesterol news. Nature. 1999;400:816 – 817.
lesterol and phospholipids by apolipoprotein A-I in Tangier disease. 114. Becq F, Hamon Y, Bajetto A, Gola M, Verrier B, Chimini G. ABC1, an
J Clin Invest. 1995;96:78 – 87. ATP binding cassette transporter required for phagocytosis of apoptotic
93. Walter M, Gerdes U, Seedorf U, Assmann G. The high density cells, generates a regulated anion flux after expression in Xenopus laevis
lipoprotein- and apolipoprotein A-I-induced mobilization of cellular oocytes. J Biol Chem. 1997;272:2695–2915.
cholesterol is impaired in fibroblasts from Tangier disease subjects. 115. Hamon Y, Luciani MF, Becq F, Verrier B, Rubartelli A, Chimini G.
Biochem Biophys Res Commun. 1994;205:850 – 856. Interleukin 1␤ secretion is impaired by inhibitors of the ATP binding
94. Fielding PE, Fielding CJ. Plasma membrane caveolae mediate the efflux cassette transporter ABC1. Blood. 1997;90:2911–2915.
of cellular free cholesterol. Biochemistry. 1995;34:14288 –14292. 116. Bevers EM, Comfurius P, Dekkers DWC, Zwaal RFA. Lipid translo-
95. Theret N, Delbart C, Aquie G, Fruchart JC, Vassaux G, Ailhaud G. cation across the plasma membrane of mammalian cells. Biochim
Cholesterol efflux from adipose cells is coupled to diacylglycerol pro- Biophys Acta. 1999;1439:317–330.
duction and protein kinase C activation. Biochem Biophys Res Commun. 117. Costet P, Luo Y, Wang N, Tall AR. Sterol-dependent transactivation of
1990;137:1361–1368. the ABC1 promoter by LXR/RXR: LXRs transactivate ABC1 promotor.
96. Mendez AJ, Oram JF, Bierman EL. Protein kinase C as a mediator of J Biol Chem. 2000;275:28240 –28245.
high density lipoprotein dependent efflux of intracellular cholesterol. 118. Repa JJ, Turley SD, Lobaccaro JA, Medina J, Li L, Lustig K, Shan B,
J Biol Chem. 1991;266:10104 –10111. Heyman RA, Dietschy JM, Mangelsdorf DJ. Regulation of absorption
97. Sakr SW, Williams DL, Stoudt GW, Phillips MC, Rothblat GH. and ABC1-mediated efflux of cholesterol by RXR heterodimers.
Induction of cholesterol efflux to lipid-free apolipoprotein A-I by Science. 2000;289:1524 –1529.
cAMP. Biochim Biophys Acta. 1999;1438:85–98. 119. Wang N, Silver DL, Costet P, Tall AR. Specific binding of apoA-I,
98. Smith JD, Miyata M, Ginsberg M, Grigaux C, Shmookler E, Plump AS. enhanced cholesterol efflux and altered plasma membrane morphology
Cyclic AMP induces apolipoprotein E binding activity and promotes in cells expressing ABC1. J Biol Chem. 2000;275:33053–33058.
cholesterol efflux from a macrophage cell line to apolipoprotein 120. Oram JF, Lawn RM, Garver MR, Wade DP. ABCA1 is the cAMP-
acceptors. J Biol Chem. 1996;271:30647–30655. inducible receptor that mediates cholesterol secretion from macro-
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

