875
Views
23
CrossRef citations to date
0
Altmetric
Review

Discrete roles of apoA‐I and apoE in the biogenesis of HDL species: Lessons learned from gene transfer studies in different mouse models

, , , , , & show all
Pages 14-28 | Published online: 08 Jul 2009

Abstract

Using adenovirus‐mediated gene transfer in apolipoprotein A‐I (apoA‐I)‐deficient mice, we have established that apoA‐I mutations inhibit discrete steps in a pathway that leads to the biogenesis and remodeling of high‐density lipoprotein (HDL).

To this point, five discrete categories of apoA‐I mutants have been characterized that may affect the interactions of apoA‐I with ATP‐binding cassette superfamily A, member 1 (ABCA1) or lecithin:cholesterol acyl transferase (LCAT) or may influence the plasma phospholipid transfer protein activity or may cause various forms of dyslipidemia.

Biogenesis of HDL is not a unique property of apoA‐I. Using adenovirus‐mediated gene transfer of apoE in apoA‐I‐ or ABCA1‐deficient mice, we have established that apolipoprotein E (apoE) also participates in a novel pathway of biogenesis of apoE‐containing HDL particles. This process requires the functions of the ABCA1 lipid transporter and LCAT, and it is promoted by substitution of hydrophobic residues in the 261 to 269 region of apoE by Ala.

The apoE‐containing HDL particles formed in the circulation may have atheroprotective properties. ApoE‐containing HDL may also have important biological functions in the brain that confer protection from Alzheimer's disease.

Abbreviations
ABCA1=

ATP‐binding cassette superfamily A, member 1

ABCA1−/−=

ABCA1‐deficient

apoA‐I=

apolipoprotein A‐I

apoA‐I−/−=

apoA‐I‐deficient mice

apoE=

apolipoprotein E

apoE−/−=

apoE‐deficient

ATP=

adenosine triphosphate

cAMP=

cyclic adenosine monophosphate

CE=

cholesteryl esters

CETP=

cholesteryl ester transfer protein

EM=

electron microscopy

FPLC=

fast pressure liquid chromatography

GFP=

green fluorescence protein

HDL=

high‐density lipoprotein

IDL=

intermediate‐density lipoprotein

LCAT=

lecithin:cholesterol acyl transferase

LDL=

low‐density lipoprotein

pfu=

plaque‐forming unit

PLTP=

phospholipid transfer protein

SR‐BI=

scavenger receptor class B type I

SR‐BI−/−=

SR‐BI‐deficient

TC=

total cholesterol

VLDL=

very‐low‐density lipoprotein

WT=

wild type

Introduction

ApoA‐I participates in several steps in the biogenesis and catabolism of HDL

High‐density lipoprotein (HDL) is synthesized and catabolized through a complex pathway shown in Figure , Citation1. HDL assembly is initiated by an ATP‐binding cassette superfamily A, member 1 (ABCA1)‐mediated transfer of cellular phospholipids and cholesterol to extracellular lipid‐poor apolipoprotein A‐I (apoA‐I). Through a series of intermediate steps that are not fully understood, lipidated apoA‐I is gradually converted to discoidal particles and subsequently to spherical α‐HDL particles by the action of lecithin:cholesterol acyl transferase (LCAT). Under normal conditions human plasma contains 3%–5% preβ‐HDL particles and 95%–97% α‐HDL particles Citation2, Citation3, whereas discoidal HDL is not detectable in normal human plasma. Following synthesis, HDL interacts with the HDL receptor scavenger receptor class B type I (SR‐BI), and this interaction mediates selective uptake of cholesterol esters and other lipids by the cell Citation4, Citation5, as well as bidirectional movement of unesterified cholesterol Citation6–9. Cellular cholesterol may also be effluxed to HDL by the cell surface transporter ABCG1 Citation10. Finally, hydrolysis of lipids of HDL is mediated by various lipases (lipoprotein lipase, hepatic lipase, endothelial lipase), and exchange of lipids by the phospholipid transfer protein (PLTP) and by the cholesteryl ester transfer protein (CETP). Lipid‐free apoA‐I secreted by the liver, intestine, and few other tissues Citation11 may also interact with ABCA1 present in peripheral cells and tissues and thus contribute to lipid efflux and potentially to HDL biogenesis.

Figure 1 The pathway of biogenesis(A) and catabolism (B) of HDL. Numbers 1–11 indicate key cell membrane or plasma proteins shown to influence HDL levels or composition. They are: 1) apolipoprotein A‐I; 2) ATP binding cassette transporter A1; 3) lecithin:cholesterol acyl transferase; 4) cholesteryl ester transfer protein; 5) hepatic lipase; 6) endothelial lipase; 7) phospholipid transfer protein; 8) lipoprotein lipase; 9) scavenger receptor class B type I; 10) LDL receptor; 11) ATP binding cassette transporter G1 (ABCG1). The figure is modified from reference Citation1. C: Secondary structure of apoA‐I based on high‐resolution X‐ray crystallography Citation58. The domains of apoA‐I that affect its various biological functions are shown in colors as follows. Pink and blue colors indicate regions that affect interactions of apoA‐I with ABCA1 and LCAT, respectively. Red indicates the region associated with inhibition of PLTP activity and dyslipidemia. Green (and green asterisks) indicates regions and amino acids associated with hypertriglyceridemia. Gray asterisks indicate amino acids that are involved in interactions of apoA‐I with SR‐BI.

Figure 1 The pathway of biogenesis(A) and catabolism (B) of HDL. Numbers 1–11 indicate key cell membrane or plasma proteins shown to influence HDL levels or composition. They are: 1) apolipoprotein A‐I; 2) ATP binding cassette transporter A1; 3) lecithin:cholesterol acyl transferase; 4) cholesteryl ester transfer protein; 5) hepatic lipase; 6) endothelial lipase; 7) phospholipid transfer protein; 8) lipoprotein lipase; 9) scavenger receptor class B type I; 10) LDL receptor; 11) ATP binding cassette transporter G1 (ABCG1). The figure is modified from reference Citation1. C: Secondary structure of apoA‐I based on high‐resolution X‐ray crystallography Citation58. The domains of apoA‐I that affect its various biological functions are shown in colors as follows. Pink and blue colors indicate regions that affect interactions of apoA‐I with ABCA1 and LCAT, respectively. Red indicates the region associated with inhibition of PLTP activity and dyslipidemia. Green (and green asterisks) indicates regions and amino acids associated with hypertriglyceridemia. Gray asterisks indicate amino acids that are involved in interactions of apoA‐I with SR‐BI.

To probe the pathway of biogenesis and catabolism of HDL, we have used adenovirus‐mediated gene transfer of apoA‐I mutants in apoA‐I‐deficient and other mouse models. The expectation was that these apoA‐I mutants will inhibit discrete steps of the HDL pathway and either lead to rapid catabolism or accumulation of intermediate products. The first part of this review will provide examples of characteristic mutations of apoA‐I (Figure ) that affect different steps of the HDL biogenesis and catabolism Citation12–17. The second part will describe a novel pathway of biogenesis of apolipoprotein E (apoE)‐containing HDL and how certain apoE mutations promote formation of spherical apoE‐containing HDL Citation18–20.

