Vous êtes sur la page 1sur 8

Atherosclerosis 196 (2008) 219–226

Low phospholipid transfer protein (PLTP) is a risk factor for


peripheral atherosclerosis
Wilfried Schgoer a , Thomas Mueller b , Matti Jauhiainen d,1 ,
Andreas Wehinger a,c,1 , Roland Gander a , Ivan Tancevski a ,
Karin Salzmann a , Philipp Eller a , Andreas Ritsch a , Meinhard Haltmayer b ,
Christian Ehnholm d , Josef R. Patsch a , Bernhard Foeger a,c,∗
a Department of Internal Medicine, Medical University Innsbruck, Austria
bDepartment of Laboratory Medicine, Konventhospital Barmherzige Brueder, Linz, Austria
c Department of Internal Medicine, Landeskrankenhaus Bregenz, Austria
d Department of Molecular Medicine, National Public Health Institute, Biomedicum, Helsinki, Finland
Received 27 March 2006; received in revised form 18 April 2007; accepted 27 April 2007
Available online 5 June 2007

Abstract
Objective: Phospholipid transfer protein (PLTP) facilitates cholesterol efflux from cells, intravascular HDL remodelling and transfer of
vitamin E and endotoxin. In humans, the relationship of PLTP to atherosclerosis is unknown. However, strong coronary risk factors like
obesity, diabetes, cigarette smoking and inflammation increase circulating levels of active PLTP. The aim of the present, cross-sectional study
was to analyze the relationship of PLTP to peripheral arterial disease, a marker of generalized atherosclerosis, independently of potentially
confounding factors like obesity, diabetes and smoking.
Methods: We performed a case control study in 153 patients with symptomatic peripheral arterial disease (PAD) and 208 controls free of
vascular disease. Smokers and patients with diabetes mellitus were excluded. A lipoprotein-independent assay was used for measurement of
circulating bioactive PLTP and an ELISA utilizing a monoclonal antibody was used to analyze PLTP mass.
Results: PLTP activity was significantly decreased in patients with PAD 5.5 (4.6–6.4)(median (25th–75th percentile)) versus 5.9
(5.1–6.9) ␮mol/mL/h in controls (p = 0.001). In contrast, PLTP mass was similar in patients with PAD 8.5 ␮g/mL (7.3–9.5) and in con-
trols 8.3 ␮g/mL (6.9–9.7) (p = 0.665). Multivariate logistic regression analysis revealed that PLTP activity is independently associated with
the presence of PAD. PLTP activity was similar in patients with and without lipid-lowering drugs (p = 0.396).
Conclusion: Our results show that in non-diabetic, non-smoking subjects low rather than high PLTP activity is a marker for the presence of
peripheral arterial disease and that distribution of PLTP between high-activity and low-activity forms may be compromised in atherosclerosis.
© 2007 Elsevier Ireland Ltd. All rights reserved.

Keywords: Phospholipid transfer protein (PLTP); Peripheral arterial disease; Atherosclerosis; High density lipoproteins (HDL); Triglycerides

1. Introduction
Abbreviations: ABI, ankle brachial index; CAD, coronary arterial dis-
Phospholipid transfer protein (PLTP), a member of the
ease; CVD, cerebral vascular disease; HbA1C , glycohemoglobine A1C ; HDL,
high-density lipoprotein; hs-CRP, high-sensitivity C-reactive protein; LDL, lipid transfer/lipopolysaccharide binding protein gene family,
low-density lipoprotein; OR, odds ratio; PAD, peripheral arterial disease; plays a major role in the metabolism of HDL, vitamin E and
PC, phosphatidyl choline; PLTP, phospholipid transfer protein lipopolysaccharide [1]. PLTP facilitates cholesterol efflux
∗ Corresponding author at: Carl-Pedenzstr. 2, Department of Internal
from cells [2,3], enhances the transfer of surface remnants
Medicine, Landeskrankenhaus Bregenz, 6900 Bregenz, Austria.
from triglyceride-rich lipoproteins to HDL during lipolysis
Tel.: +43 5574 401 6401; fax: +43 5574 401 80.
E-mail address: bernhard.foeger@lkhb.at (B. Foeger). [4], modulates size and composition of HDL particles [5–7],
1 These authors contributed equally to the manuscript. promotes the generation of pre-ß HDL [8,9] and stimulates