99. Bortnick AE, Rothblat GH, Stoudt G, Hoppe KL, Royer LJ, McNeish J, phages. J Biol Chem. 2000;275:34508 –34511.
Francone OL. The correlation of ATP-binding cassette 1 mRNA levels 121. Orso E, Broccardo C, Kaminski WE, Böttcher A, Liebisch G, Drobnik
with cholesterol efflux from various cell lines. J Biol Chem. 2000;275: W, Götz A, Chambenoit O, Diederich W, Langmann T, et al. Transport
28634 –28640. of lipids from Golgi to plasma membrane is defective in Tangier disease:
100. Hamon Y, Broccardo C, Chambenoit O, Lucuani M-F, Oti F, Chaslin S, patients and Abc1-deficient mice. Nat Genet. 2000;24:192–196.
Freyssinet J-M, Devaux PF, McNeish J, Marguet D, et al. ABC1 122. Bradbury NA. Intracellular CFTR: localization and function. Physiol
promotes engulfment of apoptotic cells and transbilayer redistribution of Rev. 1999;79:S175–S191.
phosphatidylserine. Nat Cell Biol. 2000;2:399 – 406. 123. Weng J, Mata NL, Azarian SM, Tzekov RT, Birch DG, Travis GH.
101. Czarnecka H, Yokoyama S. Lecithin:cholesterol acyltransferase reaction Insights into the function of Rim protein in photoreceptors and etiology
on cellular lipid released by free apolipoprotein-mediated efflux. Bio- of Stargardt’s disease from the phenotype in abcr knockout mice. Cell.
chemistry. 1995;34:4385– 4392. 1999;98:12–23.
102. Ji Y, Jian B, Wang N, Sun Y, Moya ML, Phillips MC, Rothblat GH, 124. Kaminski WE, Orso E, Diederich W, Klucken J, Drobnik W, Schmitz G.
Swaney JB, Tall AR. Scavenger receptor BI promotes high density Identification of a novel human sterol-sensitive ATP-binding cassette
lipoprotein-mediated cellular cholesterol efflux. J Biol Chem. 1997;272: transporter (ABCA7). Biochem Biophys Res Commun. 2000;273:
20982–20985. 532–538.
103. De la Llera-Moya M, Rothblat GH, Connelly MA, Kellner-Weibel G, 125. Mazzone T. Apolipoprotein E secretion by macrophages: its potential
Sakr SW, Phillipis MC, Williams DL. Scavenger receptor B1 mediates functions. Curr Opin Lipidol. 1996;7:303–310.
free cholesterol flux independent of HDL tethering to the cell surface. J 126. Linton MF, Atkinson JB, Fazio S. Prevention of atherosclerosis in
Lipid Res. 1999;40:575–580. apolipoprotein E-deficient mice by bone marrow transplantation.
104. Walter M, Reinecke H, Nofer J-R, Seedorf U, Assmann G. HDL3 Science. 1995;267:1034 –1037.
stimulates multiple signaling pathways in human skin fibroblasts. Arte- 127. Bolsvert WA, Spangenberg J, Curtiss LK. Treatment of severe hyper-
rioscler Thromb Vasc Biol. 1995;15:1975–1983. lipidemia in apolipoprotein E-deficient mice by bone marrow transplan-
105. Walter M, Reinecke H, Gerdes U, Nofer JR, Höbbel G, Seedorf U, tation. J Clin Invest. 1995;96:1118 –1124.
Assmann G. Defective regulation of phosphatidylcholine-specific phos- 128. Bellosta S, Mahley RW, Sanan DA, Newland DL, Taylor JM, Pitas RE.
pholipases C and D in a kindred with Tangier disease: evidence for the Macrophage-specific expression of human apolipoprotein E reduces
involvement of phosphatidylcholine breakdown in HDL-mediated cho- atherosclerosis in hypercholesterolemic apolipoproteinE-null mice.
lesterol efflux mechanisms. J Clin Invest. 1996;98:2315–2323. J Clin Invest. 1995;96:2170 –2179.
106. Pörn MI, Ackerman KEO, Slotte JP. High density lipoprotein induces a 129. Fazio S, Babaev VR, Murray AB, Hasty AH, Carter KJ, Gleaves LA,
rapid and transient release of Ca2⫹ in cultured fibroblasts. Biochem J. Atkinson JB, Linton MF. Increased atherosclerosis in mice reconstituted
1991;279:29 –33. with apolipoprotein E null macrophages. Proc Natl Acad Sci U S A.
107. Mollers C, Drobnik W, Resink T, Schmitz G. High density lipoproteins 1997;94:4647– 4652.
and low density lipoprotein mediated signal transduction in cultured 130. Basu S, Goldstein JL, Brown MS. Independent pathways for secretion of
human skin fibroblasts. Cell Signal. 1995;7:695–707. free cholesterol and apolipoprotein E by macrophages. Science. 1983;
108. Rogler GB, Trümbach B, Klima B, Lackner KJ, Schmitz G. HDL- 219:871– 873.
mediated efflux of intracellular cholesterol is impaired in fibroblasts 131. Dory L. Synthesis and secretion of apo E in thioglycolate-elicited mouse
from Tangier disease patients. Arterioscler Thromb Vasc Biol. 1995;15: peritoneal macrophages: effect of cholesterol efflux. J Lipid Res. 1989;
683– 690. 30:809 – 816.
109. Drobnik W, Möllers C, Resink T, Schmitz G. Activation of 132. Mazzone T, Reardon C. Expression of heterologous human apoli-
phosphatidylinositol-specific phospholipase C in response to HDL3 and poprotein E by J774 macrophages enhances cholesterol efflux to HDL3.
LDL is markedly reduced in cultured fibroblasts from Tangier patients. J Lipid Res. 1994;35:1345–1353.
Arterioscler Thromb Vasc Biol. 1995;15:1369 –1377. 133. Zhang WY, Gaynor PM, Kruth HS. Apolipoprotein E produced by
110. Nofer JR, Fobker M, Höbbel G, Voss R, Wolinska I, Tepel M, Zidek W, human monocyte-derived macrophages mediates cholesterol efflux that
Junker R, Seedorf U, von Eckardstein A, et al. Activation of occurs in the absence of added cholesterol acceptors. J Biol Chem.
phosphatidylinositol-specific phospholipase C (PI-PLC) by HDL- 1996;271:28641–28646.
associated lysosphingolipids: involvement in mitogenesis but not in 134. Cullen P, Cignarella A, Brennhausen B, Mohr S, Assmann G, von
cholesterol efflux. Biochemistry. 2000;39:15199 –15207. Eckardstein A. Phenotype-dependent differences in apolipoprotein E
111. Khoo JC, Mahoney EM, Steinberg DS. Neutral cholesterol esterase metabolism and cholesterol homeostasis in human monocyte-derived
activity in macrophages and its enhancement by cyclic AMP-dependent macrophages. J Clin Invest. 1998;101:1670 –1677.
protein kinase. J Biol Chem. 1981;256:12659 –12663. 135. Bielicki JK, McCall MR, Forte TM. Apolipoprotein A-I promotes cho-
112. Santamarina-Fojo S, Peterson K, Knapper C, Qiu Y, Freeman L, Cheng lesterol release and apolipoprotein E recruitment from THP-1 macro-
J-F, Osorio J, Remaley A, Yang X-P, Haudenschild C, et al. Complete phage like foam cells. J Lipid Res. 1999;40:85–92.
genomic sequence of the human ABCA1 gene: analysis of the human 136. Rees D, Sloane T, Jessup W, Dean RT, Kritharides L. Apolipoprotein
and mouse ATP biding cassette A promoter. Proc Natl Acad Sci U S A. A-I stimulates secretion of apolipoprotein E by foam cell macrophages.
2000;97:7987–7992. J Biol Chem. 1999;274:27925–27933.
26 Arterioscler Thromb Vasc Biol. January 2001