Search for mutations in apoA‐I with defective ABCA1‐mediated cholesterol efflux that may prevent the biogenesis of HDL

To explore the mechanism of ABCA1‐mediated efflux of cellular lipid to apoA‐I, we expressed in vitro wild‐type (WT) apoA‐I and numerous variants carrying point mutations or deletions. The apoA‐I mutants thus generated were used as lipid acceptors either in cultured cells expressing endogenous ABCA1 (J774 macrophages activated by a cyclic adenosine monophosphate (cAMP) analog), or in HEK293 cells transfected with a cDNA encoding ABCA1, and were used for in vitro ABCA1‐mediated lipid efflux and apoA‐I/ABCA1 chemical cross‐linking experiments. The questions addressed were: whether apoA‐I interacts directly or indirectly with ABCA1 to promote cholesterol and phospholipid efflux; which domains of apoA‐I are required for ABCA1‐dependent lipid efflux; and how lipid efflux correlates with apoA‐I/ABCA1 cross‐linking.

These in vitro studies showed that ABCA1‐mediated cholesterol and phospholipid efflux was moderately decreased by amino‐terminal deletions and was diminished by carboxy‐terminal deletions in which residues 220–231 were removed (Figure ; Table ) Citation12. Chemical cross‐linking/immunoprecipitation studies showed that the relative ability of apoA‐I mutants to promote ABCA1‐mediated lipid efflux correlated with the ability of these mutants to be cross‐linked efficiently to ABCA1 Citation21.

Table I. In vivo phenotypes of mice expressing apoA‐I deletion and point mutants and changes in the in vitro functions of these apoA‐I mutants Citation12–17, Citation59, Citation60.

Figure 2 A: cAMP‐dependent (ABCA1‐mediated) cholesterol efflux in J774 mouse macrophages. The experiment was performed as described Citation12. B: FPLC profile of total plasma cholesterol of apoA‐I−/− mice expressing the WT apoA‐I, or the carboxy‐terminal mutants apoA‐I[Δ(185–243)], apoA‐I[Δ(220–243)], apoA‐I[Δ(232–243)], apoA‐I[Glu191Ala/His193Ala/Lys195Ala], or the control protein GFP. Plasma samples were obtained from mice infected with 1×109 pfu of the recombinant adenoviruses expressing the WT or mutant forms of apoA‐I or the control protein GFP 4 days postinfection. The experiment was performed as described Citation16. C: Electron microscopy pictures of the HDL fractions obtained by density gradient ultracentrifugation from the plasma of apoA‐I−/− mice expressing the WT or mutant forms of apoA‐I or the control protein GFP. The experiment was performed as described Citation16. D: Two‐dimensional gel electrophoresis and Western blotting analysis of plasma of apoA‐I−/− mice expressing the WT, the point mutant or the truncated mutant forms of apoA‐I described above, or the control protein GFP. HDL subpopulations containing apoA‐I were detected using antihuman apoA‐I antibodies Citation16.

Figure 2 A: cAMP‐dependent (ABCA1‐mediated) cholesterol efflux in J774 mouse macrophages. The experiment was performed as described Citation12. B: FPLC profile of total plasma cholesterol of apoA‐I−/− mice expressing the WT apoA‐I, or the carboxy‐terminal mutants apoA‐I[Δ(185–243)], apoA‐I[Δ(220–243)], apoA‐I[Δ(232–243)], apoA‐I[Glu191Ala/His193Ala/Lys195Ala], or the control protein GFP. Plasma samples were obtained from mice infected with 1×109 pfu of the recombinant adenoviruses expressing the WT or mutant forms of apoA‐I or the control protein GFP 4 days postinfection. The experiment was performed as described Citation16. C: Electron microscopy pictures of the HDL fractions obtained by density gradient ultracentrifugation from the plasma of apoA‐I−/− mice expressing the WT or mutant forms of apoA‐I or the control protein GFP. The experiment was performed as described Citation16. D: Two‐dimensional gel electrophoresis and Western blotting analysis of plasma of apoA‐I−/− mice expressing the WT, the point mutant or the truncated mutant forms of apoA‐I described above, or the control protein GFP. HDL subpopulations containing apoA‐I were detected using antihuman apoA‐I antibodies Citation16.

A functional relationship between apoA‐I/ABCA1 cross‐linking and lipid efflux was also seen in the concurrent inhibition of both apoA‐I cross‐linking to ABCA1 and lipid efflux by two small molecule inhibitors: block lipid transport‐4 (BLT4) and (1‐(2‐methoxy‐phenyl)‐3‐naphthalen‐2‐yl‐urea) (glyburide) Citation22. All these in vitro data are consistent with a lipid efflux mechanism that involves direct interaction between apoA‐I and ABCA1, although other earlier studies suggested that protein‐protein interactions may not be required for lipid efflux Citation23.

In the in vivo studies, apoA‐I‐deficient (apoA‐I−/−) mice were infected with 1 to 2×109 plaque‐forming units (pfu) of an adenovirus expressing WT apoA‐I or the various apoA‐I mutants that were previously studied for ABCA1‐mediated lipid efflux and ABCA1 cross‐linking. Four to five days following gene transfer, lipid analyses, gel filtration chromatography, and two‐dimensional gel electrophoretic analyses were performed on the plasma of the apoA‐I−/− mice expressing the WT or mutant apoA‐I forms Citation12, Citation16, Citation24. Hepatic apoA‐I mRNA levels were also determined to ensure comparable expression of the mutant genes. These experiments showed that the WT apoA‐I, the apoA‐I[Δ(232–243)], and the apoA‐I[Glu191A/His193A/Lys195A] generated HDL in vivo as determined by fast pressure liquid chromatography (FPLC) fractionation (Figure ) Citation16. Electron microscopy (EM) analysis showed that the HDL consisted of spherical particles (Figure  I–III; Table ) Citation16. In contrast, apoA‐I[Δ(185–243)] and apoA‐I[Δ(220–243)] generated few HDL particles that were similar in numbers with the particles found in the plasma of apoA‐I−/− mice that express the green fluorescence protein (GFP) (Figure  IV–VI) Citation16, Citation24. Two‐dimensional gel electrophoresis showed that the majority of HDL derived from apoA‐I[Δ(232–243)] and apoA‐I[Glu191Ala/His193Ala/Lys195Ala] consisted of particles with α electrophoretic mobility and a small fraction consisted of particles with β electrophoretic mobility (Figure  I–III) Citation16. In contrast, the carboxy‐terminal deletion mutants apoA‐I[Δ(185–243)] and apoA‐I[Δ(220–243)] formed very little HDL, which in the case of apoA‐I[Δ(185–243)] contained exclusively preβ1‐HDL particles (Figure  IV) and in the case of apoA‐I[Δ(220–243)] contained preβ‐HDL and a few α‐HDL particles (Figure  V) Citation16. These findings suggested that carboxy‐terminal deletion mutants of apoA‐I that lacked the 220–231 region prevent the lipidation of apoA‐I, thus blocking the first step in the biogenesis of HDL Citation25. The combined in vitro and in vivo findings suggest that apoA‐I mutants that lack residues 220–231 are defective in the ABCA1‐mediated efflux of cellular phospholipids and cholesterol, cross‐link poorly to ABCA1, and fail to form discoidal or spherical HDL in vivo but form exclusively or predominantly preβ‐HDL particles. Furthermore, the apoA‐I[Δ(232–243)] mutant that retains the 220–231 region and the point mutant in the 191–195 region tested, promote normally the biogenesis of HDL.