0021-9150/$ – see front matter © 2007 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.atherosclerosis.2007.04.046
220 W. Schgoer et al. / Atherosclerosis 196 (2008) 219–226

apo B secretion [10]. PLTP transfers vitamin E from lipopro- In this study, we investigated the role of PLTP in peripheral
teins to endothelial cells [11] and helps to eliminate LPS from arterial disease, a reliable marker of generalized atheroscle-
the circulation [12]. rosis, by measuring PLTP activity and PLTP mass in PAD
In genetically modified mice, the net metabolic effect of patients without the major risk factors diabetes and smoking,
PLTP appears to promote atherogenesis [10,13,14]. How- which are known to increase PLTP activity.
ever, while complete deficiency of PLTP decreases both HDL,
apoB-secretion and atherosclerosis [10], no such metabolic
effects were observed for an approximately 50% decrease 2. Methods
of PLTP activity in heterozygotes [15]. Pronounced overex-
pression of PLTP, in turn, increases atherosclerosis [13,14]. 2.1. Study population
However, moderate overexpression of two-fold in transgen-
ics [13] or overexpression using low-dose adeno-associated We cross-sectionally studied 153 Caucasian patients with
virus does not [14]. Mechanistically, depletion of lipopro- PAD und 208 control subjects of the Linz Peripheral Arterial
teins of vitamin E by PLTP was one of the mechanisms Disease (LIPAD) study [36]. In order to eliminate confound-
proposed to account for reduced atherosclerosis in PLTP ing risk factors, we excluded smokers and patients with
deficient mice [16]. While vitamin E reduces atherosclero- diabetes mellitus. Diabetes mellitus was considered present
sis in mice [17], supplementation of humans with vitamin if a patient was treated with insulin or oral agents or had a
E to prevent atherosclerosis has been completely unsuccess- fasting glucose level ≥7.0 mmol/L. Smoking was classified
ful [17], suggesting a much more prominent role of vitamin as any amount of tobacco use within the last year. The LIPAD
E in murine than in human atherosclerosis. Taken together, study objectives, its recruitment procedures, and population
only very pronounced systemic variations of PLTP appear characteristics have been previously described [36]. PAD
to influence atherosclerosis in mice; such variations have, patients were eligible for the study if they were Caucasian
hitherto, not been reported in humans [18]. To specifically men or women and had symptomatic PAD of the lower limbs
investigate the role of macrophage PLTP in atherosclerosis, (claudication or leg pain upon exertion, rest pain, or minor
bone marrow transplantations from PLTP −/− into LDL- or major tissue loss), verified by interview, physical exam-
R −/− mice have been performed by two study groups, ination, sonography, and angiography. They were ineligible
yielding contradictory results [19,20]. Valenta et al. observed if they had non-atherosclerotic causes of PAD (cardioem-
an increase in atherosclerosis after transplantation of PLTP bolic disease, vasculitis) and the history or presence of any
−/− bone marrow suggesting an atheroprotective poten- malignancy. All control subjects were free of vascular disease
tial of macrophage PLTP [19]. In contrast, Vikstedt et al. and were characterized as follows: no clinical indication of
observed a decrease in atherosclerosis after transplanta- PAD by history and physical examination; systolic brachial
tion of PLTP −/− bone marrow suggesting an atherogenic blood pressure equal to or less than the blood pressure in
potential of macrophage PLTP [20]. ApoE −/− mice, trans- each of the right and left anterior tibial and posterior tibial
planted with PLTP −/− macrophages showed a pronounced arteries, respectively (i.e. ABI ≥ 1.0); no pathologic pattern
increase in atherosclerosis, which, however, mainly reflects of pulse waves in lower limbs by continuous-wave spectral
lower secretion of apoE by macrophages and, consequently, analysis; no manifest CAD; no manifest CVD; no previous
much higher plasma cholesterol levels [21]. Moreover, mech- vascular surgery or stenting of the internal carotid arteries;
anisms leading to atherosclerosis, such as transport of and no stenosis of the internal carotid artery 50% or greater
vitamin E by lipoproteins, may profoundly differ between on colour duplex ultrasound scans. Cases and control subjects
species. gave informed consent and the study protocol was approved
In humans, PLTP activity is increased in subjects with by the Review Committee.
a number of risk factors for atherosclerosis. PLTP activ-
ity increases with age [22], body mass index [18,23–27], 2.2. Laboratory methods
cigarette smoking [28,29] and diabetes mellitus [30,31]. The
relationship of PLTP to human atherosclerosis remains to be Venous blood was drawn from all subjects under standard-
clarified. Studying CAD patients with stable angina, unsta- ized conditions after an overnight fasting period. Creatinine,
ble angina and acute myocardial infarction, Schlitt et al. [32] fasting glucose, glycohemoglobin A1c (HbA1c ), total choles-
found in a cross-sectional study that increased plasma PLTP terol, and triglyceride concentrations were analyzed with
activity is a risk factor for CAD. In a prospective study standard assays on a COBAS Integra analyzer (Roche
in CAD patients, however, PLTP activity was not predic- Diagnostics). For determination of high-density lipoprotein
tive of future CAD events [33]. On the other hand, in a cholesterol (HDL) and low-density lipoprotein (LDL) choles-
prospective Japanese study, PLTP mass protects from CAD terol quantitative electrophoresis with enzymatic staining
[34]. Finally, a small cross-sectional study comparing nor- (Helena BioSciences) was used. Total homocysteine (tHcy),
molipidemic PAD patients with controls matched for plasma folate, and vitamin B12 assays were performed on an AxSYM
lipoproteins found a non-significant trend for increased PLTP analyzer (Abbott Diagnostics). Concentration of C-reactive
activity in cases [35]. protein (CRP) was measured with a high-sensitivity analyzer
W. Schgoer et al. / Atherosclerosis 196 (2008) 219–226 221