137. Lin YC, Duan H, Mazzone T. Apolipoprotein E-dependent cholesterol transfer protein gene in vivo but is not required for sterol up-regulation
efflux from macrophages: kinetic study and divergent mechanisms for of gene expression. J Biol Chem. 1998;273:22409 –22414.
endogenous versus exogenous apolipoprotein E. J Lipid Res. 1999;40: 157. Gauthier B, Robb M, Gaudet F, Ginsburg GS, McPherson R. Character-
1618 –1626. ization of a cholesterol response element (CRE) in the promoter of the
138. Langer C, Huang Y, Cullen P, Wiesenhütter B, Mahley RW, Assmann cholesteryl ester transfer protein gene: functional role of the transcription
G, von Eckardstein A. Endogenous apolipoprotein E modulates choles- factors SREBP-1a, -2, and YY1. J Lipid Res. 1999;40:1284–1293.
terol efflux and cholesterol ester hydrolysis mediated by high-density 158. Luo Y, Tall AR. Sterol upregulation of human CETP expression in vitro
lipoprotein-3 and lipid-free apolipoproteins in mouse peritoneal macro- and in transgenic mice by an LXR element. J Clin Invest. 2000;105:
phages. J Mol Med. 2000;78:217–227. 513–520.
139. Fidge N. High density lipoprotein receptors, binding proteins, and 159. Guerin M, Lassel TS, Le Goff W, Farnier M, Chapman MJ. Action of
ligands. J Lipid Res. 1999 40;187–201. atorvastatin in combined hyperlipidemia: preferential reduction of cho-
140. Acton S, Rigotti A, Landschulz KT, Xu S, Hobbs HH, Krieger M. lesteryl ester transfer from HDL to VLDL1 particles. Arterioscler
Identification of scavenger receptor SR-BI as a high density lipoprotein Thromb Vasc Biol. 2000;20:189 –197.
receptor. Science. 1996;271:518 –520. 160. Föger B, Ritsch A, Doblinger A, Wessels H, Patsch JR. Relationship of
141. Graf GA, Connell PM, van der Westhuyzen DR, Smart EJ. The class B, plasma cholesteryl ester transfer protein to HDL cholesterol: studies in
type I scavenger receptor promotes the selective uptake of high density normotriglyceridemia and moderate hypertriglyceridemia. Arterioscler
lipoprotein cholesterol ethers into caveolae. J Biol Chem. 1999;274: Thromb Vasc Biol. 1996;16:1430 –1436.
12043–12048. 161. Christison JK, Rye KA, Stocker R. Exchange of oxidized cholesteryl
142. Gu X, Trigatti B, Xu S, Acton S, Babitt J, Krieger M. The efficient linoleate between LDL and HDL mediated by cholesteryl ester transfer
cellular uptake of high density lipoprotein lipids via scavenger receptors protein. J Lipid Res. 1995;36:2017–2026.
class B type I requires not only receptor-mediated surface binding but 162. Inazu A, Koizumi J, Mabuchi H. Cholesteryl ester transfer protein and
also receptor-specific lipid transfer mediated by its extracellular domain. atherosclerosis. Curr Opin Lipidol. 2000;11:389 –396.
J Biol Chem. 1998;273:26338 –26348. 163. Hirano K, Yamashita S, Kuga Y, Sakai N, Nozaki S, Kihara S, Arai T,
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