Low HDL syndromes caused by apoA‐I mutations can be corrected by treatment with LCAT

To explain the etiology and find a mode of therapy for some forms of genetically determined low levels of HDL, we have generated recombinant adenoviruses expressing naturally occurring apoA‐I mutations and two bioengineered mutants and studied their properties in vitro and in vivo.

Two of the natural mutants studied, apoA‐I[Leu141Arg]PisaCitation26–29 and apoA‐I[Leu159Arg]FinCitation30–32, are associated with very low HDL and apoA‐I levels in humans Citation26, Citation28, Citation29, Citation31 and premature atherosclerosis Citation26, Citation32. The other two naturally occurring mutants, apoA‐I[Arg151Cys]ParisCitation33, Citation34 and apoA‐I[Arg160Leu]OsloCitation35, are also associated with low HDL levels in humans but present milder phenotypes. The bioengineered mutants apoA‐I[Arg149Ala] and apoA‐I[Arg160Val/His162Ala] were selected because the substitution of the positively charged Arg by Ala at positions 149 and 160 reduces the positive electrostatic potential that exists in the 149–160 region of apoA‐I and diminishes the ability of the mutant forms to activate LCAT in vitroCitation36.

Our studies showed that all mutants were secreted efficiently from cells, had diminished capacity to activate LCAT, and near‐normal capacity to promote ABCA1‐mediated cholesterol efflux in vitro (Table ).

Gene transfer of the apoA‐I[Arg160Leu]Oslo, apoA‐I[Arg149Ala], apoA‐I[Arg151Cys]Paris, and apoA‐I[Arg160Val/His162Ala] mutants in apoA‐I−/− mice generated aberrant HDL phenotypes Citation15, Citation17. The total plasma cholesterol was significantly reduced (23%–55% as compared to mice expressing WT apoA‐I) (Table ). Plasma apoA‐I and HDL levels were decreased significantly compared to WT apoA‐I (Figure  I–IV; Table ) Citation15, Citation17. Electron microscopy of the HDL obtained from mice expressing the various apoA‐I mutants showed that apoA‐I[Arg151Cys]Paris formed mostly spherical and few discoidal particles (Figure  III), apoA‐I[Arg160Leu]Oslo and apoA‐I[Arg149Ala] formed a mixture of discoidal and spherical HDL particles (Figure  II and IV), and apoA‐I[Arg160Val/His162Ala] formed discoidal particles (Figure  V) Citation15, Citation17. The WT apoA‐I formed spherical HDL, whereas apoA‐I−/− mice expressing GFP formed few particles (Figure  I and VI). Two‐dimensional gel electrophoresis of plasma obtained from mice expressing the WT apoA‐I and the various apoA‐I mutants showed that apoA‐I[Arg160Leu]Oslo, apoA‐I[Arg149Ala] and apoA‐I[Arg160Val/His162Ala] promoted the formation of preβ1 and α4‐HDL subpopulations (Figure  II, IV, and V) Citation15, Citation17. ApoA‐I[Arg151Cys]Paris generated subpopulations of different sizes that migrate between preβ and α‐HDL (Figure  III) Citation17. The WT apoA‐I formed predominantly α‐HDL and small amounts of preβ‐HDL particles (Figure  I). Simultaneous treatment of mice with adenoviruses expressing any of the four mutants and human LCAT normalized the plasma apoA‐I levels as well as the HDL cholesterol levels (Figure  I–IV), promoted the formation of spherical HDL particles (Figure  VII–X), and also promoted the formation of mostly α‐HDL subpopulations of larger size (Figure  VI–IX) Citation15, Citation17. The LCAT treatment also increased the cholesteryl ester (CE)/total cholesterol (TC) ratio of HDL from 0.3 (without treatment) to 0.8 following treatment Citation15, Citation17.

Figure 3 A:I‐IV. FPLC profiles of total plasma cholesterol of apoA‐I mice expressing apoA‐I[Arg149Ala], apoA‐I[Arg151Cys]Paris, apoA‐I[Arg160Leu]Oslo and apoA‐I[Arg160Val/His162Ala] in the presence and absence of LCAT as indicated in the Figure. The FPLC profile of mice expressing WT apoA‐I is also shown Panels AI‐IV. The experiments were performed as described Citation15, Citation17. B: Electron microscopy pictures of the HDL fractions obtained by density gradient ultracentrifugation from the plasma of apoA‐I−/− mice expressing the WT or mutant forms of apoA‐I or the control protein GFP or the same apoA‐I forms along with LCAT as indicated in Panels BI‐X. Experiments were performed as described Citation15, Citation17. C: Two‐dimensional gel electrophoresis and Western blot analysis of plasma of apoA‐I−/− mice expressing the WT or mutant forms of apoA‐I alone or in the presence of LCAT as indicated in Panels CI‐IX. HDL subpopulations containing apoA‐I were detected using antihuman apoA‐I antibodies. Experiments were performed as described Citation15, Citation17.

Figure 3 A:I‐IV. FPLC profiles of total plasma cholesterol of apoA‐I mice expressing apoA‐I[Arg149Ala], apoA‐I[Arg151Cys]Paris, apoA‐I[Arg160Leu]Oslo and apoA‐I[Arg160Val/His162Ala] in the presence and absence of LCAT as indicated in the Figure. The FPLC profile of mice expressing WT apoA‐I is also shown Panels AI‐IV. The experiments were performed as described Citation15, Citation17. B: Electron microscopy pictures of the HDL fractions obtained by density gradient ultracentrifugation from the plasma of apoA‐I−/− mice expressing the WT or mutant forms of apoA‐I or the control protein GFP or the same apoA‐I forms along with LCAT as indicated in Panels BI‐X. Experiments were performed as described Citation15, Citation17. C: Two‐dimensional gel electrophoresis and Western blot analysis of plasma of apoA‐I−/− mice expressing the WT or mutant forms of apoA‐I alone or in the presence of LCAT as indicated in Panels CI‐IX. HDL subpopulations containing apoA‐I were detected using antihuman apoA‐I antibodies. Experiments were performed as described Citation15, Citation17.

The in vivo analysis of the apoA‐I[Leu141Arg]Pisa and apoA‐I[Leu159Arg]Fin mutants in apoA‐I−/−mice indicate that apoA‐I[Leu141Arg]Pisa and apoA‐I[Leu159Arg]Fin mutations appear to inhibit an early step in the biogenesis of HDL due to inefficient esterification of the cholesterol of the preβ1‐HDL particles by the endogenous LCAT (27,32,37). The LCAT insufficiency appears to result from depletion of the plasma LCAT mass Citation37. The defects in apoA‐I appear to promote rapid catabolism of newly lipidated apoA‐I as well as plasma LCAT and thus prevent the formation of either discoidal or spherical HDL particles Citation37. In contrast, the apoA‐I[Arg151Cys]Paris, apoA‐I[Arg160Leu]Oslo, and apoA‐I[Arg149Ala] allow formation of discoidal particles but, due to insufficiency of the endogenous LCAT, prevent the conversion of discoidal to spherical HDL Citation17. The correction of the aberrant HDL phenotypes for all mutants studied here by treatment with LCAT suggests a potential therapeutic intervention to correct HDL abnormalities that result from specific mutations in apoA‐I.

