SlideShare a Scribd company logo
1 of 10
Download to read offline
Free Radical Biology & Medicine 52 (2012) 1372–1381 
Contents lists available at SciVerse ScienceDirect 
Free Radical Biology & Medicine 
journal homepage: www.elsevier.com/locate/freeradbiomed 
Original Contribution 
Purified human paraoxonase-1 interacts with plasma membrane lipid rafts and 
mediates cholesterol efflux from macrophages 
Hicham Berrougui a,b, Soumaya Loued a,b,c, Abdelouahed Khalil a,b,c,⁎ 
a Research Center on Aging, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada QC J1H 4C4 
b Department of Medicine, Geriatrics Service, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada QC J1H 4C4 
c Programs of Physiology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC Canada J1H 4C4 
a r t i c l e i n f o a b s t r a c t 
Article history: 
Received 12 September 2011 
Revised 17 January 2012 
Accepted 23 January 2012 
Available online 1 February 2012 
Keywords: 
Paraoxonase-1 
HDL 
Cholesterol efflux 
Lipid raft 
Free radicals 
Macrophages 
Paraoxonase-1 (PON1) is a high-density lipoprotein (HDL)-associated serum enzyme thought to make a 
major contribution to the antioxidant and anti-inflammatory capacities of HDLs. However, the role of PON1 
in the modulation of cholesterol efflux is poorly understood. The aim of our study was to investigate the in-volvement 
of PON1 in the regulation of cholesterol efflux, especially the mechanism by which it modulates 
HDL-mediated cholesterol transport. The enrichment of HDL3 with human PON1 enhanced, in a dose-dependent 
manner, cholesterol efflux from THP-1 macrophage-like cells and ABCA1-enriched J774 macro-phages. 
Moreover, an additive effect was observed when ABCA1-enriched J774 macrophages were incubated 
with both PON1 and apo-AI. Interestingly, PON1 alone was able to mediate cholesterol efflux from J774 mac-rophages 
and to upregulate ABCA1 expression on J774 macrophages. Immunofluorescence measurement 
showed an increase in PON1 levels in the cytoplasm of J774 macrophages overexpressing ABCA1. PON1 
used an apo-AI-like mechanism to modulate cholesterol efflux from rapid and slow efflux pools derived 
from the lipid raft and nonraft domains of the plasma membrane, respectively. This was supported by the 
fact that ABCA1 protein was incrementally expressed by J774 macrophages within the first few hours of in-cubation 
with cholesterol-loaded J774 macrophages and that cyclodextrin significantly inhibited the capacity 
of PON1 to modulate cholesterol efflux from macrophages. This finding suggested that PON1 plays an impor-tant 
role in the antiatherogenic properties of HDLs and may exert its protective function outside the lipopro-tein 
environment. 
© 2012 Elsevier Inc. All rights reserved. 
It has long been recognized that the concentration of plasma high-density 
lipoproteins (HDLs) is inversely related to the risk of cardio-vascular 
disease. The atheroprotective effect of HDLs is largely attrib-uted 
to its key role in reverse cholesterol transport (RCT) whereby 
excess cholesterol from peripheral cells is transported back to the 
liver for excretion [1]. Macrophages possess a number of mechanisms 
to regulate the equilibrium between cholesterol uptake/synthesis and 
export, including transport mechanisms that promote the efflux of 
excess cholesterol to extracellular acceptors [1]. Although cholesterol 
efflux from macrophages makes up only a small fraction of overall 
flux through the RCT pathway, it is considered a relevant component 
of atheroprotection [1]. HDL-mediated cholesterol efflux is the natu-ral 
rate-limiting step of RCT [2] and occurs via three pathways. The 
first pathway is aqueous diffusion by which free cholesterol mole-cules 
spontaneously desorb from the plasma membrane, diffuse 
through the aqueous phase, and become adsorbed on acceptor 
particles by collision [3]. The second pathway involves scavenger re-ceptor 
class B type I (SR-BI)-mediated bidirectional free cholesterol 
exchanges depending on the cholesterol gradient, this pathway medi-ates 
cholesterol efflux to a wide variety of cholesterol acceptors [4]. 
The third pathway involves the ATP-binding cassette receptors 
ABCA1 and ABCG1, which mediate cholesterol efflux in a unidirec-tional 
manner to lipid-poor apolipoprotein-AI (apo-AI) and to other 
subfamily members of HDL, respectively [5–7]. 
The rate of cholesterol efflux, however, depends also on the capac-ity 
of cells to release excess cholesterol and of plasma acceptors to 
transport the released cholesterol, which is determined by the con-centration 
and physical and functional states of HDLs and apo-AI [2]. 
Apo-AI is a major protein component of HDL and is a critical ele-ment 
in cholesterol metabolism. Apo-AI reacts specifically with 
ABCA1 to generate nascent HDLs, which are then enriched with ester-ified 
cholesterol by LCAT (activated by apo-AI) to form mature spher-ical 
HDLs [8]. A number of other proteins confer additional properties 
on HDLs. Vaisar et al. identified 48 proteins in HDLs associated with 
clinically significant cardiovascular disease [9]. Paraoxonase 1 
(PON1) is one of the 8 most abundant HDL-associated proteins and 
may have atheroprotective properties [9]. PON1 has been extensively 
⁎ Corresponding author at: Research Center on Aging, 1036 rue Belvédère Sud, Sher-brooke, 
QC, Canada J1H 4C4. Fax: +1 819 829 7141. 
E-mail address: Abdelouahed.Khalil@USherbrooke.ca (A. Khalil). 
0891-5849/$ – see front matter © 2012 Elsevier Inc. All rights reserved. 
doi:10.1016/j.freeradbiomed.2012.01.019
H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1373 
studied with respect to its association with cardiovascular risk, oxida-tive 
stress, and inflammation [10]. 
PON1 is mainly synthesized in the liver and 95% is transferred to 
HDLs via an SR-BI-mediated mechanism. Small amounts of plasma 
PON1 (5%) are associated with chylomicrons and very low density li-poproteins 
[11–13]. In vitro studies have shown that PON1 inhibits 
low-density lipoprotein (LDL) and HDL lipid peroxidation and may 
be a primary determinant of the anti-inflammatory capacity of HDLs 
[14,15]. PON1 also attenuates oxidized LDL uptake via scavenger re-ceptor 
CD-36 and directly reduces oxidative stress in macrophages 
[16,17]. It also inhibits oxidized LDL-induced MCP-1 production via 
a mechanism that is probably related to its capacity to hydrolyze 
some activated phospholipids as well as lipid peroxide [18]. Cys284 
of the free sulfhydryl of PON1 is required for its atheroprotective ef-fect 
and is thought to be the active site for its antioxidant activity 
[19]. We previously showed that exposure of purified human PON1 
to oxygen free radicals induced a significant decrease in PON1 para-oxonase 
and antioxidant activities, which was accompanied by a de-crease 
in free sulfhydryl groups [19]. 
PON1 expression is inversely correlated with atherosclerotic de-velopment 
in animal models [20,21]. PON1 accumulates in arterial 
walls as human plaque progresses from fatty streaks into advanced 
lesions. This process is associated with macrophages and seems to 
protect against the increasing oxidation associated with plaque pro-gression 
[22]. Treating human carotid lesion extracts with recombi-nant 
PON1 significantly decreases the oxidative potential of the 
extracts [23]. These properties may be the main explanation for the 
beneficial effect of PON1 on the atherosclerotic process [24]. 
Recent research has focused on investigating the potential anti-atherogenic 
role played by PON1 in HDL-mediated cholesterol efflux 
from macrophages [25]. Rosenblat et al. suggested that PON1 in-creases 
lysophosphatidylcholine (lyso-PC) formation, which in turn 
stimulates HDL binding and HDL-mediated cholesterol efflux [26]. 
The H115Q and H134Q histidine residues of PON1 seem to be re-quired 
for this process [25]. 
Lipid rafts are cholesterol- and sphingolipid-rich lateral domains 
containing approximately double the molar percentages of these 
lipids and much lower proportions of protein than other areas of 
the plasma membrane [27]. Research on the role of lipid rafts in cho-lesterol 
transport and homeostasis has concentrated mainly on 
caveolin-1, a structural protein in a subset of lipid rafts (caveolae) 
that directly interacts with cholesterol [28]. Although caveolin-1 is a 
useful marker for isolating rafts and seems to be involved in intracel-lular 
cholesterol transport, there is some controversy concerning the 
role of caveolae cholesterol in cholesterol efflux [29], given that 
caveolin-1 is not expressed in primary human macrophages or J774 
macrophages [30] and that cholesterol efflux in these cells normally 
occurs via apo-AI [31]. Gaus et al. recently demonstrated that choles-terol 
exported to apo-AI from the major slow efflux pool comes from 
nonraft regions of the plasma membrane and that the interaction of 
apo-AI with lipid rafts is required to stimulate this efflux [28]. 
We investigated the role of purified human PON1 on HDL-mediated 
and non-HDL-mediated cholesterol efflux and propose an 
explanation of the mechanism by which PON1 enhances cholesterol 
efflux, especially via its interaction with lipid and nonlipid raft 
domains. We provide further evidence that the PON1–lipid raft inter-action 
is an essential event in PON1-mediated cholesterol efflux. 
Material and methods 
Acetic acid, sulfuric acid, sodium phosphate, thiobarbituric acid, 
n-butanol, methanol, ethanol, n-isopropanol, hexane, ammonium 
hydroxide, chloroform, and methanol were purchased from Fisher 
Scientific (Montreal, QC, Canada). N-ethylmaleimide (NEM), 1,1,3,3,- 
tetraethoxypropane, ethylenediaminetetraacetic acid (EDTA), 1,6- 
diphenyl-1,3,5-hexatriene, phosphatidylcholine, sphingomyelin, 8- 
(4-chlorophenylthio)adenosine 3′:5′-cyclic monophosphate (cAMP), 
methyl-β-cyclodextrin (MβCD), and [3H]cholesterol were from 
Sigma (St. Louis,MO, USA). THP-1 and J774 cellswere fromtheAmerican 
Type Culture Collection (Manassas, VA, USA). RPMI 1640 and Dulbecco's 
modified Eagle's medium (DMEM) were from Invitrogen Canada 
(Burlington, ON, Canada). Fetal bovine serum(FBS) was fromWisent 
(St-Bruno, QC, Canada). 
Subjects 
Plasma samples were obtained from young healthy volunteers 20 
to 30 years of age. They were all healthy, normolipidemic non-smokers 
and nonobese. None had clinical or laboratory signs of hy-pertension, 
inflammation, or diabetes, and all had normal thyroid 
function test results. None were taking medications or oral antioxi-dant 
supplements. The ethics committee of the Sherbrooke Geriatric 
University Institute approved the study. All the volunteers provided 
written informed consent. 
Blood collection 
After overnight fasting, 80-ml blood samples were collected in 
EDTA or citrate vacuum tubes. The plasma was separated by low-speed 
centrifugation (1000 g), and 15 ml was used immediately to 
isolate lipoproteins. The remaining plasma was stored at −80 °C 
until used for the PON1 purification procedure. 
Lipoprotein isolation 
Human plasma was collected and HDL and HDL3 were isolated by 
sequential ultracentrifugation according to the method of Sattler et al. 
[32]. In brief, whole HDL (1.063bdb1.19 g/ml) and HDL3 (1.125b 
db1.21 g/ml) were isolated by respectively 2 and 4 h of ultracentrifu-gation 
in a Beckman TLA 100.4 rotor (100,000 rpm at 15 °C). Isolated 
lipoproteins were dialyzed overnight at 4 °C in 10 mM sodium 
phosphate buffer (pH 7). Protein concentrations in the samples 
were measured using a commercial assay (Bio-Rad, Mississauga, ON, 
Canada). 
Cell cultures 
Human THP-1 monocytes and J774 macrophages were grown in 
RPMI 1640, and Fu5AH hepatoma cells were grown in DMEM. The 
media were supplemented with 10% heat-inactivated FBS, 50 nM 
2-mercaptoethanol (only for THP-1 cells), 2 mM L-glutamine, 100 U/ 
ml penicillin, and 1.5 mg/ml glucose in a humidified atmosphere 
(5% CO2 and 95% air) at 37 °C. The THP-1 monocytes (105 cells/cm2 
in six-well plates) were incubated in RPMI–FBS containing 10 ng/ml 
phorbol 12-myristate 13-acetate for 96 h to induce differentiation 
into adherent macrophage-like cells. 
Paraoxonase purification 
PON1 was purified using blue agarose and DEAE chromatography 
as described by Gan et al. [33], with some modifications. Briefly, plas-ma 
was mixed with blue agarose (Cibadron Blue 3GA; Sigma–Aldrich, 
Oakville, ON, Canada) in a solution containing 3 M NaCl, 50 mM Tris– 
HCl buffer (pH 8), 1 mM CaCl2, and 5 μM EDTA. The PON1 was eluted 
with 0.1% deoxycholate. The blue agarose-eluted PON1 was further 
purified by anion-exchange chromatography (DEAE-Biogel; Sigma) 
using a NaCl linear gradient. The paraoxonase and arylesterase activities 
of the purified PON1 were measured as described previously [33,34]. 
The purity of PON1 was verified by SDS–PAGE.
1374 H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 
Cholesterol efflux measurements 
THP-1 macrophage-like cells, J774 macrophages, and Fu5AH 
cells were incubated in fresh medium containing 2 μCi/ml [3H]cho-lesterol 
for 24 h. Labeled cells were washed and equilibrated in 
serum-free medium containing 1% bovine serum albumin (BSA) 
for an additional 12 h. To evaluate the interaction between PON1 
and the ABCA1 receptor, [3H]cholesterol-loaded J774 macrophages 
were washed three times and incubated with 1% BSA in DMEM 
alone (control) or with DMEM containing 0.3 mM cAMP for 12 h 
to yield ABCA1-enriched cells (cAMP stimulates ABCA1 gene tran-scription 
and surface protein expression) [35]. After the equilibra-tion 
period, the THP-1 macrophage-like cells and J774 
macrophages were washed three times before being incubated 
with various cholesterol acceptors (native or oxidized PON1, HDL3, 
apo-AI) depending on the experimental design. The cells were sedi-mented 
by centrifugation (350 g for 10 min) and were lysed in 
0.1 M NaOH. The counts per minute (cpm) in the supernatant and 
cell lysates were determined using a liquid scintillation counter. 
Cholesterol efflux (radiolabeled cholesterol released from cells) 
was calculated using the following formula: (radioactivity (cpm) 
in supernatant/radioactivity (cpm) in cells+medium)×100. 
Inactivation of PON1 
PON1 was inactivated by exposure to oxygen free radicals pro-duced 
by γ-radiolysis of water. Exposure of PON1 to oxygen free rad-icals 
has been shown to oxidize PON1 and inhibits its enzymatic 
activity without causing protein fragmentation [19]. PON1 was also 
inactivated by heat (1 h at 70 °C) or with 10 μM NEM, which scav-enges 
the free sulfhydryl group on PON1 (Cys284) [19]. The inactiva-tion 
of PON1 was confirmed by measuring PON1 activity. 
Lipid raft extraction 
Lipid rafts were extracted as previously described [36]. Briefly, 
cells were resuspended in 300 μl of ice-cold lipid raft lysis buffer 
and mixed with 300 μl of 85% sucrose in Hepes-buffered saline solu-tion 
(pH 6.9). The samples were overlaid with 1 ml of 35% sucrose 
and then 300 μl of 5% sucrose. The preparations were centrifuged at 
200,000 g at 4 °C for 16 h using a Beckman TLA-100.4 rotor (Beckman 
Instruments, Montreal, QC, Canada). Nine 200-μl fractions were col-lected 
from the top of the gradient. They were separated on 10% 
SDS–PAGE gels and were transferred to nitrocellulose membranes, 
which were blotted and incubated with anti-flotillin antibody. 
Immunofluorescence microscopy 
J774 macrophages were seeded in eight-well glass culture slides 
(BD Falcon, Bedford, MA, USA) at a concentration of 2.5×105 cells 
per well and were incubated with 0.3 mM cAMP for 12 h to generate 
ABCA1-enriched cells. The enriched cells were incubated with 20 U/ 
ml PON1 for 4 h, washed twice with PBS, fixed with 4% formaldehyde 
for 10 min, and incubated with 0.1% Triton X-100 in PBS for 2 min. 
After being washed with PBS, the cells were incubated with blocking 
solution (5% goat serum, 5% BSA, and 0.01% Tween 20) and then with 
primary antibody in blocking solution for 16 h at 4 °C. Alexa 488- 
labeled goat anti-mouse IgG was used as the secondary antibody. 
The slides were washed, stained with DAPI, and mounted in Vecta-shield 
(Vector Laboratories, Burlington, ON, Canada) to visualize the 
shapes of the nuclei. 
Western blotting 
Cell proteins were solubilized in RIPA buffer. Identical amounts of 
cell lysate protein were separated on 10% SDS–PAGE gels and were 
transferred to nitrocellulose membranes. The membranes were 
blocked with 5% fat-free powdered milk in TBST (10 mM Tris–HCl, 
pH 7.4, 150 mM NaCl, 0.1% Tween 20) and then incubated with 
ABCA1, ABCG1, or SR-BI primary antibodies (Abcam, Cambridge, 
MA, USA). After three washes with TBST, the membranes were incu-bated 
with horseradish peroxidase-conjugated goat anti-rabbit, goat 
anti-mouse IgG, or rabbit anti-goat IgG (Sigma), and the signals 
were visualized using the ECL Western blotting system (GE Health-care, 
Piscataway, NJ, USA). 
Coimmunoprecipitation assays 
Coimmunoprecipitation was used to confirm the formation of 
ABCA1–PON1 complex in lysates from J774 macrophages. cAMP 
(0.3 mM) was used to stimulate ABCA1 overexpression on J774 mac-rophages. 
PON1 (20 U/ml) was added to ABCA1-enriched or none-nriched 
J774 macrophages for 3 h and then cell lysates were 
prepared with RIPA buffer and protease inhibitor cocktail. Immuno-precipitations 
were carried out using Dynabeads protein G (Invitro-gen) 
according to the manufacturer's protocol. Briefly, 1.5 mg of 
Dynabeads was incubated with 5 μg of ABCA1 monoclonal antibody 
(mAb) for 60 min. After overnight incubation at 4 °C of Dynabeads– 
mAb complex with cell lysates, beads were washed three times 
with PBS and then 20 μl of 2× sample buffer (100 mM Tris, pH 6.8, 
40 g/L SDS, 200 ml/L glycerol, 20 mg/ml bromophenol blue, 0.05% 2- 
mercaptoethanol) was added to the beads. The sample/supernatant 
was separated from the beads by placing the tube on a magnet and 
then loaded on the SDS–PAGE (10%) gel. After being transferred to a 
polyvinylidene difluoride membrane, PON1 was detected using an 
anti-PON1 mouse monoclonal antibody (Abcam). 
Quantitative RT-PCR 
Total RNA from J774 macrophages treated with 20 U/ml PON1 for 
0, 1, 3, or 16 h was extracted using RNeasy extraction mini kits (Invi-trogen) 
and treated with DNase I (Qiagen, Mississauga, ON, Canada) 
according to the manufacturer's protocol. Two micrograms of RNA 
was transcribed using Reverse Transcriptase Superscript II (Invitro-gen). 
The expression of the ABCA1 gene was normalized to the corre-sponding 
amount of β-actin. Amplifications were performed using 
the following primers (sense and antisense): ABCA1, 5′-TCATCTT-CATCTGCTTCCAGC- 
3′ and 5′-GTGCTGGGGATCTTGAACAC-3′; β-actin, 
5′-GAACGGTGAAGGTGACA-3′ and 5′-TAGAGAGAAGTGGGGTGG-3′. 
The quantitative PCR assays were performed using the Stratagene 
MX3005P system (Agilent Technologies, Mississauga, ON, Canada) 
and Brilliant II SYBR Green QPCR Master Mix (Agilent). The qPCR as-says 
were performed using 25 ng of template cDNA. Samples were in-cubated 
at 95 °C for 10 min followed by 45 cycles using the following 
conditions: 95 °C for 40 s, 56 °C for 40 s, and 72 °C for 40 s. All reac-tions 
were run in triplicate for each replicate, and the average values 
were used for quantification purposes. The relative quantities of tar-get 
genes were determined using the ΔΔCt method. Briefly, the Ct 
(threshold cycle) values of target genes were normalized to an en-dogenous 
control gene, β-actin (ΔCt=Ct target−Ct β-actin), and were 
compared with a calibrator (ΔΔCt=ΔCt sample−ΔCt calibrator). Relative 
expression (RQ) was calculated using a sequence detection system 
(MxPro and QPCR software; Agilent) and the formula RQ=2−ΔΔCt. 
Statistical analysis 
Values are expressed as means ± SEM. A one-way analysis of var-iance 
was used for multiple comparisons. A linear regression analysis 