143. Connelly MA, Klein SM, Azhar S, Abumrad NA, Williams DL. Com- Yanagi K, Takami S, Menju M, et al. Atherosclerotic disease in marked
parison of class B scavenger receptors, CD36 and scavenger receptor BI hyperalphalipoproteinemia: combined reduction of cholesteryl ester
(SR-BI) shows that both receptors mediate high density lipoprotein- transfer protein and hepatic triglyceride lipase. Arterioscler Thromb
cholesteryl ester selective uptake but SR-BI exhibits a unique Vasc Biol. 1995;15:1849 –1856.
enhancement of cholesteryl ester uptake. J Biol Chem. 1999;274:41– 47. 164. Bruce C, Sharp DS, Tall AR. Relationship of HDL and coronary heart
144. Rodrigueza WV, Thuahnai ST, Temel RE, Lund-Katz S, Phillips MC, disease to a common amino acid polymorphism in the cholesteryl ester
Williams DL. Mechanism of scavenger receptor class B type I-mediated transfer protein in men with and without hypertriglyceridemia. J Lipid
selective uptake of cholesteryl esters from high density lipoprotein to Res. 1998;39:1071–1078.
165. Agerholm-Larsen B, Nordestgaard BG, Steffensen R, Jensen G,
adrenal cells. J Biol Chem. 1999;274:20344 –20350.
Tybjærg-Hansen A. Elevated HDL cholesterol is a risk factor for ische-
145. Lambert G, Chase MB, Dugi K, Bensadoun A, Brewer HB Jr,
mic heart disease in white women when caused by a common mutation
Santamarina-Fojo S. Hepatic lipase promotes the selective uptake of
in the cholesteryl ester transfer protein gene. Circulation. 2000;101:
high density lipoprotein-cholesteryl esters via the scavenger receptors
1907–1912.
B1. J Lipid Res. 1999;40:1294 –1303.
166. Zhong S, Sharp DS, Grove JS, Bruce C, Yano K, Curb JD, Tall AR.
146. Collet X, Tall AR, Serajuddin H, Guendouzi K, Royer L, Oliveira H,
Increased coronary heart disease in Japanese-American men with
Barbaras R, Jiang XC, Francone OL. Remodeling of HDL by CETP in
mutation in the cholesteryl ester transfer protein gene despite increased
vivo and by CETP and hepatic lipase in vitro results in enhanced uptake
HDL levels. J Clin Invest. 1996;97:2917–2923.
of HDL CE by cells expressing scavenger receptor B-I. J Lipid Res.
167. Ordovas JM, Cupples A, Corella D, Otvos JD, Osgood D, Martinez A,
1999;40:1185–1193.
Lahoz C, Coltell O, Wilson PWF, Schaefer EJ. Association of cho-
147. Arai T, Rinninger F, Varban L, Fairchild, Huntress V, Liang CP, Chen
lesteryl ester protein TaqIB polymorphism with variations in lipoprotein
W, Seo T, Deckelbaum R, Huszar D, et al. Decreased selective uptake
subclasses and coronary heart disease risk: the Framingham Study.
of high density lipoprotein cholesteryl esters in apolipoprotein E
Arterioscler Thromb Vasc Biol. 2000;20:1323–1329.
knockout mice. Proc Natl Acad Sci U S A. 1999;96:12050 –12055. 168. Rittershaus CW, Miller DP, Thomas LJ, Picard MD, Honan CM,
148. Trigatti B, Rayburn H, Vinals M, Braun A, Miettinen H, Penman M, Emmett CD, Pettey CL, Aldari H, Hammond RA, Beattie DT, et al.
Hertz M, Schrenzel M, Amigo L, Rigotti A, et al. Influence of the high Vaccine induced antibodies inhibit CETP activity in vivo and reduce
density lipoprotein receptor SR-BI on reproductive and cardiovascular aortic lesions in a rabbit model of atherosclerosis. Arterioscler Thromb
pathophysiology. Proc Natl Acad Sci U S A. 1999;96:9322–9327. Vasc Biol. 2000;20:2106 –2112.