Mutations in apoA‐I may induce hypertriglyceridemia and other dyslipidemias

We have identified three mutations of apoA‐I that have a profound effect on the overall cholesterol and triglyceride homeostasis Citation13, Citation14. Two of these, apoA‐I[Δ(62–78)] and apoA‐I[Glu110Ala/Glu111Ala], caused combined hyperlipidemia, characterized by high plasma cholesterol and severe hypertriglyceridemia Citation13, Citation14. All the triglycerides and the majority of the excess cholesterol were distributed in very‐low‐density lipoprotein (VLDL)/intermediate‐density lipoprotein (IDL)‐sized lipoproteins. Density gradient ultracentrifugation showed that this VLDL/IDL fraction was enriched in apoA‐I (Figure ). Also the VLDL/IDL fraction had decreased levels of apoE and apoCII and increased levels of apoB‐48 (Figure ) Citation13, Citation14. The presence of apoA‐I and the depletion of apoCII in the VLDL/IDL fraction raised the possibility that the mutations were associated with inhibition of lipolysis in vivo. Coinfection of mice with adenoviruses expressing apoA‐I[Glu110Ala/Glu111Ala] and the human lipoprotein lipase reduced plasma and VLDL triglyceride levels but had a smaller effect on plasma cholesterol levels.

Figure 4 A: Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS‐PAGE) analysis of density gradient ultracentrifugation fractions of plasma obtained from apoA‐I−/− mice 4 days postinfection with adenoviruses expressing WT apoA‐I or the apoA‐I[Glu110Ala/Glu111Ala] mutant. The densities of the fraction are indicated on the top of the figure. ApoA‐I was detected by Western blotting using polyclonal antibody to human apoA‐I Citation13. B: Assessment of the apoprotein composition of VLDL/LDL isolated from plasma of mice infected with WT apoA‐I and apoA‐I[Glu110Ala/Glu111Ala] mutant. The apoproteins were detected by Western blotting using polyclonal antibodies to mouse apoB, apoE and apoCII Citation13. C: SDS‐PAGE analysis of density gradient ultracentrifugation fractions of plasma of apoA‐I−/− mice expressing the WT apoA‐I or the apoA‐I[Δ(89–99)] mutant. ApoA‐I was detected by staining with Coomassie Brilliant Blue. On the right side of the panel is shown the CE/TC ratio from a pool of lipoprotein fractions that correspond to the HDL region. Experiments were performed as described Citation14. D, E: Electron microscopy pictures of HDL fractions obtained from apoA‐I−/− mice expressing the WT apoA‐I (D) or the apoA‐I[Δ(89–99)] mutant (E) following density gradient ultracentrifugation of plasma. The photomicrographs were taken at 75,000× magnification and enlarged three times. The experiment was performed as described Citation14. F, G: Analysis of plasma obtained from mice expressing the WT apoA‐I (F) or the apoA‐I[Δ(89–99)] mutant (G) following two‐dimensional gel electrophoresis and Western blotting. The experiment was performed as described Citation14.

Figure 4 A: Sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS‐PAGE) analysis of density gradient ultracentrifugation fractions of plasma obtained from apoA‐I−/− mice 4 days postinfection with adenoviruses expressing WT apoA‐I or the apoA‐I[Glu110Ala/Glu111Ala] mutant. The densities of the fraction are indicated on the top of the figure. ApoA‐I was detected by Western blotting using polyclonal antibody to human apoA‐I Citation13. B: Assessment of the apoprotein composition of VLDL/LDL isolated from plasma of mice infected with WT apoA‐I and apoA‐I[Glu110Ala/Glu111Ala] mutant. The apoproteins were detected by Western blotting using polyclonal antibodies to mouse apoB, apoE and apoCII Citation13. C: SDS‐PAGE analysis of density gradient ultracentrifugation fractions of plasma of apoA‐I−/− mice expressing the WT apoA‐I or the apoA‐I[Δ(89–99)] mutant. ApoA‐I was detected by staining with Coomassie Brilliant Blue. On the right side of the panel is shown the CE/TC ratio from a pool of lipoprotein fractions that correspond to the HDL region. Experiments were performed as described Citation14. D, E: Electron microscopy pictures of HDL fractions obtained from apoA‐I−/− mice expressing the WT apoA‐I (D) or the apoA‐I[Δ(89–99)] mutant (E) following density gradient ultracentrifugation of plasma. The photomicrographs were taken at 75,000× magnification and enlarged three times. The experiment was performed as described Citation14. F, G: Analysis of plasma obtained from mice expressing the WT apoA‐I (F) or the apoA‐I[Δ(89–99)] mutant (G) following two‐dimensional gel electrophoresis and Western blotting. The experiment was performed as described Citation14.

The third mutation, apoA‐I[Δ(89–99)] Citation14, caused (relative to WT apoA‐I) hypercholesterolemia characterized by increased cholesterol and phospholipids in the VLDL/IDL/low‐density lipoprotein (LDL) size lipoproteins, normal triglyceride levels, and a substantially decreased CE/TC ratio in HDL and LDL (Figure ). The Δ(89–99) deletion also increased apoA‐I in the LDL, caused accumulation of discoidal HDL (compare Figure  with Figure ), and increased the levels of preβ1‐ relative to the α‐HDL subpopulation particles (compare Figure  with Figure ) Citation14. Strikingly, mice expressing the apoA‐I[Δ(89–99)] mutant exhibited PLTP activity that was only 32% of that of mice expressing the WT apoA‐I Citation14. It has been proposed that PLTP functions by linking the donor and acceptor lipoprotein particles thus facilitating the net transfer of phospholipids to HDL Citation38. Previous studies also have suggested that apoA‐I interacts physically with PLTP Citation39 and that this interaction may underlie the transfer of the phospholipids from the donor molecule to HDL.

Figure  shows the normal pathway of biogenesis and catabolism of HDL and various steps where the pathway can be affected and lead to dyslipidemia. This includes: 1) lack of synthesis of HDL due to mutations in ABCA1 or apoA‐I that prevent apoA‐I/ABCA1 interactions; 2) failure to synthesize discoidal or spherical HDL due to fast catabolism of minimally lipidated apoA‐I; 3) induction of hypertriglyceridemia by the apoA‐I[Δ(62–78)] and apoA‐I[Glu110Ala/Glu111Ala] mutants; 4) accumulation of discoidal HDL, inhibition of PLTP, and the induction of hypercholesterolemia by the apoA‐[Δ(89–99)] mutant; and 5) the accumulation of discoidal HDL due to the mutations in the 149–160 region of apoA‐I. The phenotypes produced by targeted mutagenesis of apoA‐I will be valuable for the detection of similar phenotypes in humans and can serve as diagnostic and prognostic markers of dyslipidemia and/or atherosclerosis.