was used to assess the association between two continuous variables. 
Statistical analyses were performed using Prism 5.0 version software. 
A p value of b0.05 was considered statistically significant.
Results 
PON1 has an effect on HDL-mediated cholesterol efflux 
HDL-associated proteins other than apo-AI can stimulate HDL-mediated 
cholesterol efflux [37,38]. The aim of this study was to in-vestigate 
the role of PON1 in cholesterol efflux. Loading THP1 
macrophage-like cells with [3H]cholesterol and purified human plas-ma 
PON1 5, 10, and 20 U/ml (corresponding to 12.5, 25, and 50 μg/ml 
of protein) increased HDL3-mediated cholesterol efflux from THP-1 
macrophage-like cells by 20.8, 24, and 63% (pb0.05), respectively 
(Fig. 1A). The increase in cholesterol efflux from THP1 macrophage-like 
cells was dependent on the concentration of PON1 (r2=0.91, 
pb0.05, data not shown). It is noteworthy that the physiological con-centration 
of human plasma PON1 is between 50 and 100 μg/ml pro-tein 
[39,40]. 
Although PON1 significantly increased cholesterol efflux to J774 
macrophages (Fig. 1B) in a concentration-dependent manner 
(r2=0.94, pb0.05, data not shown), stimulating J774 macrophages 
with cAMP to induce ABCA1 overexpression significantly potentiated 
the effect (r2=0.97, pb0.05). A comparison of the slopes of the two 
correlations shows that cholesterol efflux increased 1.82-fold when 
PON1 was added to cAMP-stimulated J774 macrophages compared 
to unstimulated cells (1.03±0.11 vs 0.55±0.1, respectively, 
pb0.05). This suggests that PON1 enhances HDL3-mediated choles-terol 
efflux and that the ABCA1 transporter pathway mediates this 
effect. 
PON1 is a novel independent transporter involved in cholesterol efflux 
We performed experiments in more than one cell type to deter-mine 
which of the three cholesterol efflux pathways might be stimu-lated 
by PON1. PON1-mediated cholesterol efflux was first studied in 
THP-1 macrophage-like cells, which equally express ABCA1/ABCG1 
transporters and the SR-BI receptor. Our results show that incubation 
of cholesterol-loaded THP-1 with 0, 5, 10, or 20 U/ml PON1 alone re-sults 
in a significant increment in cholesterol efflux by 2.5-, 3-, and 
4.6-fold compared to the control (Fig. 2A). 
To better understand the mechanism by which PON1 mediates 
cholesterol efflux, we investigated the role of the ABCA1 pathway in 
this process. For that purpose we used cAMP-pretreated J774 macro-phages, 
which express high levels of ABCA1 [4]. ABCA1-enriched and 
nonenriched J774 macrophages (control) were incubated with in-creasing 
concentrations of PON1. The results show that PON1 signifi-cantly 
promoted cholesterol efflux in ABCA1-enriched macrophages 
in a concentration-dependent manner, whereas no significant in-crease 
was observed in the control (Fig. 2B). 
Taking into account that PON1 and apo-AI are both associated 
with HDL, we investigated a possible synergistic effect between 
apo-AI and PON1 to mediate cholesterol efflux. For that purpose, we 
incubated [3H]cholesterol-loaded ABCA1-enriched J774 cells with in-creasing 
concentrations of PON1 (0, 5, 10, and 20 U/ml) in the pres-ence 
or not of apo-AI (25 μg/ml) for 4 h. Results in Fig. 2C, shows 
that apo-AI-mediated cholesterol efflux was significantly potentiated, 
and in a dose-dependent manner, in the presence of PON1 by an av-erage 
of 37.24±5.35% (pb0.01). 
The incubation of cholesterol-loaded ABCA1-enriched J774 with 
increasing concentrations of BSA (negative control) had no effect, 
whereas 25 μg/ml apo-AI (positive control) significantly enhanced 
cholesterol efflux (data not shown). This confirms that PON1 en-hances 
ABCA1-dependent cholesterol efflux from J774 macrophages. 
To assess SR-BI-mediated cholesterol efflux, we used Fu5AH rat 
hepatoma cells, which express high levels of SR-BI and lack a func-tional 
ABCA1 [41]. Interestingly, incubating PON1 (0, 5, 10, or 20 U/ 
ml) with SR-BI-overexpressing Fu5AH cells did not cause a significant 
change in cholesterol efflux (Fig. 2D). This effect was nonsignificant 
even when PON1 was incubated in the presence of HDL (50 μg/ml), 
which leads to the suggestion that the SR-BI receptor was not or 
was less implicated in the PON1-mediated cholesterol efflux. 
ABCA1 overexpression increases PON1 internalization 
cAMP-treated J774 macrophages exhibited more intense immuno-fluorescence 
than the control cells (Figs. 3A and B), indicating that 
ABCA1 overexpression increases the internalization of PON1, which 
leads to its localization in the cell's cytoplasm (Fig. 3C, 100× original 
magnification). This is in accordance with the results reported by 
Efrat and Aviram [42], who used FITC fluorescence and confocal mi-croscopy 
to demonstrate an internalization of PON1 and its localiza-tion 
in the cell's cytoplasm compartment. 
ABCA1-enriched and nonenriched J774 macrophages were incu-bated 
with PON1 for 3 h. PON1 was detected in the ABCA1 
A 
40 
30 
20 
10 
0 
* * 
** 
+ + + + HDL3 [50μg/ml] 
0 5 10 20 PON1 [U/ml] 
Cholesterol efflux (%) 
30 
20 
10 
0 
ns 
* 
** 
** 
+ + + + + + + + HDL3 [50 μg/ml] 
- + - + - + - + cAMP [0.3 mM] 
0 5 10 20 PON1 [U/ml] 
B 
Cholesterol efflux (%) 
Fig. 1. HDL3-associated PON1 mediates cholesterol efflux from THP-1 and J774 macro-phages. 
(A) THP-1 macrophages were loaded with [3H]cholesterol (2 μCi/ml) for 24 h. 
The cells were then washed and further incubated for 24 h with 50 μg/ml HDL3 alone 
(control) or in the presence of increasing concentrations of PON1 (0–20 U/ml; 
0–50 μg/ml PON1 protein concentration). (B) [3H]Cholesterol-loaded J774 macro-phages 
were incubated overnight at 37 °C without (control) or with 0.3 mM cAMP 
(ABCA1-enriched cells). After being washed, the cells were incubated for 24 h with 
HDL3 alone or with HDL3 supplemented with increasing concentrations of PON1 
(0–20 U/ml; 0–50 μg/ml). Results are expressed as the means ± SEM of three indepen-dent 
experiments. *pb0.05, **pb0.01. 
H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1375
1376 H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 
0 5 10 20 PON1 [U/ml] 5 10 20 5 10 20 
0 5 10 15 20 25 0 5 10 20 20 
+ 
20 
15 
10 
5 
0 
** 
THP1 
Cholesterol efflux (%) 
15 
10 
5 
PON1 
cAMP - 
+ 
+ 
0 
0 0 0 0 0 
- - - + + + 
+ 
+ 
Magnification (x 100) 
** 
*** 
10 
8 
6 
4 
2 
0 
Cholesterol efflux (%) 
apo-AI 25μg/ml 
15 
10 
5 
A B C 
Fig. 3. ABCA1 overexpression increases PON1 internalization. (A) Control and (B and C) ABCA1-enriched J774 cells (original magnification 40× and 100×, respectively) were incu-bated 
with PON1 (20 U/ml; 50 μg/ml) before their fixation with 4% paraformaldehyde and their immunostaining with primary antibody for PON1 (mouse). The cells were then 
incubated with Alexa 488-labeled goat anti-mouse IgG secondary antibody and processed for immunofluorescence microscopy. 
PON1 [U/ml] 
** 
*** 
*** 
J774 
HDL 
cAMP [0.3 mM] 
0 
apo-AI-free 
PON1 [U/ml] 
Cholesterol efflux (%) 
0 
ns Fu5AH 
ns 
PON1 [U/ml] 
HDL [μg/ml] 
0 
50 50 
0 0 0 0 
Cholesterol efflux (%) 
ns 
A B 
C D 
Fig. 2. Effects of purified PON1 on the cholesterol efflux. (A) [3H]Cholesterol-loaded THP-1 cells were incubated with PON1 (0–20 U/ml; 0–50 μg/ml) for 24 h. (B) J774 macrophages 
were loaded with [3H]cholesterol and then incubated for 12 h in the presence of cAMP to yield ABCA1 expression. Cholesterol efflux from ABCA1-enriched and nonenriched J774 
was then initiated by the addition of PON1 (0–20 U/ml; 0–50 μg/ml) for 4 h. (C) [3H]Cholesterol-loaded and ABCA1-enriched J774 cells were incubated for 4 h with increasing con-centrations 
of PON1 (0–50 μg/ml) in presence or not of apo-AI. (D) [3H]Cholesterol-loaded Fu5AH cells were incubated with PON1 (0–20 U/ml; 0–50 μg/ml) and/or HDL3 (50 μg/ml, 
positive control) for 24 h. To investigate if PON1 require HDL for its interaction with SR-BI receptors, [3H]cholesterol-loaded Fu5AH cells were incubated with PON1 (20 U/ml; 50 μg/ 
ml) in the presence of HDL3 (50 μg/ml) for 24 h. Results are expressed as the means ± SEM of more than three independent experiments. **pb0.01, ***pb0.001; ns, nonsignificant.
PON1 (20 U/ml) 
1 2 
+ + 
immunoprecipitate, with the highest amount in the precipitate from 
ABCA1-enriched J774 (Fig. 4), suggesting that PON1 binds with 
ABCA1 to form a protein complex. 
We previously showed that the oxidation of purified human PON1 
causes a decrease in its antioxidant and paraoxonase activities [19]. 
This effect was attributed to the loss of the free sulfhydryl group 
[19]. In this study, we show that exposure of PON1 to oxygen free rad-icals 
produced by γ-radiolysis affects significantly its ability to pro-mote 
HDL3-mediated cholesterol efflux from THP-1 macrophage-like 
cells (−29.04%, pb0.01, Fig. 5A). In addition, heat (70 °C for 
1 h) and NEM treatment, which irreversibly blocks SH groups by 
covalent binding [43], significantly decreased (pb0.01) paraoxonase 
activity as well as its ability to promote cholesterol efflux, especially 
from ABCA1-enriched macrophages (Fig. 5B). 
PON1 interacts with lipid raft domains to initiate cholesterol efflux from 
macrophages 
In the second part of our study, we investigated the capacity of 
PON1 to interact with lipid rafts and to mediate cholesterol efflux. Su-crose 
gradients separated the plasma membranes into two fractions: 
40 
30 
20 
10 
0 
* 
A 
Cholesterol efflux (%) 
10 
8 
6 
4 
2 
0 
HDL3-nPON1 [20U/ml] HDL3-oxPON1 [20U/ml] 
** ** 
nPON1 Heat-inactivated 
PON1 
NEM-inactivated 
PON1 
B 
Cholesterol efflux (%) 
Fig. 5. PON1 oxidation impairs its capacity to stimulate HDL-mediated cholesterol 
efflux. (A) [3H]Cholesterol-loaded THP-1 macrophages were incubated with HDL3 in 
the presence of native (nPON1) or oxidized PON1 (oxPON1) for 24 h. Oxidation of 
PON1 was induced by its exposure to oxygen free radicals produced by γ-radiolysis 
of water. (B) Denaturing PON1 reduces its capacity to interact with ABCA1 receptors 
and to promote cholesterol efflux. 20 U/ml (50 μg/ml) PON1 was inactivated by heat 
(70 °C/1 h) or by NEM treatment (1 h) before starting the experiments. J774 macro-phages 
were loaded with [3H]cholesterol and treated with cAMP to upregulate 
ABCA1 expression. Treated and control cells were incubated with native PON1, heat-inactivated 
PON1, or NEM-treated PON1 for 4 h. Data are given as means ± SEM of 
more than three independent experiments. *pb0.05, **pb0.01. 
8×10 04 
6×10 04 
4×10 04 
2×10 04 
2.0×1004 
1.5×1004 
1.0×1004 
5.0×1003 
Control 
* 
apoA1 
PON1 
Control 
apoA1 
PON1 
0.0 
Raft fractions 
** 
* 
Cholesterol contents 
(cpm) 
12 ** 
9 
6 
3 
Control-CD free 
apoA1-CD 
PON1-CD free 
Control-CD 
apoA1-CD free 
PON1-CD 
0 
* 
Cholesterol efflux (%) 
A 
B 
Non-Raft fractions 
Fig. 6. PON1 and apo-AI start mediating cholesterol efflux from lipid raft domains. THP- 
1 macrophages were loaded with [3H]cholesterol and then incubated for 4 h with apo- 
AI or PON1. Plasma membranes were isolated on a sucrose gradient as described under 
Material and methods. (A) Cholesterol contents (cpm) in associated lipid raft and non-raft 
fractions. (B) [3H]Cholesterol-loaded and ABCA1-enriched J774 macrophages were 
pretreated for 1 h at 37 °C with medium containing BSA alone or plus methyl-β- 
cyclodextrin (CD; 2 mg/ml). After the above preincubations, the media were removed 
and efflux to fresh medium containing or not apo-AI or PON1 over 4 h was measured. 
Results are expressed as the means ± SEM of more than three independent experi-ments. 
*pb0.05, **pb0.01 vs control. 
PON1 45Kda 
cAMP 
+ 
Fig. 4. Coimmunoprecipitation of ABCA1 and PON1 in lysates from J774 macrophages 
shows that PON1 binds to ABCA1. PON1 (20 U/ml) was added to ABCA1-enriched 
(lane 1) or nonenriched (lane 2) J774 for 3 h and then ABCA1 immunoprecipitation 
(IP) from cell lysates was performed using Dynabeads protein-G and ABCA1 monoclo-nal 
antibody. ABCA1 IP was followed by PON1 Western blot (10% SDS–PAGE) using 
mouse anti-human PON1 monoclonal antibody. 
H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1377
1378 H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 
light membranes (rafts) and heavy membranes (nonrafts). Western 
blotting using fotilin-1, which migrated with the rafts, confirmed 
the separation (data not shown). 
Incubating [3H]cholesterol-loaded THP-1 macrophage-like cells 
with PON1 and apo-AI resulted in a significant decrease in the choles-terol 
content of the lipid raft fractions, by approximately 38.0 
(pb0.01) and 20.5% (pb0.05), respectively. However, in the nonraft 
fractions, cholesterol decreased by only 23.1 and 12.3% (pb0.05) in 
the presence of PON1 and apo-AI, respectively (Fig. 6A). This sug-gested 
that PON1 interacts with lipid rafts to initiate cholesterol ef-flux 
from macrophages. 
To gain more insight into the mechanismby which the PON1–lipid 
raft interaction modulates cholesterol efflux, we studied the effect of 
a short MβCD pretreatment on the increase in cholesterol efflux from 
cholesterol-loaded macrophages by PON1 and apo-AI (control). The 
MβCD treatment caused a selective removal of cholesterol from 
lipid raft domains and induced the dispersal of their lipid and protein 
components [28]. Interestingly, the exposure of ABCA1-enriched J774 
macrophages to MβCD for 1 h resulted in a significant impairment of 
the cholesterol efflux mediated by PON1 (57.21%, pb0.05) and by 
apo-AI (63.89%, pb0.01) (Fig. 6B). This suggested that the presence 
of intact lipid rafts seems to be essential for initiating PON1 to stimu-late 
the cholesterol efflux process. 
Modeling the kinetics of cholesterol export from macrophages to 
apo-AI predicted a rapid initial efflux from the small pool, followed 
by a slower but substantial and sustained efflux from the separate 
larger pool, which contributed to most of the exported cholesterol 
[44].We examined the kinetics model of cholesterol efflux from mac-rophages 
to PON1 compared to apo-AI. Fig. 7 shows a typical choles-terol 
efflux experiment (0 to 16 h) from cholesterol-loaded J774 
macrophages treated with cAMP. Cholesterol efflux to DMEM alone 
containing 1% BSA was minimal compared to apo-AI- and PON1- 
mediated cholesterol efflux. 
Like apo-AI, PON1-induced cholesterol efflux can be qualitatively 
split into two phases, an initial high cholesterol efflux rate phase 
that lasts from 0 to 3 h followed by a slower cholesterol efflux rate 
phase. 
PON1 has an effect on ABCA1 protein and gene expression 
To investigate the effects of PON1 on the ABCA1 level during the 
distinct phases of the cholesterol efflux process, ABCA1 protein and 
gene expression was analyzed by Western blotting and qPCR, 
10000 
8000 
6000 
4000 
2000 
15 
10 
5 
1 3 16 1 3 16 
0 1 3 16 1 3 16 
*** *** 
A 
respectively, in J774 macrophages incubated in the presence or 
absence of PON1. Interestingly, a short incubation of the macrophages 
with PON1 increased ABCA1 protein levels after 1 and 3 h, but the 
effect disappeared after a longer incubation period (16 h; Fig. 8A). 
More importantly, the upregulation by PON1 was accompanied by 
an increase in ABCA1 mRNA levels (Fig. 8B). The same effect on 
ABCA1 protein expression was observed when apo-AI was used as a 
control. These results demonstrate that PON1 modulates ABCA1 
expression on macrophages. 
Discussion 
Cholesterol efflux is thought to be one of the most important 
atheroprotective properties of HDLs. This process is mediated by the 
interaction of lipid-free apo-AI and HDLs with ABCA1, ABCG1, and 
SR-B1 membrane proteins as well as via other mechanisms, including 
passive diffusion. Apo-AI is a key factor in the enhancement of HDL-mediated 
cholesterol efflux. However, plasma HDL particles contain 
0 4 8 12 16 
25 
20 
15 
10 
5 
0 
Apo-AI [25 μg/ml] 
PON1 [20 U/ml: 50 μg/ml] 
Control [BSA: 50 μg/ml] 
Time [hours] 
Cholesterol efflux (%) 
Fig. 7. PON1 enhances cholesterol efflux from the ABCA1-dependent pathway in rapid-low 
and slow-high pools. Kinetics of cholesterol efflux to apo-AI (25 μg/ml) and PON1 
(20 U/ml; 50 μg/ml) over the first 16 h in J774 [3H]cholesterol-loaded macrophages 
enriched in ABCA1 is shown. BSA (50 μg/ml) was used as control. Results are expressed 
as the means ± SEM of more than three independent experiments. 
0 
** 
*** 
PON1 
expression 
gene ABCA1 Relative 0 1 3 16 
PON1 Incubation time [hours] 0 
*** *** 
*** *** 
Incubation time [Hours] 
PON1 
apo-AI 
Hours 
ABCA1 
0 
apo-AI PON1 
Densitometry Analysis 
B 
Fig. 8. Effects of short and long pretreatment of J774 macrophages with apo-AI or PON1 
on ABCA1 protein and RNA expression. Kinetics analysis of ABCA1 expression (0, 3, and 
16 h) upon incubation with apo-AI (25 μg/ml) or PON1 (20 U/ml; 50 μg/ml) was car-ried 
out in non-ABCA1-enriched J774 cells. Protein levels and RNA expression of 
ABCA1 were respectively determined by (A) Western blot and (B) RT-PCR analyses. 
Quantitative analysis was determined using densitometry from three independent 
experiments. Results are expressed as means ± SEM. **pb0.01, ***pb0.001.
H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1379 
a variety of proteins with antiatherogenic potential, including PON1, 
that may also contribute significantly to the cholesterol efflux pro-cess. 
PON1 is a protein that is mainly complexed to HDLs and has 
been associated, in part, with the antioxidant and anti-inflammatory 
properties of HDLs. The goal of this study was to investigate the in-volvement 
of PON1 in the regulation of the cholesterol efflux process, 
especially the mechanism by which PON1 modulates HDL-mediated 
cholesterol transport. 
Our results showed that PON1 enhances HDL-mediated cholester-ol 
efflux in THP-1 macrophage-like cells and in J774 macrophages. 
The effect was more pronounced with ABCA1-enriched J774 macro-phages, 
suggesting that an interaction between PON1 and ABCA1 re-ceptors 
promotes cholesterol efflux. Our results showed that purified 
PON1 is highly effective in stimulating cholesterol efflux via the 
ABCA1 pathway, whereas no effect was observed with respect to 
SR-BI receptors. This finding is in agreement with that of Rosenblat 
et al., who used purified human PON1 and HDLs from PON1 knockout 
and PON1Tg mice [26]. Moreover, Fuhrman et al. recently reported 
that PON1 may protect macrophages from apoptosis by upregulating 
macrophage SR-BI-mediated HDL binding to the cells. However, the 
involvement of SR-BI in the interaction of HDLs with macrophages 
does not affect cellular cholesterol efflux [45]. 
Although apo-AI is the principal HDL-associated protein that in-teracts 
with membrane proteins to promote cholesterol efflux, other 
HDL-associated proteins may play this role. A recent study by De 
Beer et al. [46] showed that lipid-free serum amyloid A (SAA) acts 
like apo-AI by mediating sequential cholesterol efflux via ABCA1 
and ABCG1 [46]. Remaley et al. showed that ABCA1-mediated lipid ef-flux 
is not specific to apo-AI but can also occur with other apolipopro-teins 
that contain multiple amphipathic helical domains [47]. A study 
by Xie et al. [49] showed that an apo-AI mimetic peptide (D-4F), 
which has no sequence homology with apo-AI but possesses the 
class-A amphipathic helical motif, promotes cholesterol efflux 
through the ABCA1 pathway [48,49]. It is thus not surprising that 
PON1 modulates cholesterol efflux from macrophages principally 
via the ABCA1 pathway. Indeed, like apo-AI and SAA, the secondary 
structure of PON1 contains amphipathic α-helices with approximate-ly 
22 amino acids [50] that enable HDL particles to tightly bind, stabi-lize, 
and stimulate PON1 [51]. 
Our results demonstrate that PON1 enhances cholesterol efflux 
and induces an upregulation of ABCA1. Although we did not investi-gate 
the mechanism of PON1-induced upregulation of ABCA1, it has 
been suggested that this effect could be attributed to the capacity of 