149. Kozarsky KF, Donahee MH, Rigotti A, Iqbal SN, Edelman ER, Krieger 169. Marotti KR, Castle CK, Boyle TP, Lin AH, Murray RW, Melchior GW.
M. Overexpression of the HDL receptor SR-BI alters plasma HDL and Severe atherosclerosis in transgenic mice expressing simian cholesteryl
bile cholesterol levels. Nature. 1997;387:414 – 417. ester transfer protein. Nature. 1993;364:73–75.
150. Arai T, Wang N, Bezouevski M, Welch C, Tall AR. Decreased athero- 170. Hayek T, Masucci, Magoulas L, Jiang X, Walsh A, Rubin E, Breslow
sclerosis in heterozygous low density lipoprotein receptor-deficient mice JL, Tall AR. Decreased early atherosclerotic lesions in hypertriglyceri-
expressing the scavenger receptor BI transgene. J Biol Chem. 1999;274: demic mice expressing cholesteryl ester transfer protein transgene.
2366 –2371. J Clin Invest. 1995;96:2071–2074.
151. Kozarsky KF, Donahee MH, Glick JM, Krieger M, Rader DJ. Gene 171. Plump AS, Masucci-Magoulas L, Bruce C, Bisgaier CL, Breslow JL,
transfer and hepatic overexpression of the HDL receptor SR-BI reduces Tall AR. Increased atherosclerosis in apoE and LDL receptor gene
atherosclerosis in the cholesterol-fed LDL receptor– deficient mouse. knock-out mice as a result of human cholesteryl ester transfer protein
Arterioscler Thromb Vasc Biol. 2000;20:721–727. transgene expression. Arterioscler Thromb Vasc Biol. 1999;19:
152. Fluiter K, van der Westhuijzen DR, van-Berkel TJ. In vivo regulation of 1105–1110.
scavenger receptor BI and the selective uptake of high density 172. Föger B, Chase M, Amar MJ, Vaisman BL, Shamburek RD, Paigen B,
lipoprotein cholesteryl esters in rat liver parenchymal and Kupffer cells. Fruchart, Najib J, Paiz JA, Koch CA, et al. Cholesteryl ester transfer
J Biol Chem. 1998;273:8434 – 8438. protein corrects dysfunctional high density lipoproteins and reduces
153. Morton RE. Cholesteryl ester transfer protein and its plasma regulator: aortic atherosclerosis in lecithin cholesterol acyltransferase transgenic
lipid transfer inhibitor protein. Curr Opin Lipidol. 1999;10:321–327. mice. J Biol Chem. 1999;274:36912–36920.
154. Ikewaki K, Rader DJ, Sakamoto T, Nishiwaki M, Wakimoto N, Schaefer 173. Herrera VL, Makrides SC, Xie HX, Adari H, Krauss RM, Ryan US,
JR, Ishikawa T, Fairwell T, Zech LA, Nakamura H, et al. Delayed Ruiz-Opazo N. Spontaneous combined hyperlipidemia, coronary heart
catabolism of high density lipoprotein apolipoproteins A-I and A-II in disease and decreased survival in Dahl salt-sensitive hypertensive rats
human cholesteryl ester transfer protein deficiency. J Clin Invest. 1993; transgenic for human cholesteryl ester transfer protein. Nat Med. 1999;
92:1650 –1658. 5:1383–1389.
155. Wang X, Driscoll DM, Morton RE. Molecular cloning and expression of 174. Conelly PW, Hegele RA. Hepatic lipase deficiency. Crit Rev Clin Lab
lipid transfer inhibitor protein reveals its identity with apolipoprotein F. Sci. 1998;35:547–572.
J Biol Chem. 1999;274:1814 –1820. 175. Santamarina-Fojo S, Haudenschild C, Amar M. The role of hepatic
156. Chouinard RA Jr, Luo Y, Osborne TF, Walsh A, Tall AR. Sterol lipase in lipoprotein metabolism and atherosclerosis. Curr Opin Lipidol.
regulatory element binding protein-1 activates the cholesteryl ester 1998;9:211–219.
von Eckardstein et al HDLs and Arteriosclerosis 27