Figure 5 Schematic representation of the defects in the HDL pathway that can account for 1) lack of HDL synthesis due to mutation in ABCA1 or carboxy‐terminal apoA‐I deletions that affect cholesterol efflux; 2) hypertriglyceridemia induced by the apoA‐I[Δ(62–78)] and apoA‐I[Glu110Ala/Glu111Ala] mutants; 3) the high plasma cholesterol levels, inhibition of PLTP activity, and accumulation of discoidal HDL particles that is induced by the apoA‐I[Δ(89–99)] mutant; and 4) accumulation of discoidal HDL due to LCAT deficiency or apoA‐I mutations that cause depletion of plasma LCAT and either prevent formation of discoidal and spherical HDL (apoA‐I[Leu141Arg]PISA, apoA‐I[Leu159Arg]Fin) or cause accumulation of discoidal HDL (apoA‐I[Arg149Ala], apoA‐I[Arg151Cys]Paris, apoA‐I[Arg160Leu]Oslo and apoA‐I[Arg160Val/His162Ala)]). PL = phospholipids; C = cholesterol; TC = total cholesterol; CE = cholesteryl esters.

Figure 5 Schematic representation of the defects in the HDL pathway that can account for 1) lack of HDL synthesis due to mutation in ABCA1 or carboxy‐terminal apoA‐I deletions that affect cholesterol efflux; 2) hypertriglyceridemia induced by the apoA‐I[Δ(62–78)] and apoA‐I[Glu110Ala/Glu111Ala] mutants; 3) the high plasma cholesterol levels, inhibition of PLTP activity, and accumulation of discoidal HDL particles that is induced by the apoA‐I[Δ(89–99)] mutant; and 4) accumulation of discoidal HDL due to LCAT deficiency or apoA‐I mutations that cause depletion of plasma LCAT and either prevent formation of discoidal and spherical HDL (apoA‐I[Leu141Arg]PISA, apoA‐I[Leu159Arg]Fin) or cause accumulation of discoidal HDL (apoA‐I[Arg149Ala], apoA‐I[Arg151Cys]Paris, apoA‐I[Arg160Leu]Oslo and apoA‐I[Arg160Val/His162Ala)]). PL = phospholipids; C = cholesterol; TC = total cholesterol; CE = cholesteryl esters.

Key messages

  • Apolipoprotein A‐I (apoA‐I) mutations inhibit discrete steps in the pathway of biogenesis and remodeling of high‐density lipoprotein (HDL).

  • The aberrant HDL phenotypes caused by naturally occurring apoA‐I mutations can be corrected by administration of lecithin:cholesterol acyl transferase (LCAT).

  • Apolipoprotein E (apoE) along with ATP‐binding cassette superfamily A, member 1 (ABCA1) and LCAT participate in the biogenesis of apoE‐containing HDL.

Novel pathway of biogenesis of apoE‐containing HDL

Using adenovirus‐mediated gene transfer in apoA‐I‐ or ABCA1‐deficient mice, we obtained unequivocal evidence that apoE participates in a novel pathway of biogenesis of apoE‐containing HDL particles that also requires the functions of the ABCA1 lipid transporter and LCAT Citation18. Infection of apoA‐I−/− mice with 2×109 pfu of an apoE4‐expressing adenovirus increased both HDL and the triglyceride‐rich VLDL/IDL/LDL fraction and generated discoidal HDL particles (compare Figure  with Figure ). ABCA1‐deficient (ABCA1−/−) mice treated similarly failed to form HDL particles (Figure ), suggesting that ABCA1 is essential for the biogenesis of apoE‐containing HDL. Combined infection of apoA‐I−/− mice with a mixture of adenoviruses expressing both apoE4 (2×109 pfu) and human LCAT (5×108 pfu) cleared the triglyceride‐rich lipoproteins, increased HDL, and converted the discoidal to spherical HDL (Figure ). Similar treatment of ABCA1−/− mice failed to promote formation of HDL, suggesting that LCAT is essential for the maturation of apoE‐containing HDL Citation18. Overall, the findings indicate that apoE has a dual functionality. In addition to its documented functions in the clearance of triglyceride‐rich lipoproteins, it participates in the biogenesis of apoE‐containing HDL in a process that is similar to that of apoA‐I (Figure ).

Figure 6 Electron microscopy pictures of the fractions corresponding to the HDL region obtained from apoA‐I−/− mice (A), apoA‐I−/− mice expressing apoE4 (B), ABCA1−/− mice (C), ABCA1−/− mice expressing apoE4 (D), and apoA‐I−/− mice expressing a combination of apoE4 and LCAT (E). Following density gradient ultracentrifugation, the fractions that float to the HDL region were obtained and analyzed by electron microscopy as described Citation18.

Figure 6 Electron microscopy pictures of the fractions corresponding to the HDL region obtained from apoA‐I−/− mice (A), apoA‐I−/− mice expressing apoE4 (B), ABCA1−/− mice (C), ABCA1−/− mice expressing apoE4 (D), and apoA‐I−/− mice expressing a combination of apoE4 and LCAT (E). Following density gradient ultracentrifugation, the fractions that float to the HDL region were obtained and analyzed by electron microscopy as described Citation18.

Figure 7 Diagram showing the participation of apoE in the chylomicron pathway(branch I), the participation of apoE in the biogenesis of apoE‐containing HDL (branch II), and the participation of apoA‐I in the biogenesis of apoA‐I‐containing HDL (branch III).

Figure 7 Diagram showing the participation of apoE in the chylomicron pathway(branch I), the participation of apoE in the biogenesis of apoE‐containing HDL (branch II), and the participation of apoA‐I in the biogenesis of apoA‐I‐containing HDL (branch III).

ApoE‐containing HDL may have antioxidant and anti‐inflammatory functions similar to those described for apoA‐I‐containing HDL, which may contribute to the atheroprotective properties of apoE Citation40–43. ApoE‐containing HDL may also have important biological functions in the brain Citation44.

ApoE mutants in the 261–269 region promote the formation of spherical apoE‐containing HDL

Overexpression of apolipoprotein E (apoE) induces hypertriglyceridemia in C57BL/6 or apoE‐deficient mice, which is abrogated by deletion of the carboxy‐terminal segment 260–299 Citation1, Citation19.

We have used adenovirus‐mediated gene transfer in apoA‐I−/− mice to test the effect of four sets of apoE mutations within the 261–269 region on the formation of spherical or discoidal apoE‐containing HDL as well as the induction of hypertriglyceridemia Citation19, Citation20.

A single amino acid substitution (apoE4[Phe265Ala]) induced hypertriglyceridemia in apoA‐I−/− mice and was associated with moderate HDL cholesterol levels (Figure  I, B I). A double substitution (apoE4[Leu261Ala,Trp264Ala]) induced milder hypertriglyceridemia and was associated with increased HDL cholesterol levels (Figure  II, B II). A triple substitution (apoE4[Leu261Ala/Trp264Ala/Phe265Ala]) or five residue substitutions (apoE4[Leu261Ala/Trp264Ala/Phe265Ala/Leu268Ala/Val269Ala]) did not induce hypertriglyceridemia and both were associated with greatly increased HDL cholesterol levels (Figure  III, B III and A IV, B IV, respectively) Citation19, Citation20. EM analysis of the HDL fractions showed that apoE4[Leu261Ala/Trp264Ala/Phe265Ala] and the apoE4[Leu261Ala/Trp264Ala/Phe265Ala/Leu268Ala/Val269Ala] formed spherical HDL (Figure  III, IV), apoE4[Phe265Ala] formed discoidal HDL (Figure I), and the apoE4[Leu261Ala/Trp264Ala] formed mostly spherical and few discoidal HDL particles (Figure  II) Citation20. The phenotype of apoE‐mutC carrying three amino acid substitions, in terms of lipid levels and HDL formation was similar in apoE4 as well as in apoE2 background Citation20.