PON1 to hydrolyze oxidized phospholipids to form lyso-PC, which 
in turn induces ABCA1 expression [26]. In accordance, Hou et al. dem-onstrated 
that lyso-PC increases the mRNA and protein expression of 
PPARγ, LXRα, and ABCA1 of macrophage foamcells in a concentration-dependent 
manner [52]. The upregulation of ABCA1, in this study, 
was confirmed by the measurement of ABCA1 protein and mRNA 
expression and also evidenced by the internalization of PON1 in mac-rophages. 
Our results from the immunofluorescence and cholesterol 
efflux assays showed that the internalization of PON1 in macrophages 
is ABCA1 dependent. A reduction in ABCA1 content or an alteration in 
the structure of PON1 significantly affected PON1-mediated cholesterol 
efflux. 
Sorenson et al. showed that purified human and rabbit PON1 both 
have a single free sulfhydryl group (Cys284) and suggested that this 
cysteine is part of the active site of PON1 required for its antioxidant 
activity [53]. Previous studies, including ours, showed that PON1 mu-tants 
display significantly less antioxidant activity and that blocking 
the free sulfhydryl groups with NEM or p-hydroxymercuribenzoate 
significantly reduces the antioxidant activity of PON1 [19,54]. In this 
study, we showed that exposure of PON1 to free radicals or its inacti-vation 
by heat or NEM treatment induced a net reduction in its ability 
to promote cholesterol efflux from THP-1 macrophage-like cells and 
ABCA1-enriched J774 macrophages. These results confirm that the 
capacity of PON1 to stimulate cholesterol efflux is dependent on its 
structural integrity. 
The mechanism by which apo-AI removes excess cholesterol and 
phospholipids from macrophages has been extensively investigated. 
Gaus et al. [28,44] studied the interaction between apo-AI and lipid 
raft microdomains and concluded that cholesterol efflux to apo-AI oc-curs 
sequentially from two membrane pools with different kinetics, 
that is, rapid release from a small rapid efflux pool within the first 
few hours followed by progressive release from a major slow efflux 
pool over several hours. They proposed that cholesterol exported to 
apo-AI from this major slow efflux pool comes from nonraft regions 
of the plasma membrane. However, the interaction of apo-AI with 
lipid rafts is required to stimulate this efflux. Our results showed 
that PON1-mediated cholesterol efflux also occurs via two steps: 
rapid cholesterol efflux during the first 3 h followed by slow choles-terol 
efflux thereafter. The level of cholesterol efflux mediated by 
PON1 was comparable to that mediated by apo-AI, at least during 
the rapid cholesterol efflux step. This suggested that PON1 acts via 
an apo-AI-like mechanism to mediate cholesterol efflux. This mecha-nistic 
assumption was supported by the results of ABCA1 expression 
measurements and experiments involving a pretreatment with cyclo-dextrin, 
which selectively depletes cholesterol from lipid raft do-mains 
(~50%) but not from nonraft domains [28]. First, the ABCA1 
transporter was overexpressed within the first 3 h of incubation of 
PON1 with cholesterol-loaded J774 macrophages, which covers the 
period during which cholesterol is released from the small rapid ef-flux 
pool. After 16 h, ABCA1 expression was downregulated, which 
was probably related to the involvement of the ABCG1 transporter 
or SR-BI receptor. These two proteins do not interact with lipid-free 
apo-AI [55,56]. Second, cyclodextrin significantly affected the ability 
of PON1 and apo-AI to mediate cholesterol efflux from macrophages 
(Fig. 6B). 
Studies investigating the relationship between membrane plasma 
microdomains and apo-AI with respect to the cholesterol efflux ca-pacity 
of apo-AI have given rise to conflicting results. The distribution 
of ABCA1 between lipid raft and nonraft domains depends on the 
macrophage cell line and the method used to prepare the rafts. 
ABCA1 is not present in Triton-resistant membrane domains, whereas 
a significant proportion can be detected in Lubrol-resistant domains 
[57,58]. Mendez et al. reported that membrane raft lipids are not di-rectly 
available for lipid efflux by apolipoprotein-dependent mecha-nisms 
[58]. In contrast, cholesterol efflux from cells to HDL particles 
or to plasma can be partially explained by the depletion of cholesterol 
from membrane rafts. This suggests that apo-AI– and PON1–ABCA1- 
mediated cellular cholesterol efflux occurs by mechanisms distinct 
from those mediated by lipoprotein particles containing lipids as 
their major constituent. More interestingly, this study showed that 
PON1 plays an important role in the antiatherogenic properties of 
HDLs and can exert its protective function outside the lipoprotein en-vironment. 
This is supported by a recent study by Deakin et al. show-ing 
that PON1 is not a fixed component of HDLs because it can be 
transferred from HDLs to the external face of the plasma membrane 
of cells in an enzymatically active form [59]. 
These findings open a new window on the association between 
PON1 and HDLs with respect to their antiatherogenic function as 
well as on the mechanisms by which PON1 may exert its atheropro-tective 
effect, especially by regulating cholesterol efflux from macro-phages 
and maintaining cholesterol homeostasis. However, further 
studies are needed to elucidate the nature of the pathways involved 
in the promotion of cholesterol efflux by PON1 and to determine 
the activity of free, non-HDL-associated PON1. 
Acknowledgment 
This work was supported by a grant from the Canadian Institutes 
of Health Research (MOP-89912).
1380 H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 
References 
[1] Cuchel, M.; Rader, D. J. Macrophage reverse cholesterol transport: key to the 
regression of atherosclerosis? Circulation 113:2548–2555; 2006. 
[2] Ruano, J.; Lopez-Miranda, J.; Fuentes, F.; Moreno, J. A.; Bellido, C.; Perez-Martinez, 
P.; Lozano, A.; Gomez, P.; Jimenez, Y.; Perez Jimenez, F. Phenolic content of virgin 
olive oil improves ischemic reactive hyperemia in hypercholesterolemic patients. 
J. Am. Coll. Cardiol. 46:1864–1868; 2005. 
[3] Tall, A. R.; Costet, P.; Wang, N. Regulation and mechanisms of macrophage choles-terol 
efflux. J. Clin. Invest. 110:899–904; 2002. 
[4] Yancey, P. G.; Bortnick, A. E.; Kellner-Weibel, G.; de la Llera-Moya, M.; Phillips, 
M. C.; Rothblat, G. H. Importance of different pathways of cellular cholesterol ef-flux. 
Arterioscler. Thromb. Vasc. Biol. 23:712–719; 2003. 
[5] Smith, J. D. Insight into ABCG1-mediated cholesterol efflux. Arterioscler. Thromb. 
Vasc. Biol. 26:1198–1200; 2006. 
[6] Favari, E.; Lee, M.; Calabresi, L.; Franceschini, G.; Zimetti, F.; Bernini, F.; Kovanen, 
P. T. Depletion of pre-β-high density lipoprotein by human chymase impairs ATP-binding 
cassette transporter A1- but not scavenger receptor class B type I-mediated 
lipid efflux to high density lipoprotein. J. Biol. Chem. 279:9930–9936; 
2004. 
[7] Baldan, A.; Bojanic, D. D.; Edwards, P. A. The ABCs of sterol transport. J. Lipid Res. 
50:S80–S85 (Suppl.); 2009. 
[8] Gu, F.; Jones, M. K.; Chen, J.; Patterson, J. C.; Catte, A.; Jerome, W. G.; Li, L.; Segrest, 
J. P. Structures of discoidal high density lipoproteins: a combined computational– 
experimental approach. J. Biol. Chem. 285:4652–4665; 2010. 
[9] Vaisar, T.; Pennathur, S.; Green, P. S.; Gharib, S. A.; Hoofnagle, A. N.; Cheung, 
M. C.; Byun, J.; Vuletic, S.; Kassim, S.; Singh, P.; Chea, H.; Knopp, R. H.; 
Brunzell, J.; Geary, R.; Chait, A.; Zhao, X. Q.; Elkon, K.; Marcovina, S.; Ridker, P.; 
Oram, J. F.; Heinecke, J. W. Shotgun proteomics implicates protease inhibition 
and complement activation in the antiinflammatory properties of HDL. J. Clin. 
Invest. 117:746–756; 2007. 
[10] Visioli, F.; Poli, A.; Gall, C. Antioxidant and other biological activities of phenols 
from olives and olive oil. Med. Res. Rev. 22:65–75; 2002. 
[11] Costa, L. G.; Vitalone, A.; Cole, T. B.; Furlong, C. E. Modulation of paraoxonase 
(PON1) activity. Biochem. Pharmacol. 69:541–550; 2005. 
[12] James, R. W.; Brulhart-Meynet, M. C.; Singh, A. K.; Riederer, B.; Seidler, U.; Out, R.; 
Van Berkel, T. J.; Deakin, S. The scavenger receptor class B, type I is a primary de-terminant 
of paraoxonase-1 association with high-density lipoproteins. Arterios-cler. 
Thromb. Vasc. Biol. 30:2121–2127; 2010. 
[13] Deakin, S.; Moren, X.; James, R. W. Very low density lipoproteins provide a vector 
for secretion of paraoxonase-1 from cells. Atherosclerosis 179:17–25; 2005. 
[14] Jaouad, L.; Milochevitch, C.; Khalil, A. PON1 paraoxonase activity is reduced dur-ing 
HDL oxidation and is an indicator of HDL antioxidant capacity. Free Radic. Res. 
37:77–83; 2003. 
[15] Marsillach, J.; Aragones, G.; Mackness, B.; Mackness, M.; Rull, A.; Beltran-Debon, 
R.; Pedro-Botet, J.; Alonso-Villaverde, C.; Joven, J.; Camps, J. Decreased 
paraoxonase-1 activity is associated with alterations of high-density lipoprotein 
particles in chronic liver impairment. Lipids Health Dis. 9:46; 2010. 
[16] Fuhrman, B.; Volkova, N.; Aviram, M. Oxidative stress increases the expression of 
the CD36 scavenger receptor and the cellular uptake of oxidized low-density lipo-protein 
in macrophages from atherosclerotic mice: protective role of antioxidants 
and of paraoxonase. Atherosclerosis 161:307–316; 2002. 
[17] Rozenberg, O.; Shih, D. M.; Aviram, M. Paraoxonase 1 (PON1) attenuates macro-phage 
oxidative status: studies in PON1 transfected cells and in PON1 transgenic 
mice. Atherosclerosis 181:9–18; 2005. 
[18] Watson, A. D.; Berliner, J. A.; Hama, S. Y.; La Du, B. N.; Faull, K. F.; Fogelman, A. M.; 
Navab, M. Protective effect of high density lipoprotein associated paraoxonase: 
inhibition of the biological activity of minimally oxidized low density lipoprotein. 
J. Clin. Invest. 96:2882–2891; 1995. 
[19] Jaouad, L.; de Guise, C.; Berrougui, H.; Cloutier, M.; Isabelle, M.; Fulop, T.; Payette, 
H.; Khalil, A. Age-related decrease in high-density lipoproteins antioxidant activ-ity 
is due to an alteration in PON1's free sulfhydryl groups. Atherosclerosis 185: 
191–200; 2006. 
[20] Tward, A.; Xia, Y. R.; Wang, X. P.; Shi, Y. S.; Park, C.; Castellani, L. W.; Lusis, A. J.; 
Shih, D. M. Decreased atherosclerotic lesion formation in human serum paraoxo-nase 
transgenic mice. Circulation 106:484–490; 2002. 
[21] Shih, D. M.; Gu, L.; Xia, Y. R.; Navab, M.; Li, W. F.; Hama, S.; Castellani, L. W.; 
Furlong, C. E.; Costa, L. G.; Fogelman, A. M.; Lusis, A. J. Mice lacking serum paraox-onase 
are susceptible to organophosphate toxicity and atherosclerosis. Nature 
394:284–287; 1998. 
[22] Mackness, B.; Hunt, R.; Durrington, P. N.; Mackness, M. I. Increased immunoloca-lization 
of paraoxonase, clusterin, and apolipoprotein A-I in the human artery 
wall with the progression of atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 17: 
1233–1238; 1997. 
[23] Tavori, H.; Khatib, S.; Aviram, M.; Vaya, J. Characterization of the PON1 active site 
using modeling simulation, in relation to PON1 lactonase activity. Bioorg. Med. 
Chem. 16:7504–7509; 2008. 
[24] Navab, M.; Hama, S. Y.; Hough, G. P.; Hedrick, C. C.; Sorenson, R.; La Du, B. N.; 
Kobashigawa, J. A.; Fonarow, G. C.; Berliner, J. A.; Laks, H.; Fogelman, A. M. High 
density associated enzymes: their role in vascular biology. Curr. Opin. Lipidol. 9: 
449–456; 1998. 
[25] Rosenblat, M.; Gaidukov, L.; Khersonsky, O.; Vaya, J.; Oren, R.; Tawfik, D. S.; 
Aviram, M. The catalytic histidine dyad of high density lipoprotein-associated 
serum paraoxonase-1 (PON1) is essential for PON1-mediated inhibition of low 
density lipoprotein oxidation and stimulation of macrophage cholesterol efflux. 
J. Biol. Chem. 281:7657–7665; 2006. 
[26] Rosenblat, M.; Vaya, J.; Shih, D.; Aviram, M. Paraoxonase 1 (PON1) enhances HDL-mediated 
macrophage cholesterol efflux via the ABCA1 transporter in association 
with increased HDL binding to the cells: a possible role for lysophosphatidylcho-line. 
Atherosclerosis 179:69–77; 2005. 
[27] Pike, L. J.; Han, X.; Chung, K. N.; Gross, R. W. Lipid rafts are enriched in arachidonic 
acid and plasmenylethanolamine and their composition is independent of 
caveolin-1 expression: a quantitative electrospray ionization/mass spectrometric 
analysis. Biochemistry 41:2075–2088; 2002. 
[28] Gaus, K.; Kritharides, L.; Schmitz, G.; Boettcher, A.; Drobnik, W.; Langmann, T.; 
Quinn, C. M.; Death, A.; Dean, R. T.; Jessup, W. Apolipoprotein A-1 interaction 
with plasma membrane lipid rafts controls cholesterol export from macrophages. 
FASEB J. 18:574–576; 2004. 
[29] Gillotte, K. L.; Davidson, W. S.; Lund-Katz, S.; Rothblat, G. H.; Phillips, M. C. Re-moval 
of cellular cholesterol by pre-β-HDL involves plasma membrane microso-lubilization. 
J. Lipid Res. 39:1918–1928; 1998. 
[30] Gargalovic, P.; Dory, L. Caveolins and macrophage lipid metabolism. J. Lipid Res. 
44:11–21; 2003. 
[31] Rees, D.; Sloane, T.; Jessup, W.; Dean, R. T.; Kritharides, L. Apolipoprotein A-I stim-ulates 
secretion of apolipoprotein E by foam cell macrophages. J. Biol. Chem. 274: 
27925–27933; 1999. 
[32] Sattler, W.; Mohr, D.; Stocker, R. Rapid isolation of lipoproteins and assessment of 
their peroxidation by high-performance liquid chromatography postcolumn 
chemiluminescence. Methods Enzymol. 233:469–489; 1994. 
[33] Gan, K. N.; Smolen, A.; Eckerson, H. W.; La Du, B. N. Purification of human serum 
paraoxonase/arylesterase: evidence for one esterase catalyzing both activities. 
Drug Metab. Dispos. 19:100–106; 1991. 
[34] Eckerson,H.W.;Wyte, C.M.; La Du, B.N. The human serumparaoxonase/arylesterase 
polymorphism. Am. J. Hum. Genet. 35:1126–1138; 1983. 
[35] Hajj Hassan, H.; Blain, S.; Boucher, B.; Denis, M.; Krimbou, L.; Genest, J. Structural 
modification of plasma HDL by phospholipids promotes efficient ABCA1- 
mediated cholesterol release. J. Lipid Res. 46:1457–1465; 2005. 
[36] Fortin, C. F.; Larbi, A.; Lesur, O.; Douziech, N.; Fulop Jr., T. Impairment of SHP-1 
down-regulation in the lipid rafts of human neutrophils under GM-CSF stimula-tion 
contributes to their age-related, altered functions. J. Leukocyte Biol. 79: 
1061–1072; 2006. 
[37] Abe-Dohmae, S.; Kato, K. H.; Kumon, Y.; Hu, W.; Ishigami, H.; Iwamoto, N.; 
Okazaki, M.; Wu, C. A.; Tsujita, M.; Ueda, K.; Yokoyama, S. Serum amyloid A gen-erates 
high density lipoprotein with cellular lipid in an ABCA1- or ABCA7- 
dependent manner. J. Lipid Res. 47:1542–1550; 2006. 
[38] Hu, W.; Abe-Dohmae, S.; Tsujita, M.; Iwamoto, N.; Ogikubo, O.; Otsuka, T.; 
Kumon, Y.; Yokoyama, S. Biogenesis of HDL by SAA is dependent on ABCA1 in 
the liver in vivo. J. Lipid Res. 49:386–393; 2008. 
[39] Parra, S.; Alonso-Villaverde, C.; Coll, B.; Ferre, N.; Marsillach, J.; Aragones, G.; 
Mackness, M.; Mackness, B.; Masana, L.; Joven, J.; Camps, J. Serum paraoxonase- 
1 activity and concentration are influenced by human immunodeficiency virus 
infection. Atherosclerosis 194:175–181; 2007. 
[40] Seres, I.; Paragh, G.; Deschene, E.; Fulop Jr., T.; Khalil, A. Study of factors influenc-ing 
the decreased HDL associated PON1 activity with aging. Exp. Gerontol. 39: 
59–66; 2004. 
[41] Attia, N.; Fournier, N.; Vedie, B.; Cambillau, M.; Beaune, P.; Ziegler, O.; Grynberg, 
A.; Paul, J. L.; Guerci, B. Impact of android overweight or obesity and insulin resis-tance 
on basal and postprandial SR-BI and ABCA1-mediated serum cholesterol 
efflux capacities. Atherosclerosis 209:422–429; 2010. 
[42] Efrat, M.; Aviram, M. Macrophage paraoxonase 1 (PON1) binding sites. Biochem. 
Biophys. Res. Commun. 376:105–110; 2008. 
[43] Blumrich, M.; Petzinger, E. Two distinct types of SH-groups are necessary for 
bumetanide and bile acid uptake into isolated rat hepatocytes. Biochim. Biophys. 
Acta 1149:278–284; 1993. 
[44] Gaus, K.; Gooding, J. J.; Dean, R. T.; Kritharides, L.; Jessup, W. A kinetic model to 
evaluate cholesterol efflux from THP-1 macrophages to apolipoprotein A-1. 
Biochemistry 40:9363–9373; 2001. 
[45] Fuhrman, B.; Gantman, A.; Aviram, M. Paraoxonase 1 (PON1) deficiency in mice is 
associated with reduced expression of macrophage SR-BI and consequently the 
loss of HDL cytoprotection against apoptosis. Atherosclerosis 211:61–68; 2010. 
[46] de Beer, M. C.; Ji, A.; Jahangiri, A.; Vaughan, A. M.; de Beer, F. C.; van der 
Westhuyzen, D. R.; Webb, N. R. ATP binding cassette G1-dependent cholesterol 
efflux during inflammation. J. Lipid Res. 52:345–353; 2011. 
[47] Remaley, A. T.; Stonik, J. A.; Demosky, S. J.; Neufeld, E. B.; Bocharov, A. V.; 
Vishnyakova, T. G.; Eggerman, T. L.; Patterson, A. P.; Duverger, N. J.; Santamarina- 
Fojo, S.; Brewer Jr., H. B. Apolipoprotein specificity for lipid efflux by the human 
ABCAI transporter. Biochem. Biophys. Res. Commun. 280:818–823; 2001. 
[48] Moreno, J. A.; Perez-Jimenez, F.; Marin, C.; Perez-Martinez, P.; Moreno, R.; Gomez, P.; 
Jimenez-Gomez, Y.; Paniagua, J. A.; Lairon, D.; Lopez-Miranda, J. The apolipoprotein E 
gene promoter (−219G/T) polymorphism determines insulin sensitivity in response 
to dietary fat in healthy young adults. J. Nutr. 135:2535–2540; 2005. 
[49] Xie, Q.; Zhao, S. P.; Li, F. D-4F, an apolipoprotein A-I mimetic peptide, promotes 
cholesterol efflux from macrophages via ATP-binding cassette transporter A1. 
Tohoku J. Exp. Med. 220:223–228; 2010. 
[50] Harel, M.; Aharoni, A.; Gaidukov, L.; Brumshtein, B.; Khersonsky, O.; Meged, R.; 
Dvir, H.; Ravelli, R. B.; McCarthy, A.; Toker, L.; Silman, I.; Sussman, J. L.; Tawfik, 
D. S. Structure and evolution of the serum paraoxonase family of detoxifying 
and anti-atherosclerotic enzymes. Nat. Struct. Mol. Biol. 11:412–419; 2004. 
[51] Perez-Jimenez, F.; Lopez-Miranda, J. University of Health Sciences: a blow of 
hope. Med. Clin. (Barcelona) 125:477; 2005. 
[52] Hou, M.; Xia,M.; Zhu, H.;Wang, Q.; Li, Y.; Xiao, Y.; Zhao, T.; Tang, Z.;Ma, J.; Ling,W. 
Lysophosphatidylcholine promotes cholesterol efflux from mouse macrophage
H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1381 
foam cells via PPARγ–LXRα–ABCA1-dependent pathway associated with apoE. Cell 
Biochem. Funct. 25:33–44; 2007. 
[53] Sorenson, R. C.; Primo-Parmo, S. L.; Kuo, C. L.; Adkins, S.; Lockridge, O.; La Du, B. N. 
Reconsideration of the catalytic center and mechanism of mammalian paraoxonase/ 
arylesterase. Proc. Natl. Acad. Sci. U. S. A. 92:7187–7191; 1995. 
[54] Aviram, M.; Billecke, S.; Sorenson, R.; Bisgaier, C.; Newton, R.; Rosenblat, M.; 
Erogul, J.; Hsu, C.; Dunlop, C.; La Du, B. Paraoxonase active site required for protec-tion 
against LDL oxidation involves its free sulfhydryl group and is different from 
that required for its arylesterase/paraoxonase activities: selective action of human 
paraoxonase allozymes Q and R. Arterioscler. Thromb. Vasc. Biol. 18:1617–1624; 
1998. 
[55] Fu, Y. Rate-limiting factors of cholesterol efflux in reverse cholesterol transport: 
acceptors and donors. Clin. Exp. Pharmacol. Physiol. 37:703–709; 2010. 
[56] Wang, N.; Lan, D.; Chen, W.; Matsuura, F.; Tall, A. R. ATP-binding cassette trans-porters 
G1 and G4 mediate cellular cholesterol efflux to high-density lipopro-teins. 
Proc. Natl. Acad. Sci. U. S. A. 101:9774–9779; 2004. 
[57] Drobnik, W.; Borsukova, H.; Bottcher, A.; Pfeiffer, A.; Liebisch, G.; Schutz, G. J.; 
Schindler, H.; Schmitz, G. Apo AI/ABCA1-dependent and HDL3-mediated lipid ef-flux 
from compositionally distinct cholesterol-based microdomains. Traffic 3: 
268–278; 2002. 
[58] Mendez, A. J.; Lin, G.; Wade, D. P.; Lawn, R. M.; Oram, J. F. Membrane lipid 
domains distinct from cholesterol/sphingomyelin-rich rafts are involved in the 
ABCA1-mediated lipid secretory pathway. J. Biol. Chem. 276:3158–3166; 2001. 
[59] Deakin, S. P.; Bioletto, S.; Bochaton-Piallat, M. L.; James, R. W. HDL-associated 
paraoxonase-1 can redistribute to cell membranes and influence sensitivity to 
oxidative stress. Free Radic. Biol. Med. 50:102–109; 2011.