Table 1
Clinical data of patients with PAD and control subjects
PAD group (n = 153) Control group (n = 208) p-Valuea
Male sex (n) 92 (60%) 131 (63%) 0.586
Age (years) 74 (68–79) 72 (65–78) 0.054
BMI (kg/m2 ) 26 (24–28) 26 (24–28) 0.209
Arterial hypertension (n) 106 (69%) 88 (42%) <0.001
CAD (n) 61 (40%) 0 NA
CVD (n) 25 (16%) 0 NA
Carotid stenosis ≥50% (n) 47 (31%) 0 NA
Previous PTA/stenting (n) 39 (26%) 0 NA
Previous vascular surgery (n) 37 (24%) 0 NA
Previous amputation (n) 4 (3%) 0 NA
ABI (mmHg/mmHg) 0.62 (0.43–0.82) 1.17 (1.08–1.29) <0.001
Abbreviations: ABI, resting ankle brachial index; BMI, body mass index; CAD, coronary artery disease; CVD, cerebrovascular disease; PAD, peripheral arterial
disease; PTA, percutaneous transluminal angioplasty. Age, BMI and ABI are presented as median (25th–75th percentiles).
a Nonparametric Mann–Whitney U-test or Fisher’s exact test as appropriate.

(N High Sensitivity CRP) on a BN ProSpec analyzer (Dade age, arterial hypertension, LDL-cholesterol, triglycerides,
Behring) with polystyrene particles coated with monoclonal fasting glucose, hs-CRP and PLTP mass were included as
mouse antibodies to CRP. covariates into the final model. Covariate selection was based
Serum samples for the determination of PLTP activity on significant univariate differences and clinical relevance.
were placed on ice immediately and within 30 min they were Spearman’s coefficient of rank correlation (rs ) was used for
centrifuged at 4000 rpm for 10 min, divided into aliquots, the assessment of the relationship of quantitative and cat-
and frozen at −80 ◦ C until analysis. Corresponding patient egorial data in the study population. p-Values <0.05 were
and control samples were collected and analyzed in par- considered to indicate statistical significance. Statistical anal-
allel to minimize preanalytical error. PLTP activity was ysis was performed using SPSS version 12 (SPSS Inc.,
measured as the ability of serum to facilitate the transfer Illinois, USA).
of [3 H]dipalmitoyl-phosphatidylcholine ([3 H]PC) from PC
vesicles to HDL3 , both added in excess compared to endoge-
nous plasma lipoproteins, as previously described [37], thus 3. Results
yielding a lipoprotein-independent assay measuring circulat-
ing bioactive PLTP. In brief, after sonication of a mixture Clinical and biochemical data of 153 PAD patients and
containing 10 ␮mol of L-␣-PC (Sigma), 2 ␮Ci 3 H-PC and 208 controls are presented in Tables 1–3. Due to the study
0.1 ␮mol of butylated hydroxytoluene (Sigma), respectively, design, none of the patients or controls suffered from dia-