Figure 8 Plasma cholesterol and FPLC profiles of apoA‐I−/− mice infected with recombinant adenoviruses expressing WT apoE4, apoE4‐mutA (one amino acid substitution), apoE4‐mutB (two amino acids substitutions), apoE4‐mutC (three amino acids substitutions), or apoE4‐mut1 (five amino acids substitutions) as indicated on the top of the Figure. ApoA‐I−/− mice were infected with 2×109 pfu of recombinant adenovirus, and serum samples were isolated 4 days postinfection as described Citation20. A: total plasma cholesterol and triglycerides; B: cholesterol FPLC profiles; C: electron microscopy pictures of HDL.

Figure 8 Plasma cholesterol and FPLC profiles of apoA‐I−/− mice infected with recombinant adenoviruses expressing WT apoE4, apoE4‐mutA (one amino acid substitution), apoE4‐mutB (two amino acids substitutions), apoE4‐mutC (three amino acids substitutions), or apoE4‐mut1 (five amino acids substitutions) as indicated on the top of the Figure. ApoA‐I−/− mice were infected with 2×109 pfu of recombinant adenovirus, and serum samples were isolated 4 days postinfection as described Citation20. A: total plasma cholesterol and triglycerides; B: cholesterol FPLC profiles; C: electron microscopy pictures of HDL.

These studies established that substitutions of Leu261, Trp264 and Phe265 by Ala in either the apoE4 or apoE2 background promoted formation of spherical apoE‐containing HDL and also prevented apoE‐induced hypertriglyceridemia. Bioengineered apoE variants that can promote formation of spherical apoE‐containing HDL may find therapeutic applications in the future for the prevention of atherosclerosis.

Functions of apoE‐containing HDL in the brain

ApoE is the only medium‐size apolipoprotein present in the brain Citation11, Citation45 and alone may contribute to lipid homeostasis in the brain Citation44. The interactions of apoE with ABCA1 in the brain promote cholesterol efflux and lead to the biogenesis of apoE‐containing HDL Citation25,44. Once formed, apoE‐containing HDL can interact with the HDL receptor SR‐BI, and this interaction leads to the uptake of cholesteryl esters and cholesterol efflux Citation46. ApoE‐containing HDL may further have antioxidant and anti‐inflammatory functions in the brain similar to those attributed to apoA‐I‐containing HDL Citation40–43. Existing evidence indicated that apoE‐containing HDL through receptor‐mediated processes may sequester Aβ away from the brain parenchyma and prevent amyloid deposition and plaque formation Citation47–53. In contrast, lipid‐free apoE that cannot bind to apoE‐recognizing receptors Citation54 may promote Aβ polymerization and plaque formation (Figure ) Citation55–57.

Figure 9 Schematic representation of the pathway of biogenesis of apoE‐containing HDL in the brain. The picture depicts a potential role of apoE‐containing HDL in lipid homeostasis in the brain and the removal of the amyloid peptide β (Aβ). It also depicts a potential role of lipid‐free apoE in the Aβ polymerization and formation of amyloid plaques.

Figure 9 Schematic representation of the pathway of biogenesis of apoE‐containing HDL in the brain. The picture depicts a potential role of apoE‐containing HDL in lipid homeostasis in the brain and the removal of the amyloid peptide β (Aβ). It also depicts a potential role of lipid‐free apoE in the Aβ polymerization and formation of amyloid plaques.

Materials and methods

Materials used and various assays, such as cholesterol efflux assays, cross‐linking of apoA‐I to ABCA1, lipid‐lipoprotein apoA‐I and apoE measurements have been described Citation12–21.

Acknowledgements

This work was supported by grants from the National Institutes of Health (HL48739 and HL68216), the 6th Framework Programme of the European Union (No. LSHM‐CT‐2006‐0376331), and a grant from the American Heart Association (SDG 0535443T). Alexander Vezeridis is a trainee of the Predoctoral Training Grant in Cardiovascular Biology (T32 HL07969).