176. Thuren T, Wilcox RW, Sisson P, Waite M. Hepatic lipase hydrolysis of on cholesterol efflux into plasma and lipoproteins of normal, apoli-
lipid monolayers: regulation by apolipoproteins. J Biol Chem. 1991;266: poprotein A-I–, and apolipoprotein E– deficient mice. Arterioscler
4853– 4861. Thromb Vasc Biol. 1997;17:2010 –2019.
177. Weng W, Brandenburg NA, Zhong S, Halkias J, Wu L, Jiang XC, Tall 198. Rubin EM, Krauss RM, Spangler EA, Verstuyft JG, Clift S. Inhibition
A, Breslow JL. ApoA-II maintains HDL levels in part by inhibition of of early atherogenesis in transgenic mice by human apolipoprotein AI.
hepatic lipase: studies in apoA-II and hepatic lipase double knockout Nature. 1991;353:265–267.
mice. J Lipid Res. 1999;40:1064 –1070. 199. Duverger N, Kruth H, Emmanuel F, Caillaud JM, Viglietta C, Castro G,
178. Garcia A, Barbaras R, Collet X, Bogyo A, Chap H, Perret B. High-density Tailleux A, Fievet C, Fruchart JC, Houdebine LM, et al. Inhibition of
lipoprotein 3 receptor-dependent endocytosis pathway in a human hepatoma atherosclerosis development in cholesterol-fed human apolipoprotein
cell line (HepG2). Biochemistry. 1996;35:13064–13071. A-I-transgenic rabbits. Circulation. 1996;94:713–717.
179. Brinton EA. Oral estrogen replacement therapy in postmenopausal 200. Plump AS, Scott CJ, Breslow JL. Human apolipoprotein A-I gene
women selectively raises levels and production rates of lipoprotein A-I expression increases high density lipoprotein and suppresses atheroscle-
and lowers hepatic lipase activity without lowering the fractional cat- rosis in the apolipoprotein E-deficient mouse. Proc Natl Acad Sci U S A.
abolic rate. Arterioscler Thromb Vasc Biol. 1996;16:431– 440. 1997;91:9607–9611.
180. Tan KC, Shiu SW, Pang RW, Kung AW. Effects of testosterone 201. Dansky HM, Charlton SA, Barlow CB, Tamminen M, Smith JD, Frank JS,
replacement on HDL subfractions and apolipoprotein A-I containing Breslow JL. Apo A-I inhibits foam cell formation in apo E-deficient mice
lipoproteins. Clin Endocrinol (Oxf). 1998;48:187–194. after monocyte adherence to endothelium. J Clin Invest. 1999;104:31–39.
181. Dugi KA, Brandauer K, Schmidt N, Ramacher D, Kreuzer J. Low 202. Benoit P, Emmanuel F, Caillaud JM, Bassinet L, Castro G, Gallix P,
hepatic lipase activity is a risk factor for coronary artery disease. Ath- Fruchart JC, Branellec D, Denefle P, Duverger N. Somatic gene transfer
erosclerosis. 2000;151:126. Abstract. of human apoA-I inhibits atherosclerosis progression in mouse models.
182. Mezdour H, Jones R, Dengremont C, Castro G, Maeda N. Hepatic lipase Circulation. 1999;99:105–110.
deficiency increases plasma cholesterol but reduces susceptibility to 203. Tangirala RK, Tsukamoto K, Chun SH, Usher D, Pure E, Rader DJ.
atherosclerosis in apolipoprotein E deficient mice. J Biol Chem. 1997; Regression of atherosclerosis induced by liver-directed gene transfer of
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