More Related Content

What's hot

Biochemistry of blood bds
Biochemistry of blood  bdsBiochemistry of blood  bds
Biochemistry of blood bdsUrooj Arif
 
Krill yağının karaciğer üzerinde etkileri
Krill yağının karaciğer üzerinde etkileriKrill yağının karaciğer üzerinde etkileri
Krill yağının karaciğer üzerinde etkileriyekosan
 
Cholestrol metabolism
Cholestrol metabolismCholestrol metabolism
Cholestrol metabolismHamza Qureshi
 
Artificial blood - PFC & HBOC
Artificial blood - PFC & HBOCArtificial blood - PFC & HBOC
Artificial blood - PFC & HBOCManobalaSelvaraj2
 
Reverse cholesterol metabolism
Reverse cholesterol metabolismReverse cholesterol metabolism
Reverse cholesterol metabolismWajeeha123
 
Lipo proteins
Lipo proteinsLipo proteins
Lipo proteinsAli iqbal
 
Lipoproteins: Structure, classification, metabolism and significance
Lipoproteins:  Structure, classification, metabolism and significanceLipoproteins:  Structure, classification, metabolism and significance
Lipoproteins: Structure, classification, metabolism and significanceenamifat
 
Cholesterol synthesis, transport and excretion ppt 1
Cholesterol synthesis, transport and excretion ppt 1Cholesterol synthesis, transport and excretion ppt 1
Cholesterol synthesis, transport and excretion ppt 1Shailendra Shah
 
Abnormalities in lipoproteinemia
Abnormalities in  lipoproteinemiaAbnormalities in  lipoproteinemia
Abnormalities in lipoproteinemiaPranatiChavan
 
Lipoprotein metabolism - (transport of lipids in the Blood)
Lipoprotein metabolism - (transport of lipids in the Blood)Lipoprotein metabolism - (transport of lipids in the Blood)
Lipoprotein metabolism - (transport of lipids in the Blood)Ashok Katta
 
Biosynthesis of cholesterol
Biosynthesis of cholesterolBiosynthesis of cholesterol
Biosynthesis of cholesterolmadhu_mathi
 
Cholesterol metabolism (2)
Cholesterol metabolism (2)Cholesterol metabolism (2)
Cholesterol metabolism (2)Mahdi Albakaa
 
Is measuring apo B and Lp (a) of value ?
Is measuring apo B and Lp (a) of value ?Is measuring apo B and Lp (a) of value ?
Is measuring apo B and Lp (a) of value ?cardiositeindia
 

What's hot (20)

Biochemistry of blood bds
Biochemistry of blood  bdsBiochemistry of blood  bds
Biochemistry of blood bds
 
Krill yağının karaciğer üzerinde etkileri
Krill yağının karaciğer üzerinde etkileriKrill yağının karaciğer üzerinde etkileri
Krill yağının karaciğer üzerinde etkileri
 
Cholestrol metabolism
Cholestrol metabolismCholestrol metabolism
Cholestrol metabolism
 
Artificial blood - PFC & HBOC
Artificial blood - PFC & HBOCArtificial blood - PFC & HBOC
Artificial blood - PFC & HBOC
 
Reverse cholesterol metabolism
Reverse cholesterol metabolismReverse cholesterol metabolism
Reverse cholesterol metabolism
 
Lipo proteins
Lipo proteinsLipo proteins
Lipo proteins
 
Lipoproteins: Structure, classification, metabolism and significance
Lipoproteins:  Structure, classification, metabolism and significanceLipoproteins:  Structure, classification, metabolism and significance
Lipoproteins: Structure, classification, metabolism and significance
 
Cholesterol synthesis, transport and excretion ppt 1
Cholesterol synthesis, transport and excretion ppt 1Cholesterol synthesis, transport and excretion ppt 1
Cholesterol synthesis, transport and excretion ppt 1
 
Abnormalities in lipoproteinemia
Abnormalities in  lipoproteinemiaAbnormalities in  lipoproteinemia
Abnormalities in lipoproteinemia
 
Lipoprotein metabolism - (transport of lipids in the Blood)
Lipoprotein metabolism - (transport of lipids in the Blood)Lipoprotein metabolism - (transport of lipids in the Blood)
Lipoprotein metabolism - (transport of lipids in the Blood)
 
nihms-691736
nihms-691736nihms-691736
nihms-691736
 
Biosynthesis of cholesterol
Biosynthesis of cholesterolBiosynthesis of cholesterol
Biosynthesis of cholesterol
 
Cholesterol & its metabolism
Cholesterol & its metabolismCholesterol & its metabolism
Cholesterol & its metabolism
 
Lipid transport 2020
Lipid transport 2020Lipid transport 2020
Lipid transport 2020
 
Cholesterol
CholesterolCholesterol
Cholesterol
 
Blood
BloodBlood
Blood
 
Cholesterol metabolism (2)
Cholesterol metabolism (2)Cholesterol metabolism (2)
Cholesterol metabolism (2)
 
Abetalipoproteinemia
AbetalipoproteinemiaAbetalipoproteinemia
Abetalipoproteinemia
 
Lipoprotein
LipoproteinLipoprotein
Lipoprotein
 
Is measuring apo B and Lp (a) of value ?
Is measuring apo B and Lp (a) of value ?Is measuring apo B and Lp (a) of value ?
Is measuring apo B and Lp (a) of value ?
 