References

  • Zannis V. I., Chroni A., Kypreos K. E., Kan H. Y., Cesar T. B., Zanni E. E., et al. Probing the pathways of chylomicron and HDL metabolism using adenovirus‐mediated gene transfer. Curr Opin Lipidol 2004; 15: 151–66
  • Fielding C. J., Fielding P. E. Molecular physiology of reverse cholesterol transport. J Lipid Res 1995; 36: 211–28
  • Nanjee M. N., Cooke C. J., Olszewski W. L., Miller N. E. Concentrations of electrophoretic and size subclasses of apolipoprotein A‐I‐containing particles in human peripheral lymph. Arterioscler Thromb Vasc Biol 2000; 20: 2148–55
  • Acton S., Rigotti A., Landschulz K. T., Xu S., Hobbs H. H., Krieger M. Identification of scavenger receptor SR‐BI as a high density lipoprotein receptor. Science 1996; 271: 518–20
  • Stangl H., Hyatt M., Hobbs H. H. Transport of lipids from high and low density lipoproteins via scavenger receptor‐BI. J Biol Chem 1999; 274: 32692–8
  • Ji Y., Jian B., Wang N., Sun Y., Moya M. L., Phillips M. C., et al. Scavenger receptor BI promotes high density lipoprotein‐mediated cellular cholesterol efflux. J Biol Chem 1997; 272: 20982–5
  • Gu X., Kozarsky K., Krieger M. Scavenger receptor class B, type I‐mediated [3H]cholesterol efflux to high and low density lipoproteins is dependent on lipoprotein binding to the receptor. J Biol Chem 2000; 275: 29993–30001
  • Liu T., Krieger M., Kan H. Y., Zannis V. I. The effects of mutations in helices 4 and 6 of apoA‐I on scavenger receptor class B type I (SR‐BI)‐mediated cholesterol efflux suggest that formation of a productive complex between reconstituted high density lipoprotein and SR‐BI is required for efficient lipid transport. J Biol Chem 2002; 277: 21576–84
  • Krieger M. Scavenger receptor class B type I is a multiligand HDL receptor that influences diverse physiologic systems. J Clin Invest 2001; 108: 793–7
  • Wang N., Lan D., Chen W., Matsuura F., Tall A. R. ATP‐binding cassette transporters G1 and G4 mediate cellular cholesterol efflux to high‐density lipoproteins. Proc Natl Acad Sci U S A 2004; 101: 9774–9
  • Zannis V. I., Cole F. S., Jackson C. L., Kurnit D. M., Karathanasis S. K. Distribution of apolipoprotein A‐I, C‐II, C‐III, and E mRNA in fetal human tissues. Time‐dependent induction of apolipoprotein E mRNA by cultures of human monocyte‐macrophages. Biochemistry 1985; 24: 4450–5
  • Chroni A., Liu T., Gorshkova I., Kan H. Y., Uehara Y., von Eckardstein A., et al. The central helices of apoA‐I can promote ATP‐binding cassette transporter A1 (ABCA1)‐mediated lipid efflux. Amino acid residues 220–231 of the wild‐type apoA‐I are required for lipid efflux in vitro and high density lipoprotein formation in vivo. J Biol Chem 2003; 278: 6719–30
  • Chroni A., Kan H. Y., Kypreos K. E., Gorshkova I. N., Shkodrani A., Zannis V. I. Substitutions of glutamate 110 and 111 in the middle helix 4 of human apolipoprotein A‐I (apoA‐I) by alanine affect the structure and in vitro functions of apoA‐I and induce severe hypertriglyceridemia in apoA‐I‐deficient mice. Biochemistry 2004; 43: 10442–57
  • Chroni A., Kan H. Y., Shkodrani A., Liu T., Zannis V. I. Deletions of Helices 2 and 3 of Human ApoA‐I Are Associated with Severe Dyslipidemia following Adenovirus‐Mediated Gene Transfer in ApoA‐I‐Deficient Mice. Biochemistry 2005; 44: 4108–17
  • Chroni A., Duka A., Kan H. Y., Liu T., Zannis V. I. Point mutations in apolipoprotein A‐I mimic the phenotype observed in patients with classical lecithin:cholesterol acyltransferase deficiency. Biochemistry 2005; 44: 14353–66
  • Chroni A., Koukos G., Duka A., Zannis V. I. The Carboxy‐Terminal Region of apoA‐I Is Required for the ABCA1‐Dependent Formation of alpha‐HDL But Not Prebeta‐HDL Particles in Vivo. Biochemistry 2007; 46: 5697–708
  • Koukos G., Chroni A., Duka A., Kardassis D., Zannis V. I. Naturally occurring and bioengineered apoA‐I mutations that inhibit the conversion of discoidal to spherical HDL: The abnormal HDL phenotypes can be corrected by treatment with LCAT. Biochem J 2007; 406: 167–74
  • Kypreos K. E., Zannis V. I. Pathway of biogenesis of apolipoprotein E‐containing HDL in vivo with the participation of ABCA1 and LCAT. Biochem J 2007; 403: 359–67
  • Kypreos K. E., Van Dijk K. W., Havekes L. M., Zannis V. I. Generation of a recombinant apolipoprotein E variant with improved biological functions: hydrophobic residues (LEU‐261, TRP‐264, PHE‐265, LEU‐268, VAL‐269) of apoE can account for the apoE‐induced hypertriglyceridemia. J Biol Chem 2005; 280: 6276–84
  • Drosatos K., Kypreos K. E., Zannis V. I. Residues Leu261, Trp264, and Phe265 account for apolipoprotein E‐induced dyslipidemia and affect the formation of apolipoprotein E‐containing high‐density lipoprotein. Biochemistry 2007; 46: 9645–53
  • Chroni A., Liu T., Fitzgerald M. L., Freeman M. W., Zannis V. I. Cross‐linking and lipid efflux properties of apoA‐I mutants suggest direct association between apoA‐I helices and ABCA1. Biochemistry 2004; 43: 2126–39
  • Nieland T. J., Chroni A., Fitzgerald M. L., Maliga Z., Zannis V. I., Kirchhausen T., et al. Cross‐inhibition of SR‐BI‐ and ABCA1‐mediated cholesterol transport by the small molecules BLT‐4 and glyburide. J Lipid Res 2004; 45: 1256–65
  • Chambenoit O., Hamon Y., Marguet D., Rigneault H., Rosseneu M., Chimini G. Specific docking of apolipoprotein A‐I at the cell surface requires a functional ABCA1 transporter. J Biol Chem 2001; 276: 9955–60
  • Reardon C. A., Kan H. Y., Cabana V., Blachowicz L., Lukens J. R., Wu Q., et al. In vivo studies of HDL assembly and metabolism using adenovirus‐mediated transfer of ApoA‐I mutants in ApoA‐I‐deficient mice. Biochemistry 2001; 40: 13670–80
  • Fitzgerald M. L., Morris A. L., Chroni A., Mendez A. J., Zannis V. I., Freeman M. W. ABCA1 and amphipathic apolipoproteins form high‐affinity molecular complexes required for cholesterol efflux. J Lipid Res 2004; 45: 287–94
  • Miccoli R., Bertolotto A., Navalesi R., Odoguardi L., Boni A., Wessling J., et al. Compound heterozygosity for a structural apolipoprotein A‐I variant, apo A‐I(L141R)Pisa, and an apolipoprotein A‐I null allele in patients with absence of HDL cholesterol, corneal opacifications, and coronary heart disease. Circulation 1996; 94: 1622–8
  • Miccoli R., Zhu Y., Daum U., Wessling J., Huang Y., Navalesi R., et al. A natural apolipoprotein A‐I variant, apoA‐I (L141R)Pisa, interferes with the formation of alpha‐high density lipoproteins (HDL) but not with the formation of pre beta 1‐HDL and influences efflux of cholesterol into plasma. J Lipid Res 1997; 38: 1242
  • Pisciotta L., Miccoli R., Cantafora A., Calabresi L., Tarugi P., Alessandrini P., et al. Recurrent mutations of the apolipoprotein A‐I gene in three kindreds with severe HDL deficiency. Atherosclerosis 2003; 167: 335–45
  • Navalesi R., Miccoli R., Odoguardi L., Funke H., von Eckardstein A., Wiebusch H., et al. Genetically determined absence of HDL‐cholesterol and coronary atherosclerosis. Lancet 1995; 346: 708–9
  • Gylling H., Relas H., Miettinen H. E., Radhakrishnan R., Miettinen T. A. Delayed postprandial retinyl palmitate and squalene removal in a patient heterozygous for apolipoprotein A‐IFIN mutation (Leu 159–>Arg) and low HDL cholesterol level without coronary artery disease. Atherosclerosis 1996; 127: 239–43