272:13570 –13575. apolipoprotein A-I in mice. Circulation. 1999;100:1816 –1822.


183. Busch SJ, Barnhart RL, Martin GA, Fitzgerald MC, Yates MT, Mao SJ, 204. Atger V, de la Llera Moya M, Bamberger M, Francone O, Cosgrove P,
Thomas CE, Jackson RL. Human hepatic triglyceride lipase expression Tall A, Walsh A, Moatti N, Rothblat G. Cholesterol efflux potential of
reduces high density lipoprotein and aortic cholesterol in cholesterol-fed sera from mice expressing human cholesteryl ester transfer protein
transgenic mice. J Biol Chem. 1994;269:13376 –13382. and/or human apolipoprotein AI. J Clin Invest. 1995 96;2613–2622.
205. Eriksson M, Carlson LA, Miettinen TA, Angelin B. Stimulation of fecal steroid
184. Jaye M, Lynch KJ, Krawiec J, Marchadier D, Maugeais C, Doan K,
excretion after infusion of recombinant proapolipoprotein A-I: potential reverse
South V, Amin D, Perrone M, Rader DJ. A novel endothelial-derived
cholesterol transport in humans Circulation. 1999;100:594–598.
lipase that modulates HDL metabolism. Nat Genet. 1999;21:424 – 428.
206. Duverger N, Tremp G, Caillaud JM, Emmanuel F, Castro G, Fruchart
185. Hirata K, Dichek HL, Cioffi JA, Leeper NJ, Quintana L, Kronmal GS,
JC, et al. Protection against atherogenesis in mice mediated by human
Cooper AD, Quertermous T. Cloning of unique lipase from endothelial cells
apolipoprotein A-IV. Science. 1996;273:966 –968.
extends the lipase gene family. J Biol Chem. 1999;274:14170–14175.
207. Cohen RD, Castellani LW, Qiao JH, Van-Lenten BJ, Lusis AJ, Reue K.
186. Moestrup SK, Kozyraki R, Kristiansen M, Kaysen JH, Rasmussen HH,
Reduced aortic lesions and elevated high density lipoprotein levels in
Brault D, Pontillon F, Goda FO, Christensen EI, Hammond TG, et al. The
transgenic mice overexpressing mouse apoA-IV. J Clin Invest. 1997;99:
intrinsic factor-vitamin B12 receptor and target of teratogenic antibodies is
1906 –1916.
a megalin-binding peripheral membrane protein with homology to devel-
208. Kuivenhoven JA, Pritchard H, Hill J, Frohlich J, Assmann G, Kastelein
opmental proteins. J Biol Chem. 1998 27;273:5235–5242.
JJ. The molecular pathology of lecithin:cholesterol acyltransferase
187. Mahley RW. Apolipoprotein E: cholesterol transport protein with
(LCAT) deficiency syndromes. J Lipid Res. 1997;38:191–205.
expanding role in cell biology. Science. 1988;240:622– 630. 209. Serfaty-Lacrosniere C, Civeira F, Lanzberg A, Isaia P, Berg J, Janus ED,
188. Assmann G, von Eckardstein A, Brewer HB Jr. Familial high density Smith MP Jr, Pritchard PH, Frohlich J, Lees RS, et al. Homozygous Tangier
lipoprotein deficiency: Tangier disease. In: Scriver CA, Beaudet AL, Sly disease and cardiovascular disease. Atherosclerosis. 1994;107:85–98.
WS, Valle D, eds. The Metabolic and Molecular Bases of Inherited 210. Sakai N, Vaisman BL, Koch CA, Hoyt RF Jr, Meyn SM, Talley GD,
Disease. 7th ed. New York, NY: McGraw-Hill; 1995:2053–2072. Paiz JA, Brewer HB Jr, Santamarina-Fojo S. Targeted disruption of the
189. DeLamatre JG, Sarphie TG, Archibold RC, Hornick CA. Metabolism of mouse lecithin:cholesterol acyltransferase (LCAT) gene: generation of a
apoE-free high density lipoproteins in rat hepatoma cells: evidence for a new animal model for human LCAT deficiency. J Biol Chem. 1997;14;
retroendocytic pathway. J Lipid Res. 1990;31:191–202. 272:7506 –7510.
190. Kambouris AM, Roach PD, Calvert GD, Nestel PJ. Retroendocytosis of 211. Berard AM, Foger B, Remaley A, Shamburek R, Vaisman BL, Talley G,
high density lipoproteins by the human hepatoma cell line, HepG2. Paigen B, Hoyt RF, Marcovina S, Brewer HB, et al. High plasma HDL
Arteriosclerosis. 1990;10:582–590. concentrations associated with enhanced atherosclerosis in transgenic
191. Silver DL, Wang N, Tall AR. Defective HDL particle uptake in ob/ob mice overexpressing lecithin-cholesteryl acyltransferase. Nat Med.
hepatocytes causes decreased recycling, degradation, and selective lipid 1997;3:744 –749.
uptake. J Clin Invest. 2000;105:151–159. 212. Hoeg JM, Santamarina-Fojo S, Berard AM, Cornhill JF, Herderick EE,
192. Silver DL, Jiang XC, Tall AR. Increased high density lipoprotein Feldman SH, Haudenschild CC, Vaisman BL, Hoyt RF Jr, Demosky SJ
(HDL), defective hepatic catabolism of ApoA-I and ApoA-II, and Jr, et al. Overexpression of lecithin:cholesterol acyltransferase in
decreased ApoA-I mRNA in ob/ob mice: possible role of leptin in transgenic rabbits prevents diet-induced atherosclerosis. Proc Natl Acad
stimulation of HDL turnover. J Biol Chem. 1999;274:4140 – 4146. Sci U S A. 1996;93:11448 –11453.
193. Funke H. Familial HDL deficiency syndromes. In: Jacotot B, Mathe D, 213. Schultz JR, Verstuyft JG, Gong EL, Nichols AV, Rubin EM. Protein
Fruchart JC, eds. Atherosclerosis XI. Singapore: Elsevier Science; 1997: composition determines the anti-atherogenic properties of HDL in
713–731. Excerpta Medica, Vol 1155. transgenic mice. Nature. 1993;365:762–764.
194. Li H, Reddick RL, Maeda N. Lack of apoA-I is not associated with 214. Warden CH, Hedrick CC, Qiao JH, Castellani LW, Lusis AJ. Athero-
increased susceptibility to atherosclerosis in mice. Arterioscler Thromb. sclerosis in transgenic mice overexpressing apolipoprotein A-II.
1993;13:1814 –1821. Science. 1993;261:469 – 472.
195. Voyiaziakis E, Goldberg IJ, Plump AS, Rubin EM, Breslow JL, Huang LS. 215. Escola-Gil JC, Marzal-Casacuberta A, Julve-Gil J, Ishida BY, Ordonez-
ApoA-I deficiency causes both hypertriglyceridemia and increased atheroscle- Llanos J, Chan L, et al. Human apolipoprotein A-II is a pro-atherogenic
rosis in human apoB transgenic mice. J Lipid Res. 1998;39: 313–321. molecule when it is expressed in transgenic mice at a level similar to that
196. von Eckardstein A, Huang Y, Wu S, Noseda G, Assmann G. Reverse in humans: evidence of a potentially relevant species-specific interaction
cholesterol transport in plasma of patients with different forms of with diet. J Lipid Res. 1998;39:457– 462.
familial high density lipoprotein deficiency. Arterioscler Thromb Vasc 216. Tailleux A, Bouly M, Luc G, Castro G, Caillaud M, Hennuyer N, Pulain
Biol. 1995;15:690 –701. B, Fruchart JC, Duverger N, Fiévet C. Decreased susceptibility to diet
197. Huang Y, Langer C, Raabe M, Wiesenhütter B, Wu S, Seedorf U, induced atherosclerosis in human apolipoprotein A-II transgenic mice.
Maeda N, Assmann G, von Eckardstein A. Effects of genotype and diet Arterioscler Thromb Vasc Biol. In press.
Downloaded from http://atvb.ahajournals.org/ by guest on December 6, 2017