Similar to Berrougui 2012 purified human paraoxonase 1 interacts with plasma membrane lipid

2011. lactate biomarker and potential therapeutic target
2011. lactate   biomarker and potential therapeutic target2011. lactate   biomarker and potential therapeutic target
2011. lactate biomarker and potential therapeutic targetFlores Hdz
 
Halliwell et al., am. j. clin. nutr. 2005 health promotiion by flavonoids, to...
Halliwell et al., am. j. clin. nutr. 2005 health promotiion by flavonoids, to...Halliwell et al., am. j. clin. nutr. 2005 health promotiion by flavonoids, to...
Halliwell et al., am. j. clin. nutr. 2005 health promotiion by flavonoids, to...REVIDOX
 
Boros-JBR-2014-review
Boros-JBR-2014-reviewBoros-JBR-2014-review
Boros-JBR-2014-reviewEszter Tuboly
 
Yang_et_al-2016-ChemMedChem-Retro-1 Analogues Differentially Affect Oligonucl...
Yang_et_al-2016-ChemMedChem-Retro-1 Analogues Differentially Affect Oligonucl...Yang_et_al-2016-ChemMedChem-Retro-1 Analogues Differentially Affect Oligonucl...
Yang_et_al-2016-ChemMedChem-Retro-1 Analogues Differentially Affect Oligonucl...Bing Yang
 
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...Trustlife
 
Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositio...
Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositio...Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositio...
Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositio...Trustlife
 
ACS Med Chem 2011 (2) 919-923
ACS Med Chem 2011 (2) 919-923ACS Med Chem 2011 (2) 919-923
ACS Med Chem 2011 (2) 919-923Dinesh Barawkar
 
Effect of polyphenols on the intestinal and placental 2012
Effect of polyphenols on the  intestinal and placental 2012Effect of polyphenols on the  intestinal and placental 2012
Effect of polyphenols on the intestinal and placental 2012gisa_legal
 
Biochemistry and Biophysics Reports 25 (2021) 100921Availa
Biochemistry and Biophysics Reports 25 (2021) 100921AvailaBiochemistry and Biophysics Reports 25 (2021) 100921Availa
Biochemistry and Biophysics Reports 25 (2021) 100921AvailaChantellPantoja184
 
© 2010 El-Ansary et al, publisher and licensee Dove Medical Pr.docx
© 2010 El-Ansary et al, publisher and licensee Dove Medical Pr.docx© 2010 El-Ansary et al, publisher and licensee Dove Medical Pr.docx
© 2010 El-Ansary et al, publisher and licensee Dove Medical Pr.docxgerardkortney
 
SF424R-R_biosketch_VerC
SF424R-R_biosketch_VerCSF424R-R_biosketch_VerC
SF424R-R_biosketch_VerCBrooks Hylemon
 
Classes of lipoproteins and their role in the
Classes of lipoproteins and their role in the Classes of lipoproteins and their role in the
Classes of lipoproteins and their role in the Fikri Abdullah Zawawi
 
Κλούρας Ε, Λυμπερόπουλος Ε, Ελισάφ Μ. Αναστολή της PCSK9: Μια νέα προσέγγιση...
Κλούρας Ε, Λυμπερόπουλος Ε, Ελισάφ Μ.  Αναστολή της PCSK9: Μια νέα προσέγγιση...Κλούρας Ε, Λυμπερόπουλος Ε, Ελισάφ Μ.  Αναστολή της PCSK9: Μια νέα προσέγγιση...
Κλούρας Ε, Λυμπερόπουλος Ε, Ελισάφ Μ. Αναστολή της PCSK9: Μια νέα προσέγγιση...MedicalWeb.gr
 

Similar to Berrougui 2012 purified human paraoxonase 1 interacts with plasma membrane lipid (20)

2011. lactate biomarker and potential therapeutic target
2011. lactate   biomarker and potential therapeutic target2011. lactate   biomarker and potential therapeutic target
2011. lactate biomarker and potential therapeutic target
 
1246.full.pdf
1246.full.pdf1246.full.pdf
1246.full.pdf
 
FXR ADH1
FXR ADH1FXR ADH1
FXR ADH1
 
Halliwell et al., am. j. clin. nutr. 2005 health promotiion by flavonoids, to...
Halliwell et al., am. j. clin. nutr. 2005 health promotiion by flavonoids, to...Halliwell et al., am. j. clin. nutr. 2005 health promotiion by flavonoids, to...
Halliwell et al., am. j. clin. nutr. 2005 health promotiion by flavonoids, to...
 
Delaine posterfinal
Delaine posterfinalDelaine posterfinal
Delaine posterfinal
 
Boros-JBR-2014-review
Boros-JBR-2014-reviewBoros-JBR-2014-review
Boros-JBR-2014-review
 
Austin Proteomics
Austin ProteomicsAustin Proteomics
Austin Proteomics
 
Yang_et_al-2016-ChemMedChem-Retro-1 Analogues Differentially Affect Oligonucl...
Yang_et_al-2016-ChemMedChem-Retro-1 Analogues Differentially Affect Oligonucl...Yang_et_al-2016-ChemMedChem-Retro-1 Analogues Differentially Affect Oligonucl...
Yang_et_al-2016-ChemMedChem-Retro-1 Analogues Differentially Affect Oligonucl...
 
Ecmo pl os
Ecmo pl osEcmo pl os
Ecmo pl os
 
Advanced medicinal chemistry
Advanced medicinal chemistryAdvanced medicinal chemistry
Advanced medicinal chemistry
 
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...
Ellagic Acid and Its Metabolites as Potent and Selective Allosteric Inhibitor...
 
Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositio...
Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositio...Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositio...
Discovery of therapeutic agents targeting PKLR for NAFLD using drug repositio...
 
ACS Med Chem 2011 (2) 919-923
ACS Med Chem 2011 (2) 919-923ACS Med Chem 2011 (2) 919-923
ACS Med Chem 2011 (2) 919-923
 
Effect of polyphenols on the intestinal and placental 2012
Effect of polyphenols on the  intestinal and placental 2012Effect of polyphenols on the  intestinal and placental 2012
Effect of polyphenols on the intestinal and placental 2012
 
nihms641125
nihms641125nihms641125
nihms641125
 
Biochemistry and Biophysics Reports 25 (2021) 100921Availa
Biochemistry and Biophysics Reports 25 (2021) 100921AvailaBiochemistry and Biophysics Reports 25 (2021) 100921Availa
Biochemistry and Biophysics Reports 25 (2021) 100921Availa
 
© 2010 El-Ansary et al, publisher and licensee Dove Medical Pr.docx
© 2010 El-Ansary et al, publisher and licensee Dove Medical Pr.docx© 2010 El-Ansary et al, publisher and licensee Dove Medical Pr.docx
© 2010 El-Ansary et al, publisher and licensee Dove Medical Pr.docx
 
SF424R-R_biosketch_VerC
SF424R-R_biosketch_VerCSF424R-R_biosketch_VerC
SF424R-R_biosketch_VerC
 
Classes of lipoproteins and their role in the
Classes of lipoproteins and their role in the Classes of lipoproteins and their role in the
Classes of lipoproteins and their role in the
 
Κλούρας Ε, Λυμπερόπουλος Ε, Ελισάφ Μ. Αναστολή της PCSK9: Μια νέα προσέγγιση...
Κλούρας Ε, Λυμπερόπουλος Ε, Ελισάφ Μ.  Αναστολή της PCSK9: Μια νέα προσέγγιση...Κλούρας Ε, Λυμπερόπουλος Ε, Ελισάφ Μ.  Αναστολή της PCSK9: Μια νέα προσέγγιση...
Κλούρας Ε, Λυμπερόπουλος Ε, Ελισάφ Μ. Αναστολή της PCSK9: Μια νέα προσέγγιση...
 

Recently uploaded

Call Girls Doddaballapur Road Just Call 7001305949 Top Class Call Girl Servic...
Call Girls Doddaballapur Road Just Call 7001305949 Top Class Call Girl Servic...Call Girls Doddaballapur Road Just Call 7001305949 Top Class Call Girl Servic...
Call Girls Doddaballapur Road Just Call 7001305949 Top Class Call Girl Servic...narwatsonia7
 
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...CALL GIRLS
 
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service BangaloreCall Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalorenarwatsonia7
 
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...narwatsonia7
 
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaCall Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaPooja Gupta
 
Russian Call Girls Chennai Madhuri 9907093804 Independent Call Girls Service ...
Russian Call Girls Chennai Madhuri 9907093804 Independent Call Girls Service ...Russian Call Girls Chennai Madhuri 9907093804 Independent Call Girls Service ...
Russian Call Girls Chennai Madhuri 9907093804 Independent Call Girls Service ...Nehru place Escorts
 
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowSonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowRiya Pathan
 
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...narwatsonia7
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Miss joya
 
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...narwatsonia7
 
Aspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliAspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliRewAs ALI
 
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...narwatsonia7
 
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% SafeBangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safenarwatsonia7
 
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls ServiceCall Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Servicenarwatsonia7
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.MiadAlsulami
 
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...narwatsonia7
 
Ahmedabad Call Girls CG Road 🔝9907093804 Short 1500 💋 Night 6000
Ahmedabad Call Girls CG Road 🔝9907093804  Short 1500  💋 Night 6000Ahmedabad Call Girls CG Road 🔝9907093804  Short 1500  💋 Night 6000
Ahmedabad Call Girls CG Road 🔝9907093804 Short 1500 💋 Night 6000aliya bhat
 
Call Girl Service Bidadi - For 7001305949 Cheap & Best with original Photos
Call Girl Service Bidadi - For 7001305949 Cheap & Best with original PhotosCall Girl Service Bidadi - For 7001305949 Cheap & Best with original Photos
Call Girl Service Bidadi - For 7001305949 Cheap & Best with original Photosnarwatsonia7
 
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Miss joya
 

Recently uploaded (20)

Call Girls Doddaballapur Road Just Call 7001305949 Top Class Call Girl Servic...
Call Girls Doddaballapur Road Just Call 7001305949 Top Class Call Girl Servic...Call Girls Doddaballapur Road Just Call 7001305949 Top Class Call Girl Servic...
Call Girls Doddaballapur Road Just Call 7001305949 Top Class Call Girl Servic...
 
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
Call Girls Service Surat Samaira ❤️🍑 8250192130 👄 Independent Escort Service ...
 
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Whitefield Just Call 7001305949 Top Class Call Girl Service Available
 
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service BangaloreCall Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
Call Girl Bangalore Nandini 7001305949 Independent Escort Service Bangalore
 
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
Russian Call Girls Chickpet - 7001305949 Booking and charges genuine rate for...
 
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaCall Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
 
Russian Call Girls Chennai Madhuri 9907093804 Independent Call Girls Service ...
Russian Call Girls Chennai Madhuri 9907093804 Independent Call Girls Service ...Russian Call Girls Chennai Madhuri 9907093804 Independent Call Girls Service ...
Russian Call Girls Chennai Madhuri 9907093804 Independent Call Girls Service ...
 
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call NowSonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
Sonagachi Call Girls Services 9907093804 @24x7 High Class Babes Here Call Now
 
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
VIP Call Girls Tirunelveli Aaradhya 8250192130 Independent Escort Service Tir...
 
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
Call Girls Service Pune Vaishnavi 9907093804 Short 1500 Night 6000 Best call ...
 
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
Russian Call Girls in Bangalore Manisha 7001305949 Independent Escort Service...
 
Aspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas AliAspirin presentation slides by Dr. Rewas Ali
Aspirin presentation slides by Dr. Rewas Ali
 
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
Low Rate Call Girls Ambattur Anika 8250192130 Independent Escort Service Amba...
 
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% SafeBangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
Bangalore Call Girls Majestic 📞 9907093804 High Profile Service 100% Safe
 
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls ServiceCall Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
Call Girls Service Bellary Road Just Call 7001305949 Enjoy College Girls Service
 
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
Artifacts in Nuclear Medicine with Identifying and resolving artifacts.
 
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
Call Girls Service in Bommanahalli - 7001305949 with real photos and phone nu...
 
Ahmedabad Call Girls CG Road 🔝9907093804 Short 1500 💋 Night 6000
Ahmedabad Call Girls CG Road 🔝9907093804  Short 1500  💋 Night 6000Ahmedabad Call Girls CG Road 🔝9907093804  Short 1500  💋 Night 6000
Ahmedabad Call Girls CG Road 🔝9907093804 Short 1500 💋 Night 6000
 
Call Girl Service Bidadi - For 7001305949 Cheap & Best with original Photos
Call Girl Service Bidadi - For 7001305949 Cheap & Best with original PhotosCall Girl Service Bidadi - For 7001305949 Cheap & Best with original Photos
Call Girl Service Bidadi - For 7001305949 Cheap & Best with original Photos
 
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
Russian Call Girls in Pune Riya 9907093804 Short 1500 Night 6000 Best call gi...
 