  • Miettinen H. E., Gylling H., Miettinen T. A., Viikari J., Paulin L., Kontula K. Apolipoprotein A‐IFin. Dominantly inherited hypoalphalipoproteinemia due to a single base substitution in the apolipoprotein A‐I gene. Arterioscler Thromb Vasc Biol 1997; 17: 83–90
  • Miettinen H. E., Jauhiainen M., Gylling H., Ehnholm S., Palomaki A., Miettinen T. A., et al. Apolipoprotein A‐IFIN (Leu159–>Arg) mutation affects lecithin cholesterol acyltransferase activation and subclass distribution of HDL but not cholesterol efflux from fibroblasts. Arterioscler Thromb Vasc Biol 1997; 17: 3021–32
  • Daum U., Langer C., Duverger N., Emmanuel F., Benoit P., Denefle P., et al. Apolipoprotein A‐I (R151C)Paris is defective in activation of lecithin: cholesterol acyltransferase but not in initial lipid binding, formation of reconstituted lipoproteins, or promotion of cholesterol efflux. J Mol Med 1999; 77: 614–22
  • Bruckert E., von Eckardstein A., Funke H., Beucler I., Wiebusch H., Turpin G., et al. The replacement of arginine by cysteine at residue 151 in apolipoprotein A‐I produces a phenotype similar to that of apolipoprotein A‐IMilano. Atherosclerosis 1997; 128: 121–8
  • Leren T. P., Bakken K. S., Daum U., Ose L., Berg K., Assmann G., et al. Heterozygosity for apolipoprotein A‐I(R160L)Oslo is associated with low levels of high density lipoprotein cholesterol and HDL‐subclass LpA‐I/A‐ II but normal levels of HDL‐subclass LpA‐I. J Lipid Res 1997; 38: 121–31
  • Roosbeek S., Vanloo B., Duverger N., Caster H., Breyne J., De Beun I., et al. Three arginine residues in apolipoprotein A‐I are critical for activation of lecithin:cholesterol acyltransferase. J Lipid Res 2001; 42: 31–40
  • Koukos G., Chroni A., Duka A., Kardassis D., Zannis V. I. Lecithin:Cholesterol Acyl Transferase Can Rescue the Abnormal Phenotype Produced by the Natural Apolipoprotein A‐I Mutations (Leu141Arg)Pisa and (Leu159Arg)FIN. Biochemistry 2007; 46: 10713–21
  • Huuskonen J., Olkkonen V. M., Jauhiainen M., Ehnholm C. The impact of phospholipid transfer protein (PLTP) on HDL metabolism. Atherosclerosis 2001; 155: 269–81
  • Pussinen P. J., Jauhiainen M., Metso J., Pyle L. E., Marcel Y. L., Fidge N. H., et al. Binding of phospholipid transfer protein (PLTP) to apolipoproteins A‐I and A‐II: location of a PLTP binding domain in the amino terminal region of apoA‐I. J Lipid Res 1998; 39: 152–61
  • Yuhanna I. S., Zhu Y., Cox B. E., Hahner L. D., Osborne‐Lawrence S., Lu P., et al. High‐density lipoprotein binding to scavenger receptor‐BI activates endothelial nitric oxide synthase. Nat Med 2001; 7: 853–7
  • Rader D. J. High‐density lipoproteins and atherosclerosis. Am J Cardiol 2002; 90: 62i–70i
  • Navab M., Hama S. Y., Anantharamaiah G. M., Hassan K., Hough G. P., Watson A. D., et al. Normal high density lipoprotein inhibits three steps in the formation of mildly oxidized low density lipoprotein: steps 2 and 3. J Lipid Res 2000; 41: 1495–508
  • Mineo C., Yuhanna I. S., Quon M. J., Shaul P. W. High density lipoprotein‐induced endothelial nitric‐oxide synthase activation is mediated by Akt and MAP kinases. J Biol Chem 2003; 278: 9142–9
  • Li X., Kypreos K., Zanni E. E., Zannis V. Domains of apoE required for binding to apoE receptor 2 and to phospholipids: Implications for the functions of apoE in the brain. Biochemistry 2003; 42: 10406–17
  • Boyles J. K., Pitas R. E., Wilson E., Mahley R. W., Taylor J. M. Apolipoprotein E associated with astrocytic glia of the central nervous system and with nonmyelinating glia of the peripheral nervous system. J Clin Invest 1985; 76: 1501–13
  • Chroni A., Nieland T. J., Kypreos K. E., Krieger M., Zannis V. I. SR‐BI mediates cholesterol efflux via its interactions with lipid‐bound ApoE. Structural mutations in SR‐BI diminish cholesterol efflux. Biochemistry 2005; 44: 13132–43
  • Cao D., Fukuchi K., Wan H., Kim H., Li L. Lack of LDL receptor aggravates learning deficits and amyloid deposits in Alzheimer transgenic mice. Neurobiol Aging 2006; 27: 1632–43
  • DeMattos R. B., Cirrito J. R., Parsadanian M., May P. C., O'Dell M. A., Taylor J. W., et al. ApoE and clusterin cooperatively suppress Abeta levels and deposition: evidence that ApoE regulates extracellular Abeta metabolism in vivo. Neuron 2004; 41: 193–202
  • Tanzi R. E., Moir R. D., Wagner S. L. Clearance of Alzheimer's Abeta peptide: the many roads to perdition. Neuron 2004; 43: 605–8
  • Shibata M., Yamada S., Kumar S. R., Calero M., Bading J., Frangione B., et al. Clearance of Alzheimer's amyloid‐ss(1–40) peptide from brain by LDL receptor‐related protein‐1 at the blood‐brain barrier. J Clin Invest 2000; 106: 1489–99
  • Deane R., Wu Z., Sagare A., Davis J., Du Y. S., Hamm K., et al. LRP/amyloid beta‐peptide interaction mediates differential brain efflux of Abeta isoforms. Neuron 2004; 43: 333–44
  • Zerbinatti C. V., Bu G. LRP and Alzheimer's disease. Rev Neurosci 2005; 16: 123–35
  • Van Uden E., Mallory M., Veinbergs I., Alford M., Rockenstein E., Masliah E. Increased extracellular amyloid deposition and neurodegeneration in human amyloid precursor protein transgenic mice deficient in receptor‐associated protein. J Neurosci 2002; 22: 9298–304
  • Ruiz J., Kouiavskaia D., Migliorini M., Robinson S., Saenko E. L., Gorlatova N., et al. The apoE isoform binding properties of the VLDL receptor reveal marked differences from LRP and the LDL receptor. J Lipid Res 2005; 46: 1721–31
  • Bales K. R., Verina T., Cummins D. J., Du Y., Dodel R. C., Saura J., et al. Apolipoprotein E is essential for amyloid deposition in the APP(V717F) transgenic mouse model of Alzheimer's disease. Proc Natl Acad Sci U S A 1999; 96: 15233–8
  • Burns M. P., Noble W. J., Olm V., Gaynor K., Casey E., LaFrancois J., et al. Co‐localization of cholesterol, apolipoprotein E and fibrillar Abeta in amyloid plaques. Brain Res Mol Brain Res 2003; 110: 119–25
  • Miao J., Vitek M. P., Xu F., Previti M. L., Davis J., Van Nostrand W. E. Reducing cerebral microvascular amyloid‐beta protein deposition diminishes regional neuroinflammation in vasculotropic mutant amyloid precursor protein transgenic mice. J Neurosci 2005; 25: 6271–7
  • Ajees A. A., Anantharamaiah G. M., Mishra V. K., Hussain M. M., Murthy H. M. Crystal structure of human apolipoprotein A‐I: insights into its protective effect against cardiovascular diseases. Proc Natl Acad Sci U S A 2006; 103: 2126–31
  • Scott B. R., McManus D. C., Franklin V., McKenzie A. G., Neville T., Sparks D. L., et al. The N‐terminal globular domain and the first class A amphipathic helix of apolipoprotein A‐I are important for lecithin:cholesterol acyltransferase activation and the maturation of high density lipoprotein in vivo. J Biol Chem 2001; 276: 48716–24
  • Sorci‐Thomas M. G., Thomas M., Curtiss L., Landrum M. Single repeat deletion in ApoA‐I blocks cholesterol esterification and results in rapid catabolism of delta6 and wild‐type ApoA‐I in transgenic mice. J Biol Chem 2000; 275: 12156–63

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.