High Density Lipoproteins and Arteriosclerosis: Role of Cholesterol Efflux and Reverse
Cholesterol Transport
Arnold von Eckardstein, Jerzy-Roch Nofer and Gerd Assmann

Arterioscler Thromb Vasc Biol. 2001;21:13-27


doi: 10.1161/01.ATV.21.1.13
Arteriosclerosis, Thrombosis, and Vascular Biology is published by the American Heart Association, 7272
Greenville Avenue, Dallas, TX 75231
Copyright © 2001 American Heart Association, Inc. All rights reserved.
Print ISSN: 1079-5642. Online ISSN: 1524-4636

The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://atvb.ahajournals.org/content/21/1/13

Data Supplement (unedited) at:


http://atvb.ahajournals.org/content/suppl/2001/01/02/21.1.13.DC1

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published
in Arteriosclerosis, Thrombosis, and Vascular Biology can be obtained via RightsLink, a service of the
Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for
which permission is being requested is located, click Request Permissions in the middle column of the Web
page under Services. Further information about this process is available in the Permissions and Rights
Question and Answer document.

Reprints: Information about reprints can be found online at:


http://www.lww.com/reprints

Subscriptions: Information about subscribing to Arteriosclerosis, Thrombosis, and Vascular Biology is online
at:
http://atvb.ahajournals.org//subscriptions/

Вам также может понравиться