Berrougui 2012 purified human paraoxonase 1 interacts with plasma membrane lipid

  • 1. Free Radical Biology & Medicine 52 (2012) 1372–1381 Contents lists available at SciVerse ScienceDirect Free Radical Biology & Medicine journal homepage: www.elsevier.com/locate/freeradbiomed Original Contribution Purified human paraoxonase-1 interacts with plasma membrane lipid rafts and mediates cholesterol efflux from macrophages Hicham Berrougui a,b, Soumaya Loued a,b,c, Abdelouahed Khalil a,b,c,⁎ a Research Center on Aging, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada QC J1H 4C4 b Department of Medicine, Geriatrics Service, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Canada QC J1H 4C4 c Programs of Physiology, Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC Canada J1H 4C4 a r t i c l e i n f o a b s t r a c t Article history: Received 12 September 2011 Revised 17 January 2012 Accepted 23 January 2012 Available online 1 February 2012 Keywords: Paraoxonase-1 HDL Cholesterol efflux Lipid raft Free radicals Macrophages Paraoxonase-1 (PON1) is a high-density lipoprotein (HDL)-associated serum enzyme thought to make a major contribution to the antioxidant and anti-inflammatory capacities of HDLs. However, the role of PON1 in the modulation of cholesterol efflux is poorly understood. The aim of our study was to investigate the in-volvement of PON1 in the regulation of cholesterol efflux, especially the mechanism by which it modulates HDL-mediated cholesterol transport. The enrichment of HDL3 with human PON1 enhanced, in a dose-dependent manner, cholesterol efflux from THP-1 macrophage-like cells and ABCA1-enriched J774 macro-phages. Moreover, an additive effect was observed when ABCA1-enriched J774 macrophages were incubated with both PON1 and apo-AI. Interestingly, PON1 alone was able to mediate cholesterol efflux from J774 mac-rophages and to upregulate ABCA1 expression on J774 macrophages. Immunofluorescence measurement showed an increase in PON1 levels in the cytoplasm of J774 macrophages overexpressing ABCA1. PON1 used an apo-AI-like mechanism to modulate cholesterol efflux from rapid and slow efflux pools derived from the lipid raft and nonraft domains of the plasma membrane, respectively. This was supported by the fact that ABCA1 protein was incrementally expressed by J774 macrophages within the first few hours of in-cubation with cholesterol-loaded J774 macrophages and that cyclodextrin significantly inhibited the capacity of PON1 to modulate cholesterol efflux from macrophages. This finding suggested that PON1 plays an impor-tant role in the antiatherogenic properties of HDLs and may exert its protective function outside the lipopro-tein environment. © 2012 Elsevier Inc. All rights reserved. It has long been recognized that the concentration of plasma high-density lipoproteins (HDLs) is inversely related to the risk of cardio-vascular disease. The atheroprotective effect of HDLs is largely attrib-uted to its key role in reverse cholesterol transport (RCT) whereby excess cholesterol from peripheral cells is transported back to the liver for excretion [1]. Macrophages possess a number of mechanisms to regulate the equilibrium between cholesterol uptake/synthesis and export, including transport mechanisms that promote the efflux of excess cholesterol to extracellular acceptors [1]. Although cholesterol efflux from macrophages makes up only a small fraction of overall flux through the RCT pathway, it is considered a relevant component of atheroprotection [1]. HDL-mediated cholesterol efflux is the natu-ral rate-limiting step of RCT [2] and occurs via three pathways. The first pathway is aqueous diffusion by which free cholesterol mole-cules spontaneously desorb from the plasma membrane, diffuse through the aqueous phase, and become adsorbed on acceptor particles by collision [3]. The second pathway involves scavenger re-ceptor class B type I (SR-BI)-mediated bidirectional free cholesterol exchanges depending on the cholesterol gradient, this pathway medi-ates cholesterol efflux to a wide variety of cholesterol acceptors [4]. The third pathway involves the ATP-binding cassette receptors ABCA1 and ABCG1, which mediate cholesterol efflux in a unidirec-tional manner to lipid-poor apolipoprotein-AI (apo-AI) and to other subfamily members of HDL, respectively [5–7]. The rate of cholesterol efflux, however, depends also on the capac-ity of cells to release excess cholesterol and of plasma acceptors to transport the released cholesterol, which is determined by the con-centration and physical and functional states of HDLs and apo-AI [2]. Apo-AI is a major protein component of HDL and is a critical ele-ment in cholesterol metabolism. Apo-AI reacts specifically with ABCA1 to generate nascent HDLs, which are then enriched with ester-ified cholesterol by LCAT (activated by apo-AI) to form mature spher-ical HDLs [8]. A number of other proteins confer additional properties on HDLs. Vaisar et al. identified 48 proteins in HDLs associated with clinically significant cardiovascular disease [9]. Paraoxonase 1 (PON1) is one of the 8 most abundant HDL-associated proteins and may have atheroprotective properties [9]. PON1 has been extensively ⁎ Corresponding author at: Research Center on Aging, 1036 rue Belvédère Sud, Sher-brooke, QC, Canada J1H 4C4. Fax: +1 819 829 7141. E-mail address: Abdelouahed.Khalil@USherbrooke.ca (A. Khalil). 0891-5849/$ – see front matter © 2012 Elsevier Inc. All rights reserved. doi:10.1016/j.freeradbiomed.2012.01.019
  • 2. H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1373 studied with respect to its association with cardiovascular risk, oxida-tive stress, and inflammation [10]. PON1 is mainly synthesized in the liver and 95% is transferred to HDLs via an SR-BI-mediated mechanism. Small amounts of plasma PON1 (5%) are associated with chylomicrons and very low density li-poproteins [11–13]. In vitro studies have shown that PON1 inhibits low-density lipoprotein (LDL) and HDL lipid peroxidation and may be a primary determinant of the anti-inflammatory capacity of HDLs [14,15]. PON1 also attenuates oxidized LDL uptake via scavenger re-ceptor CD-36 and directly reduces oxidative stress in macrophages [16,17]. It also inhibits oxidized LDL-induced MCP-1 production via a mechanism that is probably related to its capacity to hydrolyze some activated phospholipids as well as lipid peroxide [18]. Cys284 of the free sulfhydryl of PON1 is required for its atheroprotective ef-fect and is thought to be the active site for its antioxidant activity [19]. We previously showed that exposure of purified human PON1 to oxygen free radicals induced a significant decrease in PON1 para-oxonase and antioxidant activities, which was accompanied by a de-crease in free sulfhydryl groups [19]. PON1 expression is inversely correlated with atherosclerotic de-velopment in animal models [20,21]. PON1 accumulates in arterial walls as human plaque progresses from fatty streaks into advanced lesions. This process is associated with macrophages and seems to protect against the increasing oxidation associated with plaque pro-gression [22]. Treating human carotid lesion extracts with recombi-nant PON1 significantly decreases the oxidative potential of the extracts [23]. These properties may be the main explanation for the beneficial effect of PON1 on the atherosclerotic process [24]. Recent research has focused on investigating the potential anti-atherogenic role played by PON1 in HDL-mediated cholesterol efflux from macrophages [25]. Rosenblat et al. suggested that PON1 in-creases lysophosphatidylcholine (lyso-PC) formation, which in turn stimulates HDL binding and HDL-mediated cholesterol efflux [26]. The H115Q and H134Q histidine residues of PON1 seem to be re-quired for this process [25]. Lipid rafts are cholesterol- and sphingolipid-rich lateral domains containing approximately double the molar percentages of these lipids and much lower proportions of protein than other areas of the plasma membrane [27]. Research on the role of lipid rafts in cho-lesterol transport and homeostasis has concentrated mainly on caveolin-1, a structural protein in a subset of lipid rafts (caveolae) that directly interacts with cholesterol [28]. Although caveolin-1 is a useful marker for isolating rafts and seems to be involved in intracel-lular cholesterol transport, there is some controversy concerning the role of caveolae cholesterol in cholesterol efflux [29], given that caveolin-1 is not expressed in primary human macrophages or J774 macrophages [30] and that cholesterol efflux in these cells normally occurs via apo-AI [31]. Gaus et al. recently demonstrated that choles-terol exported to apo-AI from the major slow efflux pool comes from nonraft regions of the plasma membrane and that the interaction of apo-AI with lipid rafts is required to stimulate this efflux [28]. We investigated the role of purified human PON1 on HDL-mediated and non-HDL-mediated cholesterol efflux and propose an explanation of the mechanism by which PON1 enhances cholesterol efflux, especially via its interaction with lipid and nonlipid raft domains. We provide further evidence that the PON1–lipid raft inter-action is an essential event in PON1-mediated cholesterol efflux. Material and methods Acetic acid, sulfuric acid, sodium phosphate, thiobarbituric acid, n-butanol, methanol, ethanol, n-isopropanol, hexane, ammonium hydroxide, chloroform, and methanol were purchased from Fisher Scientific (Montreal, QC, Canada). N-ethylmaleimide (NEM), 1,1,3,3,- tetraethoxypropane, ethylenediaminetetraacetic acid (EDTA), 1,6- diphenyl-1,3,5-hexatriene, phosphatidylcholine, sphingomyelin, 8- (4-chlorophenylthio)adenosine 3′:5′-cyclic monophosphate (cAMP), methyl-β-cyclodextrin (MβCD), and [3H]cholesterol were from Sigma (St. Louis,MO, USA). THP-1 and J774 cellswere fromtheAmerican Type Culture Collection (Manassas, VA, USA). RPMI 1640 and Dulbecco's modified Eagle's medium (DMEM) were from Invitrogen Canada (Burlington, ON, Canada). Fetal bovine serum(FBS) was fromWisent (St-Bruno, QC, Canada). Subjects Plasma samples were obtained from young healthy volunteers 20 to 30 years of age. They were all healthy, normolipidemic non-smokers and nonobese. None had clinical or laboratory signs of hy-pertension, inflammation, or diabetes, and all had normal thyroid function test results. None were taking medications or oral antioxi-dant supplements. The ethics committee of the Sherbrooke Geriatric University Institute approved the study. All the volunteers provided written informed consent. Blood collection After overnight fasting, 80-ml blood samples were collected in EDTA or citrate vacuum tubes. The plasma was separated by low-speed centrifugation (1000 g), and 15 ml was used immediately to isolate lipoproteins. The remaining plasma was stored at −80 °C until used for the PON1 purification procedure. Lipoprotein isolation Human plasma was collected and HDL and HDL3 were isolated by sequential ultracentrifugation according to the method of Sattler et al. [32]. In brief, whole HDL (1.063bdb1.19 g/ml) and HDL3 (1.125b db1.21 g/ml) were isolated by respectively 2 and 4 h of ultracentrifu-gation in a Beckman TLA 100.4 rotor (100,000 rpm at 15 °C). Isolated lipoproteins were dialyzed overnight at 4 °C in 10 mM sodium phosphate buffer (pH 7). Protein concentrations in the samples were measured using a commercial assay (Bio-Rad, Mississauga, ON, Canada). Cell cultures Human THP-1 monocytes and J774 macrophages were grown in RPMI 1640, and Fu5AH hepatoma cells were grown in DMEM. The media were supplemented with 10% heat-inactivated FBS, 50 nM 2-mercaptoethanol (only for THP-1 cells), 2 mM L-glutamine, 100 U/ ml penicillin, and 1.5 mg/ml glucose in a humidified atmosphere (5% CO2 and 95% air) at 37 °C. The THP-1 monocytes (105 cells/cm2 in six-well plates) were incubated in RPMI–FBS containing 10 ng/ml phorbol 12-myristate 13-acetate for 96 h to induce differentiation into adherent macrophage-like cells. Paraoxonase purification PON1 was purified using blue agarose and DEAE chromatography as described by Gan et al. [33], with some modifications. Briefly, plas-ma was mixed with blue agarose (Cibadron Blue 3GA; Sigma–Aldrich, Oakville, ON, Canada) in a solution containing 3 M NaCl, 50 mM Tris– HCl buffer (pH 8), 1 mM CaCl2, and 5 μM EDTA. The PON1 was eluted with 0.1% deoxycholate. The blue agarose-eluted PON1 was further purified by anion-exchange chromatography (DEAE-Biogel; Sigma) using a NaCl linear gradient. The paraoxonase and arylesterase activities of the purified PON1 were measured as described previously [33,34]. The purity of PON1 was verified by SDS–PAGE.
  • 3. 1374 H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 Cholesterol efflux measurements THP-1 macrophage-like cells, J774 macrophages, and Fu5AH cells were incubated in fresh medium containing 2 μCi/ml [3H]cho-lesterol for 24 h. Labeled cells were washed and equilibrated in serum-free medium containing 1% bovine serum albumin (BSA) for an additional 12 h. To evaluate the interaction between PON1 and the ABCA1 receptor, [3H]cholesterol-loaded J774 macrophages were washed three times and incubated with 1% BSA in DMEM alone (control) or with DMEM containing 0.3 mM cAMP for 12 h to yield ABCA1-enriched cells (cAMP stimulates ABCA1 gene tran-scription and surface protein expression) [35]. After the equilibra-tion period, the THP-1 macrophage-like cells and J774 macrophages were washed three times before being incubated with various cholesterol acceptors (native or oxidized PON1, HDL3, apo-AI) depending on the experimental design. The cells were sedi-mented by centrifugation (350 g for 10 min) and were lysed in 0.1 M NaOH. The counts per minute (cpm) in the supernatant and cell lysates were determined using a liquid scintillation counter. Cholesterol efflux (radiolabeled cholesterol released from cells) was calculated using the following formula: (radioactivity (cpm) in supernatant/radioactivity (cpm) in cells+medium)×100. Inactivation of PON1 PON1 was inactivated by exposure to oxygen free radicals pro-duced by γ-radiolysis of water. Exposure of PON1 to oxygen free rad-icals has been shown to oxidize PON1 and inhibits its enzymatic activity without causing protein fragmentation [19]. PON1 was also inactivated by heat (1 h at 70 °C) or with 10 μM NEM, which scav-enges the free sulfhydryl group on PON1 (Cys284) [19]. The inactiva-tion of PON1 was confirmed by measuring PON1 activity. Lipid raft extraction Lipid rafts were extracted as previously described [36]. Briefly, cells were resuspended in 300 μl of ice-cold lipid raft lysis buffer and mixed with 300 μl of 85% sucrose in Hepes-buffered saline solu-tion (pH 6.9). The samples were overlaid with 1 ml of 35% sucrose and then 300 μl of 5% sucrose. The preparations were centrifuged at 200,000 g at 4 °C for 16 h using a Beckman TLA-100.4 rotor (Beckman Instruments, Montreal, QC, Canada). Nine 200-μl fractions were col-lected from the top of the gradient. They were separated on 10% SDS–PAGE gels and were transferred to nitrocellulose membranes, which were blotted and incubated with anti-flotillin antibody. Immunofluorescence microscopy J774 macrophages were seeded in eight-well glass culture slides (BD Falcon, Bedford, MA, USA) at a concentration of 2.5×105 cells per well and were incubated with 0.3 mM cAMP for 12 h to generate ABCA1-enriched cells. The enriched cells were incubated with 20 U/ ml PON1 for 4 h, washed twice with PBS, fixed with 4% formaldehyde for 10 min, and incubated with 0.1% Triton X-100 in PBS for 2 min. After being washed with PBS, the cells were incubated with blocking solution (5% goat serum, 5% BSA, and 0.01% Tween 20) and then with primary antibody in blocking solution for 16 h at 4 °C. Alexa 488- labeled goat anti-mouse IgG was used as the secondary antibody. The slides were washed, stained with DAPI, and mounted in Vecta-shield (Vector Laboratories, Burlington, ON, Canada) to visualize the shapes of the nuclei. Western blotting Cell proteins were solubilized in RIPA buffer. Identical amounts of cell lysate protein were separated on 10% SDS–PAGE gels and were transferred to nitrocellulose membranes. The membranes were blocked with 5% fat-free powdered milk in TBST (10 mM Tris–HCl, pH 7.4, 150 mM NaCl, 0.1% Tween 20) and then incubated with ABCA1, ABCG1, or SR-BI primary antibodies (Abcam, Cambridge, MA, USA). After three washes with TBST, the membranes were incu-bated with horseradish peroxidase-conjugated goat anti-rabbit, goat anti-mouse IgG, or rabbit anti-goat IgG (Sigma), and the signals were visualized using the ECL Western blotting system (GE Health-care, Piscataway, NJ, USA). Coimmunoprecipitation assays Coimmunoprecipitation was used to confirm the formation of ABCA1–PON1 complex in lysates from J774 macrophages. cAMP (0.3 mM) was used to stimulate ABCA1 overexpression on J774 mac-rophages. PON1 (20 U/ml) was added to ABCA1-enriched or none-nriched J774 macrophages for 3 h and then cell lysates were prepared with RIPA buffer and protease inhibitor cocktail. Immuno-precipitations were carried out using Dynabeads protein G (Invitro-gen) according to the manufacturer's protocol. Briefly, 1.5 mg of Dynabeads was incubated with 5 μg of ABCA1 monoclonal antibody (mAb) for 60 min. After overnight incubation at 4 °C of Dynabeads– mAb complex with cell lysates, beads were washed three times with PBS and then 20 μl of 2× sample buffer (100 mM Tris, pH 6.8, 40 g/L SDS, 200 ml/L glycerol, 20 mg/ml bromophenol blue, 0.05% 2- mercaptoethanol) was added to the beads. The sample/supernatant was separated from the beads by placing the tube on a magnet and then loaded on the SDS–PAGE (10%) gel. After being transferred to a polyvinylidene difluoride membrane, PON1 was detected using an anti-PON1 mouse monoclonal antibody (Abcam). Quantitative RT-PCR Total RNA from J774 macrophages treated with 20 U/ml PON1 for 0, 1, 3, or 16 h was extracted using RNeasy extraction mini kits (Invi-trogen) and treated with DNase I (Qiagen, Mississauga, ON, Canada) according to the manufacturer's protocol. Two micrograms of RNA was transcribed using Reverse Transcriptase Superscript II (Invitro-gen). The expression of the ABCA1 gene was normalized to the corre-sponding amount of β-actin. Amplifications were performed using the following primers (sense and antisense): ABCA1, 5′-TCATCTT-CATCTGCTTCCAGC- 3′ and 5′-GTGCTGGGGATCTTGAACAC-3′; β-actin, 5′-GAACGGTGAAGGTGACA-3′ and 5′-TAGAGAGAAGTGGGGTGG-3′. The quantitative PCR assays were performed using the Stratagene MX3005P system (Agilent Technologies, Mississauga, ON, Canada) and Brilliant II SYBR Green QPCR Master Mix (Agilent). The qPCR as-says were performed using 25 ng of template cDNA. Samples were in-cubated at 95 °C for 10 min followed by 45 cycles using the following conditions: 95 °C for 40 s, 56 °C for 40 s, and 72 °C for 40 s. All reac-tions were run in triplicate for each replicate, and the average values were used for quantification purposes. The relative quantities of tar-get genes were determined using the ΔΔCt method. Briefly, the Ct (threshold cycle) values of target genes were normalized to an en-dogenous control gene, β-actin (ΔCt=Ct target−Ct β-actin), and were compared with a calibrator (ΔΔCt=ΔCt sample−ΔCt calibrator). Relative expression (RQ) was calculated using a sequence detection system (MxPro and QPCR software; Agilent) and the formula RQ=2−ΔΔCt. Statistical analysis Values are expressed as means ± SEM. A one-way analysis of var-iance was used for multiple comparisons. A linear regression analysis was used to assess the association between two continuous variables. Statistical analyses were performed using Prism 5.0 version software. A p value of b0.05 was considered statistically significant.
  • 4. Results PON1 has an effect on HDL-mediated cholesterol efflux HDL-associated proteins other than apo-AI can stimulate HDL-mediated cholesterol efflux [37,38]. The aim of this study was to in-vestigate the role of PON1 in cholesterol efflux. Loading THP1 macrophage-like cells with [3H]cholesterol and purified human plas-ma PON1 5, 10, and 20 U/ml (corresponding to 12.5, 25, and 50 μg/ml of protein) increased HDL3-mediated cholesterol efflux from THP-1 macrophage-like cells by 20.8, 24, and 63% (pb0.05), respectively (Fig. 1A). The increase in cholesterol efflux from THP1 macrophage-like cells was dependent on the concentration of PON1 (r2=0.91, pb0.05, data not shown). It is noteworthy that the physiological con-centration of human plasma PON1 is between 50 and 100 μg/ml pro-tein [39,40]. Although PON1 significantly increased cholesterol efflux to J774 macrophages (Fig. 1B) in a concentration-dependent manner (r2=0.94, pb0.05, data not shown), stimulating J774 macrophages with cAMP to induce ABCA1 overexpression significantly potentiated the effect (r2=0.97, pb0.05). A comparison of the slopes of the two correlations shows that cholesterol efflux increased 1.82-fold when PON1 was added to cAMP-stimulated J774 macrophages compared to unstimulated cells (1.03±0.11 vs 0.55±0.1, respectively, pb0.05). This suggests that PON1 enhances HDL3-mediated choles-terol efflux and that the ABCA1 transporter pathway mediates this effect. PON1 is a novel independent transporter involved in cholesterol efflux We performed experiments in more than one cell type to deter-mine which of the three cholesterol efflux pathways might be stimu-lated by PON1. PON1-mediated cholesterol efflux was first studied in THP-1 macrophage-like cells, which equally express ABCA1/ABCG1 transporters and the SR-BI receptor. Our results show that incubation of cholesterol-loaded THP-1 with 0, 5, 10, or 20 U/ml PON1 alone re-sults in a significant increment in cholesterol efflux by 2.5-, 3-, and 4.6-fold compared to the control (Fig. 2A). To better understand the mechanism by which PON1 mediates cholesterol efflux, we investigated the role of the ABCA1 pathway in this process. For that purpose we used cAMP-pretreated J774 macro-phages, which express high levels of ABCA1 [4]. ABCA1-enriched and nonenriched J774 macrophages (control) were incubated with in-creasing concentrations of PON1. The results show that PON1 signifi-cantly promoted cholesterol efflux in ABCA1-enriched macrophages in a concentration-dependent manner, whereas no significant in-crease was observed in the control (Fig. 2B). Taking into account that PON1 and apo-AI are both associated with HDL, we investigated a possible synergistic effect between apo-AI and PON1 to mediate cholesterol efflux. For that purpose, we incubated [3H]cholesterol-loaded ABCA1-enriched J774 cells with in-creasing concentrations of PON1 (0, 5, 10, and 20 U/ml) in the pres-ence or not of apo-AI (25 μg/ml) for 4 h. Results in Fig. 2C, shows that apo-AI-mediated cholesterol efflux was significantly potentiated, and in a dose-dependent manner, in the presence of PON1 by an av-erage of 37.24±5.35% (pb0.01). The incubation of cholesterol-loaded ABCA1-enriched J774 with increasing concentrations of BSA (negative control) had no effect, whereas 25 μg/ml apo-AI (positive control) significantly enhanced cholesterol efflux (data not shown). This confirms that PON1 en-hances ABCA1-dependent cholesterol efflux from J774 macrophages. To assess SR-BI-mediated cholesterol efflux, we used Fu5AH rat hepatoma cells, which express high levels of SR-BI and lack a func-tional ABCA1 [41]. Interestingly, incubating PON1 (0, 5, 10, or 20 U/ ml) with SR-BI-overexpressing Fu5AH cells did not cause a significant change in cholesterol efflux (Fig. 2D). This effect was nonsignificant even when PON1 was incubated in the presence of HDL (50 μg/ml), which leads to the suggestion that the SR-BI receptor was not or was less implicated in the PON1-mediated cholesterol efflux. ABCA1 overexpression increases PON1 internalization cAMP-treated J774 macrophages exhibited more intense immuno-fluorescence than the control cells (Figs. 3A and B), indicating that ABCA1 overexpression increases the internalization of PON1, which leads to its localization in the cell's cytoplasm (Fig. 3C, 100× original magnification). This is in accordance with the results reported by Efrat and Aviram [42], who used FITC fluorescence and confocal mi-croscopy to demonstrate an internalization of PON1 and its localiza-tion in the cell's cytoplasm compartment. ABCA1-enriched and nonenriched J774 macrophages were incu-bated with PON1 for 3 h. PON1 was detected in the ABCA1 A 40 30 20 10 0 * * ** + + + + HDL3 [50μg/ml] 0 5 10 20 PON1 [U/ml] Cholesterol efflux (%) 30 20 10 0 ns * ** ** + + + + + + + + HDL3 [50 μg/ml] - + - + - + - + cAMP [0.3 mM] 0 5 10 20 PON1 [U/ml] B Cholesterol efflux (%) Fig. 1. HDL3-associated PON1 mediates cholesterol efflux from THP-1 and J774 macro-phages. (A) THP-1 macrophages were loaded with [3H]cholesterol (2 μCi/ml) for 24 h. The cells were then washed and further incubated for 24 h with 50 μg/ml HDL3 alone (control) or in the presence of increasing concentrations of PON1 (0–20 U/ml; 0–50 μg/ml PON1 protein concentration). (B) [3H]Cholesterol-loaded J774 macro-phages were incubated overnight at 37 °C without (control) or with 0.3 mM cAMP (ABCA1-enriched cells). After being washed, the cells were incubated for 24 h with HDL3 alone or with HDL3 supplemented with increasing concentrations of PON1 (0–20 U/ml; 0–50 μg/ml). Results are expressed as the means ± SEM of three indepen-dent experiments. *pb0.05, **pb0.01. H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1375
  • 5. 1376 H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 0 5 10 20 PON1 [U/ml] 5 10 20 5 10 20 0 5 10 15 20 25 0 5 10 20 20 + 20 15 10 5 0 ** THP1 Cholesterol efflux (%) 15 10 5 PON1 cAMP - + + 0 0 0 0 0 0 - - - + + + + + Magnification (x 100) ** *** 10 8 6 4 2 0 Cholesterol efflux (%) apo-AI 25μg/ml 15 10 5 A B C Fig. 3. ABCA1 overexpression increases PON1 internalization. (A) Control and (B and C) ABCA1-enriched J774 cells (original magnification 40× and 100×, respectively) were incu-bated with PON1 (20 U/ml; 50 μg/ml) before their fixation with 4% paraformaldehyde and their immunostaining with primary antibody for PON1 (mouse). The cells were then incubated with Alexa 488-labeled goat anti-mouse IgG secondary antibody and processed for immunofluorescence microscopy. PON1 [U/ml] ** *** *** J774 HDL cAMP [0.3 mM] 0 apo-AI-free PON1 [U/ml] Cholesterol efflux (%) 0 ns Fu5AH ns PON1 [U/ml] HDL [μg/ml] 0 50 50 0 0 0 0 Cholesterol efflux (%) ns A B C D Fig. 2. Effects of purified PON1 on the cholesterol efflux. (A) [3H]Cholesterol-loaded THP-1 cells were incubated with PON1 (0–20 U/ml; 0–50 μg/ml) for 24 h. (B) J774 macrophages were loaded with [3H]cholesterol and then incubated for 12 h in the presence of cAMP to yield ABCA1 expression. Cholesterol efflux from ABCA1-enriched and nonenriched J774 was then initiated by the addition of PON1 (0–20 U/ml; 0–50 μg/ml) for 4 h. (C) [3H]Cholesterol-loaded and ABCA1-enriched J774 cells were incubated for 4 h with increasing con-centrations of PON1 (0–50 μg/ml) in presence or not of apo-AI. (D) [3H]Cholesterol-loaded Fu5AH cells were incubated with PON1 (0–20 U/ml; 0–50 μg/ml) and/or HDL3 (50 μg/ml, positive control) for 24 h. To investigate if PON1 require HDL for its interaction with SR-BI receptors, [3H]cholesterol-loaded Fu5AH cells were incubated with PON1 (20 U/ml; 50 μg/ ml) in the presence of HDL3 (50 μg/ml) for 24 h. Results are expressed as the means ± SEM of more than three independent experiments. **pb0.01, ***pb0.001; ns, nonsignificant.
  • 6. PON1 (20 U/ml) 1 2 + + immunoprecipitate, with the highest amount in the precipitate from ABCA1-enriched J774 (Fig. 4), suggesting that PON1 binds with ABCA1 to form a protein complex. We previously showed that the oxidation of purified human PON1 causes a decrease in its antioxidant and paraoxonase activities [19]. This effect was attributed to the loss of the free sulfhydryl group [19]. In this study, we show that exposure of PON1 to oxygen free rad-icals produced by γ-radiolysis affects significantly its ability to pro-mote HDL3-mediated cholesterol efflux from THP-1 macrophage-like cells (−29.04%, pb0.01, Fig. 5A). In addition, heat (70 °C for 1 h) and NEM treatment, which irreversibly blocks SH groups by covalent binding [43], significantly decreased (pb0.01) paraoxonase activity as well as its ability to promote cholesterol efflux, especially from ABCA1-enriched macrophages (Fig. 5B). PON1 interacts with lipid raft domains to initiate cholesterol efflux from macrophages In the second part of our study, we investigated the capacity of PON1 to interact with lipid rafts and to mediate cholesterol efflux. Su-crose gradients separated the plasma membranes into two fractions: 40 30 20 10 0 * A Cholesterol efflux (%) 10 8 6 4 2 0 HDL3-nPON1 [20U/ml] HDL3-oxPON1 [20U/ml] ** ** nPON1 Heat-inactivated PON1 NEM-inactivated PON1 B Cholesterol efflux (%) Fig. 5. PON1 oxidation impairs its capacity to stimulate HDL-mediated cholesterol efflux. (A) [3H]Cholesterol-loaded THP-1 macrophages were incubated with HDL3 in the presence of native (nPON1) or oxidized PON1 (oxPON1) for 24 h. Oxidation of PON1 was induced by its exposure to oxygen free radicals produced by γ-radiolysis of water. (B) Denaturing PON1 reduces its capacity to interact with ABCA1 receptors and to promote cholesterol efflux. 20 U/ml (50 μg/ml) PON1 was inactivated by heat (70 °C/1 h) or by NEM treatment (1 h) before starting the experiments. J774 macro-phages were loaded with [3H]cholesterol and treated with cAMP to upregulate ABCA1 expression. Treated and control cells were incubated with native PON1, heat-inactivated PON1, or NEM-treated PON1 for 4 h. Data are given as means ± SEM of more than three independent experiments. *pb0.05, **pb0.01. 8×10 04 6×10 04 4×10 04 2×10 04 2.0×1004 1.5×1004 1.0×1004 5.0×1003 Control * apoA1 PON1 Control apoA1 PON1 0.0 Raft fractions ** * Cholesterol contents (cpm) 12 ** 9 6 3 Control-CD free apoA1-CD PON1-CD free Control-CD apoA1-CD free PON1-CD 0 * Cholesterol efflux (%) A B Non-Raft fractions Fig. 6. PON1 and apo-AI start mediating cholesterol efflux from lipid raft domains. THP- 1 macrophages were loaded with [3H]cholesterol and then incubated for 4 h with apo- AI or PON1. Plasma membranes were isolated on a sucrose gradient as described under Material and methods. (A) Cholesterol contents (cpm) in associated lipid raft and non-raft fractions. (B) [3H]Cholesterol-loaded and ABCA1-enriched J774 macrophages were pretreated for 1 h at 37 °C with medium containing BSA alone or plus methyl-β- cyclodextrin (CD; 2 mg/ml). After the above preincubations, the media were removed and efflux to fresh medium containing or not apo-AI or PON1 over 4 h was measured. Results are expressed as the means ± SEM of more than three independent experi-ments. *pb0.05, **pb0.01 vs control. PON1 45Kda cAMP + Fig. 4. Coimmunoprecipitation of ABCA1 and PON1 in lysates from J774 macrophages shows that PON1 binds to ABCA1. PON1 (20 U/ml) was added to ABCA1-enriched (lane 1) or nonenriched (lane 2) J774 for 3 h and then ABCA1 immunoprecipitation (IP) from cell lysates was performed using Dynabeads protein-G and ABCA1 monoclo-nal antibody. ABCA1 IP was followed by PON1 Western blot (10% SDS–PAGE) using mouse anti-human PON1 monoclonal antibody. H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1377
  • 7. 1378 H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 light membranes (rafts) and heavy membranes (nonrafts). Western blotting using fotilin-1, which migrated with the rafts, confirmed the separation (data not shown). Incubating [3H]cholesterol-loaded THP-1 macrophage-like cells with PON1 and apo-AI resulted in a significant decrease in the choles-terol content of the lipid raft fractions, by approximately 38.0 (pb0.01) and 20.5% (pb0.05), respectively. However, in the nonraft fractions, cholesterol decreased by only 23.1 and 12.3% (pb0.05) in the presence of PON1 and apo-AI, respectively (Fig. 6A). This sug-gested that PON1 interacts with lipid rafts to initiate cholesterol ef-flux from macrophages. To gain more insight into the mechanismby which the PON1–lipid raft interaction modulates cholesterol efflux, we studied the effect of a short MβCD pretreatment on the increase in cholesterol efflux from cholesterol-loaded macrophages by PON1 and apo-AI (control). The MβCD treatment caused a selective removal of cholesterol from lipid raft domains and induced the dispersal of their lipid and protein components [28]. Interestingly, the exposure of ABCA1-enriched J774 macrophages to MβCD for 1 h resulted in a significant impairment of the cholesterol efflux mediated by PON1 (57.21%, pb0.05) and by apo-AI (63.89%, pb0.01) (Fig. 6B). This suggested that the presence of intact lipid rafts seems to be essential for initiating PON1 to stimu-late the cholesterol efflux process. Modeling the kinetics of cholesterol export from macrophages to apo-AI predicted a rapid initial efflux from the small pool, followed by a slower but substantial and sustained efflux from the separate larger pool, which contributed to most of the exported cholesterol [44].We examined the kinetics model of cholesterol efflux from mac-rophages to PON1 compared to apo-AI. Fig. 7 shows a typical choles-terol efflux experiment (0 to 16 h) from cholesterol-loaded J774 macrophages treated with cAMP. Cholesterol efflux to DMEM alone containing 1% BSA was minimal compared to apo-AI- and PON1- mediated cholesterol efflux. Like apo-AI, PON1-induced cholesterol efflux can be qualitatively split into two phases, an initial high cholesterol efflux rate phase that lasts from 0 to 3 h followed by a slower cholesterol efflux rate phase. PON1 has an effect on ABCA1 protein and gene expression To investigate the effects of PON1 on the ABCA1 level during the distinct phases of the cholesterol efflux process, ABCA1 protein and gene expression was analyzed by Western blotting and qPCR, 10000 8000 6000 4000 2000 15 10 5 1 3 16 1 3 16 0 1 3 16 1 3 16 *** *** A respectively, in J774 macrophages incubated in the presence or absence of PON1. Interestingly, a short incubation of the macrophages with PON1 increased ABCA1 protein levels after 1 and 3 h, but the effect disappeared after a longer incubation period (16 h; Fig. 8A). More importantly, the upregulation by PON1 was accompanied by an increase in ABCA1 mRNA levels (Fig. 8B). The same effect on ABCA1 protein expression was observed when apo-AI was used as a control. These results demonstrate that PON1 modulates ABCA1 expression on macrophages. Discussion Cholesterol efflux is thought to be one of the most important atheroprotective properties of HDLs. This process is mediated by the interaction of lipid-free apo-AI and HDLs with ABCA1, ABCG1, and SR-B1 membrane proteins as well as via other mechanisms, including passive diffusion. Apo-AI is a key factor in the enhancement of HDL-mediated cholesterol efflux. However, plasma HDL particles contain 0 4 8 12 16 25 20 15 10 5 0 Apo-AI [25 μg/ml] PON1 [20 U/ml: 50 μg/ml] Control [BSA: 50 μg/ml] Time [hours] Cholesterol efflux (%) Fig. 7. PON1 enhances cholesterol efflux from the ABCA1-dependent pathway in rapid-low and slow-high pools. Kinetics of cholesterol efflux to apo-AI (25 μg/ml) and PON1 (20 U/ml; 50 μg/ml) over the first 16 h in J774 [3H]cholesterol-loaded macrophages enriched in ABCA1 is shown. BSA (50 μg/ml) was used as control. Results are expressed as the means ± SEM of more than three independent experiments. 0 ** *** PON1 expression gene ABCA1 Relative 0 1 3 16 PON1 Incubation time [hours] 0 *** *** *** *** Incubation time [Hours] PON1 apo-AI Hours ABCA1 0 apo-AI PON1 Densitometry Analysis B Fig. 8. Effects of short and long pretreatment of J774 macrophages with apo-AI or PON1 on ABCA1 protein and RNA expression. Kinetics analysis of ABCA1 expression (0, 3, and 16 h) upon incubation with apo-AI (25 μg/ml) or PON1 (20 U/ml; 50 μg/ml) was car-ried out in non-ABCA1-enriched J774 cells. Protein levels and RNA expression of ABCA1 were respectively determined by (A) Western blot and (B) RT-PCR analyses. Quantitative analysis was determined using densitometry from three independent experiments. Results are expressed as means ± SEM. **pb0.01, ***pb0.001.
  • 8. H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1379 a variety of proteins with antiatherogenic potential, including PON1, that may also contribute significantly to the cholesterol efflux pro-cess. PON1 is a protein that is mainly complexed to HDLs and has been associated, in part, with the antioxidant and anti-inflammatory properties of HDLs. The goal of this study was to investigate the in-volvement of PON1 in the regulation of the cholesterol efflux process, especially the mechanism by which PON1 modulates HDL-mediated cholesterol transport. Our results showed that PON1 enhances HDL-mediated cholester-ol efflux in THP-1 macrophage-like cells and in J774 macrophages. The effect was more pronounced with ABCA1-enriched J774 macro-phages, suggesting that an interaction between PON1 and ABCA1 re-ceptors promotes cholesterol efflux. Our results showed that purified PON1 is highly effective in stimulating cholesterol efflux via the ABCA1 pathway, whereas no effect was observed with respect to SR-BI receptors. This finding is in agreement with that of Rosenblat et al., who used purified human PON1 and HDLs from PON1 knockout and PON1Tg mice [26]. Moreover, Fuhrman et al. recently reported that PON1 may protect macrophages from apoptosis by upregulating macrophage SR-BI-mediated HDL binding to the cells. However, the involvement of SR-BI in the interaction of HDLs with macrophages does not affect cellular cholesterol efflux [45]. Although apo-AI is the principal HDL-associated protein that in-teracts with membrane proteins to promote cholesterol efflux, other HDL-associated proteins may play this role. A recent study by De Beer et al. [46] showed that lipid-free serum amyloid A (SAA) acts like apo-AI by mediating sequential cholesterol efflux via ABCA1 and ABCG1 [46]. Remaley et al. showed that ABCA1-mediated lipid ef-flux is not specific to apo-AI but can also occur with other apolipopro-teins that contain multiple amphipathic helical domains [47]. A study by Xie et al. [49] showed that an apo-AI mimetic peptide (D-4F), which has no sequence homology with apo-AI but possesses the class-A amphipathic helical motif, promotes cholesterol efflux through the ABCA1 pathway [48,49]. It is thus not surprising that PON1 modulates cholesterol efflux from macrophages principally via the ABCA1 pathway. Indeed, like apo-AI and SAA, the secondary structure of PON1 contains amphipathic α-helices with approximate-ly 22 amino acids [50] that enable HDL particles to tightly bind, stabi-lize, and stimulate PON1 [51]. Our results demonstrate that PON1 enhances cholesterol efflux and induces an upregulation of ABCA1. Although we did not investi-gate the mechanism of PON1-induced upregulation of ABCA1, it has been suggested that this effect could be attributed to the capacity of PON1 to hydrolyze oxidized phospholipids to form lyso-PC, which in turn induces ABCA1 expression [26]. In accordance, Hou et al. dem-onstrated that lyso-PC increases the mRNA and protein expression of PPARγ, LXRα, and ABCA1 of macrophage foamcells in a concentration-dependent manner [52]. The upregulation of ABCA1, in this study, was confirmed by the measurement of ABCA1 protein and mRNA expression and also evidenced by the internalization of PON1 in mac-rophages. Our results from the immunofluorescence and cholesterol efflux assays showed that the internalization of PON1 in macrophages is ABCA1 dependent. A reduction in ABCA1 content or an alteration in the structure of PON1 significantly affected PON1-mediated cholesterol efflux. Sorenson et al. showed that purified human and rabbit PON1 both have a single free sulfhydryl group (Cys284) and suggested that this cysteine is part of the active site of PON1 required for its antioxidant activity [53]. Previous studies, including ours, showed that PON1 mu-tants display significantly less antioxidant activity and that blocking the free sulfhydryl groups with NEM or p-hydroxymercuribenzoate significantly reduces the antioxidant activity of PON1 [19,54]. In this study, we showed that exposure of PON1 to free radicals or its inacti-vation by heat or NEM treatment induced a net reduction in its ability to promote cholesterol efflux from THP-1 macrophage-like cells and ABCA1-enriched J774 macrophages. These results confirm that the capacity of PON1 to stimulate cholesterol efflux is dependent on its structural integrity. The mechanism by which apo-AI removes excess cholesterol and phospholipids from macrophages has been extensively investigated. Gaus et al. [28,44] studied the interaction between apo-AI and lipid raft microdomains and concluded that cholesterol efflux to apo-AI oc-curs sequentially from two membrane pools with different kinetics, that is, rapid release from a small rapid efflux pool within the first few hours followed by progressive release from a major slow efflux pool over several hours. They proposed that cholesterol exported to apo-AI from this major slow efflux pool comes from nonraft regions of the plasma membrane. However, the interaction of apo-AI with lipid rafts is required to stimulate this efflux. Our results showed that PON1-mediated cholesterol efflux also occurs via two steps: rapid cholesterol efflux during the first 3 h followed by slow choles-terol efflux thereafter. The level of cholesterol efflux mediated by PON1 was comparable to that mediated by apo-AI, at least during the rapid cholesterol efflux step. This suggested that PON1 acts via an apo-AI-like mechanism to mediate cholesterol efflux. This mecha-nistic assumption was supported by the results of ABCA1 expression measurements and experiments involving a pretreatment with cyclo-dextrin, which selectively depletes cholesterol from lipid raft do-mains (~50%) but not from nonraft domains [28]. First, the ABCA1 transporter was overexpressed within the first 3 h of incubation of PON1 with cholesterol-loaded J774 macrophages, which covers the period during which cholesterol is released from the small rapid ef-flux pool. After 16 h, ABCA1 expression was downregulated, which was probably related to the involvement of the ABCG1 transporter or SR-BI receptor. These two proteins do not interact with lipid-free apo-AI [55,56]. Second, cyclodextrin significantly affected the ability of PON1 and apo-AI to mediate cholesterol efflux from macrophages (Fig. 6B). Studies investigating the relationship between membrane plasma microdomains and apo-AI with respect to the cholesterol efflux ca-pacity of apo-AI have given rise to conflicting results. The distribution of ABCA1 between lipid raft and nonraft domains depends on the macrophage cell line and the method used to prepare the rafts. ABCA1 is not present in Triton-resistant membrane domains, whereas a significant proportion can be detected in Lubrol-resistant domains [57,58]. Mendez et al. reported that membrane raft lipids are not di-rectly available for lipid efflux by apolipoprotein-dependent mecha-nisms [58]. In contrast, cholesterol efflux from cells to HDL particles or to plasma can be partially explained by the depletion of cholesterol from membrane rafts. This suggests that apo-AI– and PON1–ABCA1- mediated cellular cholesterol efflux occurs by mechanisms distinct from those mediated by lipoprotein particles containing lipids as their major constituent. More interestingly, this study showed that PON1 plays an important role in the antiatherogenic properties of HDLs and can exert its protective function outside the lipoprotein en-vironment. This is supported by a recent study by Deakin et al. show-ing that PON1 is not a fixed component of HDLs because it can be transferred from HDLs to the external face of the plasma membrane of cells in an enzymatically active form [59]. These findings open a new window on the association between PON1 and HDLs with respect to their antiatherogenic function as well as on the mechanisms by which PON1 may exert its atheropro-tective effect, especially by regulating cholesterol efflux from macro-phages and maintaining cholesterol homeostasis. However, further studies are needed to elucidate the nature of the pathways involved in the promotion of cholesterol efflux by PON1 and to determine the activity of free, non-HDL-associated PON1. Acknowledgment This work was supported by a grant from the Canadian Institutes of Health Research (MOP-89912).
  • 9. 1380 H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 References [1] Cuchel, M.; Rader, D. J. Macrophage reverse cholesterol transport: key to the regression of atherosclerosis? Circulation 113:2548–2555; 2006. [2] Ruano, J.; Lopez-Miranda, J.; Fuentes, F.; Moreno, J. A.; Bellido, C.; Perez-Martinez, P.; Lozano, A.; Gomez, P.; Jimenez, Y.; Perez Jimenez, F. Phenolic content of virgin olive oil improves ischemic reactive hyperemia in hypercholesterolemic patients. J. Am. Coll. Cardiol. 46:1864–1868; 2005. [3] Tall, A. R.; Costet, P.; Wang, N. Regulation and mechanisms of macrophage choles-terol efflux. J. Clin. Invest. 110:899–904; 2002. [4] Yancey, P. G.; Bortnick, A. E.; Kellner-Weibel, G.; de la Llera-Moya, M.; Phillips, M. C.; Rothblat, G. H. Importance of different pathways of cellular cholesterol ef-flux. Arterioscler. Thromb. Vasc. Biol. 23:712–719; 2003. [5] Smith, J. D. Insight into ABCG1-mediated cholesterol efflux. Arterioscler. Thromb. Vasc. Biol. 26:1198–1200; 2006. [6] Favari, E.; Lee, M.; Calabresi, L.; Franceschini, G.; Zimetti, F.; Bernini, F.; Kovanen, P. T. Depletion of pre-β-high density lipoprotein by human chymase impairs ATP-binding cassette transporter A1- but not scavenger receptor class B type I-mediated lipid efflux to high density lipoprotein. J. Biol. Chem. 279:9930–9936; 2004. [7] Baldan, A.; Bojanic, D. D.; Edwards, P. A. The ABCs of sterol transport. J. Lipid Res. 50:S80–S85 (Suppl.); 2009. [8] Gu, F.; Jones, M. K.; Chen, J.; Patterson, J. C.; Catte, A.; Jerome, W. G.; Li, L.; Segrest, J. P. Structures of discoidal high density lipoproteins: a combined computational– experimental approach. J. Biol. Chem. 285:4652–4665; 2010. [9] Vaisar, T.; Pennathur, S.; Green, P. S.; Gharib, S. A.; Hoofnagle, A. N.; Cheung, M. C.; Byun, J.; Vuletic, S.; Kassim, S.; Singh, P.; Chea, H.; Knopp, R. H.; Brunzell, J.; Geary, R.; Chait, A.; Zhao, X. Q.; Elkon, K.; Marcovina, S.; Ridker, P.; Oram, J. F.; Heinecke, J. W. Shotgun proteomics implicates protease inhibition and complement activation in the antiinflammatory properties of HDL. J. Clin. Invest. 117:746–756; 2007. [10] Visioli, F.; Poli, A.; Gall, C. Antioxidant and other biological activities of phenols from olives and olive oil. Med. Res. Rev. 22:65–75; 2002. [11] Costa, L. G.; Vitalone, A.; Cole, T. B.; Furlong, C. E. Modulation of paraoxonase (PON1) activity. Biochem. Pharmacol. 69:541–550; 2005. [12] James, R. W.; Brulhart-Meynet, M. C.; Singh, A. K.; Riederer, B.; Seidler, U.; Out, R.; Van Berkel, T. J.; Deakin, S. The scavenger receptor class B, type I is a primary de-terminant of paraoxonase-1 association with high-density lipoproteins. Arterios-cler. Thromb. Vasc. Biol. 30:2121–2127; 2010. [13] Deakin, S.; Moren, X.; James, R. W. Very low density lipoproteins provide a vector for secretion of paraoxonase-1 from cells. Atherosclerosis 179:17–25; 2005. [14] Jaouad, L.; Milochevitch, C.; Khalil, A. PON1 paraoxonase activity is reduced dur-ing HDL oxidation and is an indicator of HDL antioxidant capacity. Free Radic. Res. 37:77–83; 2003. [15] Marsillach, J.; Aragones, G.; Mackness, B.; Mackness, M.; Rull, A.; Beltran-Debon, R.; Pedro-Botet, J.; Alonso-Villaverde, C.; Joven, J.; Camps, J. Decreased paraoxonase-1 activity is associated with alterations of high-density lipoprotein particles in chronic liver impairment. Lipids Health Dis. 9:46; 2010. [16] Fuhrman, B.; Volkova, N.; Aviram, M. Oxidative stress increases the expression of the CD36 scavenger receptor and the cellular uptake of oxidized low-density lipo-protein in macrophages from atherosclerotic mice: protective role of antioxidants and of paraoxonase. Atherosclerosis 161:307–316; 2002. [17] Rozenberg, O.; Shih, D. M.; Aviram, M. Paraoxonase 1 (PON1) attenuates macro-phage oxidative status: studies in PON1 transfected cells and in PON1 transgenic mice. Atherosclerosis 181:9–18; 2005. [18] Watson, A. D.; Berliner, J. A.; Hama, S. Y.; La Du, B. N.; Faull, K. F.; Fogelman, A. M.; Navab, M. Protective effect of high density lipoprotein associated paraoxonase: inhibition of the biological activity of minimally oxidized low density lipoprotein. J. Clin. Invest. 96:2882–2891; 1995. [19] Jaouad, L.; de Guise, C.; Berrougui, H.; Cloutier, M.; Isabelle, M.; Fulop, T.; Payette, H.; Khalil, A. Age-related decrease in high-density lipoproteins antioxidant activ-ity is due to an alteration in PON1's free sulfhydryl groups. Atherosclerosis 185: 191–200; 2006. [20] Tward, A.; Xia, Y. R.; Wang, X. P.; Shi, Y. S.; Park, C.; Castellani, L. W.; Lusis, A. J.; Shih, D. M. Decreased atherosclerotic lesion formation in human serum paraoxo-nase transgenic mice. Circulation 106:484–490; 2002. [21] Shih, D. M.; Gu, L.; Xia, Y. R.; Navab, M.; Li, W. F.; Hama, S.; Castellani, L. W.; Furlong, C. E.; Costa, L. G.; Fogelman, A. M.; Lusis, A. J. Mice lacking serum paraox-onase are susceptible to organophosphate toxicity and atherosclerosis. Nature 394:284–287; 1998. [22] Mackness, B.; Hunt, R.; Durrington, P. N.; Mackness, M. I. Increased immunoloca-lization of paraoxonase, clusterin, and apolipoprotein A-I in the human artery wall with the progression of atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 17: 1233–1238; 1997. [23] Tavori, H.; Khatib, S.; Aviram, M.; Vaya, J. Characterization of the PON1 active site using modeling simulation, in relation to PON1 lactonase activity. Bioorg. Med. Chem. 16:7504–7509; 2008. [24] Navab, M.; Hama, S. Y.; Hough, G. P.; Hedrick, C. C.; Sorenson, R.; La Du, B. N.; Kobashigawa, J. A.; Fonarow, G. C.; Berliner, J. A.; Laks, H.; Fogelman, A. M. High density associated enzymes: their role in vascular biology. Curr. Opin. Lipidol. 9: 449–456; 1998. [25] Rosenblat, M.; Gaidukov, L.; Khersonsky, O.; Vaya, J.; Oren, R.; Tawfik, D. S.; Aviram, M. The catalytic histidine dyad of high density lipoprotein-associated serum paraoxonase-1 (PON1) is essential for PON1-mediated inhibition of low density lipoprotein oxidation and stimulation of macrophage cholesterol efflux. J. Biol. Chem. 281:7657–7665; 2006. [26] Rosenblat, M.; Vaya, J.; Shih, D.; Aviram, M. Paraoxonase 1 (PON1) enhances HDL-mediated macrophage cholesterol efflux via the ABCA1 transporter in association with increased HDL binding to the cells: a possible role for lysophosphatidylcho-line. Atherosclerosis 179:69–77; 2005. [27] Pike, L. J.; Han, X.; Chung, K. N.; Gross, R. W. Lipid rafts are enriched in arachidonic acid and plasmenylethanolamine and their composition is independent of caveolin-1 expression: a quantitative electrospray ionization/mass spectrometric analysis. Biochemistry 41:2075–2088; 2002. [28] Gaus, K.; Kritharides, L.; Schmitz, G.; Boettcher, A.; Drobnik, W.; Langmann, T.; Quinn, C. M.; Death, A.; Dean, R. T.; Jessup, W. Apolipoprotein A-1 interaction with plasma membrane lipid rafts controls cholesterol export from macrophages. FASEB J. 18:574–576; 2004. [29] Gillotte, K. L.; Davidson, W. S.; Lund-Katz, S.; Rothblat, G. H.; Phillips, M. C. Re-moval of cellular cholesterol by pre-β-HDL involves plasma membrane microso-lubilization. J. Lipid Res. 39:1918–1928; 1998. [30] Gargalovic, P.; Dory, L. Caveolins and macrophage lipid metabolism. J. Lipid Res. 44:11–21; 2003. [31] Rees, D.; Sloane, T.; Jessup, W.; Dean, R. T.; Kritharides, L. Apolipoprotein A-I stim-ulates secretion of apolipoprotein E by foam cell macrophages. J. Biol. Chem. 274: 27925–27933; 1999. [32] Sattler, W.; Mohr, D.; Stocker, R. Rapid isolation of lipoproteins and assessment of their peroxidation by high-performance liquid chromatography postcolumn chemiluminescence. Methods Enzymol. 233:469–489; 1994. [33] Gan, K. N.; Smolen, A.; Eckerson, H. W.; La Du, B. N. Purification of human serum paraoxonase/arylesterase: evidence for one esterase catalyzing both activities. Drug Metab. Dispos. 19:100–106; 1991. [34] Eckerson,H.W.;Wyte, C.M.; La Du, B.N. The human serumparaoxonase/arylesterase polymorphism. Am. J. Hum. Genet. 35:1126–1138; 1983. [35] Hajj Hassan, H.; Blain, S.; Boucher, B.; Denis, M.; Krimbou, L.; Genest, J. Structural modification of plasma HDL by phospholipids promotes efficient ABCA1- mediated cholesterol release. J. Lipid Res. 46:1457–1465; 2005. [36] Fortin, C. F.; Larbi, A.; Lesur, O.; Douziech, N.; Fulop Jr., T. Impairment of SHP-1 down-regulation in the lipid rafts of human neutrophils under GM-CSF stimula-tion contributes to their age-related, altered functions. J. Leukocyte Biol. 79: 1061–1072; 2006. [37] Abe-Dohmae, S.; Kato, K. H.; Kumon, Y.; Hu, W.; Ishigami, H.; Iwamoto, N.; Okazaki, M.; Wu, C. A.; Tsujita, M.; Ueda, K.; Yokoyama, S. Serum amyloid A gen-erates high density lipoprotein with cellular lipid in an ABCA1- or ABCA7- dependent manner. J. Lipid Res. 47:1542–1550; 2006. [38] Hu, W.; Abe-Dohmae, S.; Tsujita, M.; Iwamoto, N.; Ogikubo, O.; Otsuka, T.; Kumon, Y.; Yokoyama, S. Biogenesis of HDL by SAA is dependent on ABCA1 in the liver in vivo. J. Lipid Res. 49:386–393; 2008. [39] Parra, S.; Alonso-Villaverde, C.; Coll, B.; Ferre, N.; Marsillach, J.; Aragones, G.; Mackness, M.; Mackness, B.; Masana, L.; Joven, J.; Camps, J. Serum paraoxonase- 1 activity and concentration are influenced by human immunodeficiency virus infection. Atherosclerosis 194:175–181; 2007. [40] Seres, I.; Paragh, G.; Deschene, E.; Fulop Jr., T.; Khalil, A. Study of factors influenc-ing the decreased HDL associated PON1 activity with aging. Exp. Gerontol. 39: 59–66; 2004. [41] Attia, N.; Fournier, N.; Vedie, B.; Cambillau, M.; Beaune, P.; Ziegler, O.; Grynberg, A.; Paul, J. L.; Guerci, B. Impact of android overweight or obesity and insulin resis-tance on basal and postprandial SR-BI and ABCA1-mediated serum cholesterol efflux capacities. Atherosclerosis 209:422–429; 2010. [42] Efrat, M.; Aviram, M. Macrophage paraoxonase 1 (PON1) binding sites. Biochem. Biophys. Res. Commun. 376:105–110; 2008. [43] Blumrich, M.; Petzinger, E. Two distinct types of SH-groups are necessary for bumetanide and bile acid uptake into isolated rat hepatocytes. Biochim. Biophys. Acta 1149:278–284; 1993. [44] Gaus, K.; Gooding, J. J.; Dean, R. T.; Kritharides, L.; Jessup, W. A kinetic model to evaluate cholesterol efflux from THP-1 macrophages to apolipoprotein A-1. Biochemistry 40:9363–9373; 2001. [45] Fuhrman, B.; Gantman, A.; Aviram, M. Paraoxonase 1 (PON1) deficiency in mice is associated with reduced expression of macrophage SR-BI and consequently the loss of HDL cytoprotection against apoptosis. Atherosclerosis 211:61–68; 2010. [46] de Beer, M. C.; Ji, A.; Jahangiri, A.; Vaughan, A. M.; de Beer, F. C.; van der Westhuyzen, D. R.; Webb, N. R. ATP binding cassette G1-dependent cholesterol efflux during inflammation. J. Lipid Res. 52:345–353; 2011. [47] Remaley, A. T.; Stonik, J. A.; Demosky, S. J.; Neufeld, E. B.; Bocharov, A. V.; Vishnyakova, T. G.; Eggerman, T. L.; Patterson, A. P.; Duverger, N. J.; Santamarina- Fojo, S.; Brewer Jr., H. B. Apolipoprotein specificity for lipid efflux by the human ABCAI transporter. Biochem. Biophys. Res. Commun. 280:818–823; 2001. [48] Moreno, J. A.; Perez-Jimenez, F.; Marin, C.; Perez-Martinez, P.; Moreno, R.; Gomez, P.; Jimenez-Gomez, Y.; Paniagua, J. A.; Lairon, D.; Lopez-Miranda, J. The apolipoprotein E gene promoter (−219G/T) polymorphism determines insulin sensitivity in response to dietary fat in healthy young adults. J. Nutr. 135:2535–2540; 2005. [49] Xie, Q.; Zhao, S. P.; Li, F. D-4F, an apolipoprotein A-I mimetic peptide, promotes cholesterol efflux from macrophages via ATP-binding cassette transporter A1. Tohoku J. Exp. Med. 220:223–228; 2010. [50] Harel, M.; Aharoni, A.; Gaidukov, L.; Brumshtein, B.; Khersonsky, O.; Meged, R.; Dvir, H.; Ravelli, R. B.; McCarthy, A.; Toker, L.; Silman, I.; Sussman, J. L.; Tawfik, D. S. Structure and evolution of the serum paraoxonase family of detoxifying and anti-atherosclerotic enzymes. Nat. Struct. Mol. Biol. 11:412–419; 2004. [51] Perez-Jimenez, F.; Lopez-Miranda, J. University of Health Sciences: a blow of hope. Med. Clin. (Barcelona) 125:477; 2005. [52] Hou, M.; Xia,M.; Zhu, H.;Wang, Q.; Li, Y.; Xiao, Y.; Zhao, T.; Tang, Z.;Ma, J.; Ling,W. Lysophosphatidylcholine promotes cholesterol efflux from mouse macrophage
  • 10. H. Berrougui et al. / Free Radical Biology & Medicine 52 (2012) 1372–1381 1381 foam cells via PPARγ–LXRα–ABCA1-dependent pathway associated with apoE. Cell Biochem. Funct. 25:33–44; 2007. [53] Sorenson, R. C.; Primo-Parmo, S. L.; Kuo, C. L.; Adkins, S.; Lockridge, O.; La Du, B. N. Reconsideration of the catalytic center and mechanism of mammalian paraoxonase/ arylesterase. Proc. Natl. Acad. Sci. U. S. A. 92:7187–7191; 1995. [54] Aviram, M.; Billecke, S.; Sorenson, R.; Bisgaier, C.; Newton, R.; Rosenblat, M.; Erogul, J.; Hsu, C.; Dunlop, C.; La Du, B. Paraoxonase active site required for protec-tion against LDL oxidation involves its free sulfhydryl group and is different from that required for its arylesterase/paraoxonase activities: selective action of human paraoxonase allozymes Q and R. Arterioscler. Thromb. Vasc. Biol. 18:1617–1624; 1998. [55] Fu, Y. Rate-limiting factors of cholesterol efflux in reverse cholesterol transport: acceptors and donors. Clin. Exp. Pharmacol. Physiol. 37:703–709; 2010. [56] Wang, N.; Lan, D.; Chen, W.; Matsuura, F.; Tall, A. R. ATP-binding cassette trans-porters G1 and G4 mediate cellular cholesterol efflux to high-density lipopro-teins. Proc. Natl. Acad. Sci. U. S. A. 101:9774–9779; 2004. [57] Drobnik, W.; Borsukova, H.; Bottcher, A.; Pfeiffer, A.; Liebisch, G.; Schutz, G. J.; Schindler, H.; Schmitz, G. Apo AI/ABCA1-dependent and HDL3-mediated lipid ef-flux from compositionally distinct cholesterol-based microdomains. Traffic 3: 268–278; 2002. [58] Mendez, A. J.; Lin, G.; Wade, D. P.; Lawn, R. M.; Oram, J. F. Membrane lipid domains distinct from cholesterol/sphingomyelin-rich rafts are involved in the ABCA1-mediated lipid secretory pathway. J. Biol. Chem. 276:3158–3166; 2001. [59] Deakin, S. P.; Bioletto, S.; Bochaton-Piallat, M. L.; James, R. W. HDL-associated paraoxonase-1 can redistribute to cell membranes and influence sensitivity to oxidative stress. Free Radic. Biol. Med. 50:102–109; 2011.