lipid aggregates and titanium debris were removed by cen- betes mellitus or was a smoker. Sex, age and BMI did not
trifugation. Serum samples were incubated with [3 H]PC differ between the groups, however, expectedly, the preva-
vesicles (125 nmol of PC) and HDL3 (250 ␮g of protein) in a lence of hypertension was higher among cases (69%) than
final volume of 400 ␮L of 150 mM NaCl, 10 mM Tris–HCl, controls (42%; p < 0.001). The median ankle brachial pres-
pH 7.4, at 37 ◦ C. Vesicles were separated from HDL3 by sure index in patients with PAD was 0.62 (0.43–0.82), and
addition of 300 ␮L of a solution of 500 mM NaCl, 215 mM 1.17 (1.08–1.29) in controls (p < 0.001). Fontaine stages were
MnCl2 and 445 units/mL heparin and subsequent centrifuga- IIa in 49 (32%), IIb in 78 (51%), III in 7 (5%), and IV in
tion of 10 min at 10000 × g. 500 ␮L of the supernatant was 19 patients (12%). Clinical CAD coexistent with PAD was
counted in a LS 6500 Scintillation Counter (Beckman). Sam- present in 61 patients (40%) and clinical cerebrovascular dis-
ple volumes were chosen to keep phospholipid transfer in the
linear range of the assay. PLTP mass was determined exactly Table 2
as previously described [18]. Medications of patients with PAD and control subjects
PAD (n = 153) Controls (n = 208)
2.3. Statistical analysis Antihypertensive monotherapy (n) 38 (25%) 54 (26%)
Combination therapy (n) 45 (29%) 31 (15%)
Quantitative values are expressed as median (25th–75th 3-fold Combination therapy (n) 23 (15%) 3 (1%)
percentile) and respective differences between cases and con- Aspirin therapy (n) 67 (44%) 23 (11%)
Oral Anticoagulation (n) 24 (16%) 0
trols were determined by the nonparametric Mann–Whitney Folate supplement (n) 2 (1%) 0
U-test. Fisher’s exact test was used to compare dichotomous B-Vitamin supplement (n) 6 (4%) 0
variables. The a priori hypothesis of a relationship between Statin therapy (n) 32 (21%) 0
PLTP activity and PAD was tested using multivariate logistic Fibrate therapy (n) 4 (3%) 0
regression with case control status as dependent variable; sex, Abbreviation: PAD, peripheral arterial disease
222 W. Schgoer et al. / Atherosclerosis 196 (2008) 219–226

Table 3
Biochemical data of patients with PAD and control subjects
PAD group (n = 153) Control group (n = 208) p-Valuea
Total-cholesterol (mmol/L) 5.8 (5.1–6.7) 5.7 (4.9–6.3) 0.070
LDL-cholesterol (mmol/L) 3.8 (3.2-4.6) 3.7 (3.0–4.1) 0.014b
HDL-cholesterol (mmol/L) 1.3 (1.1–1.7) 1.4 (1.1–1.7) 0.423
Triglycerides (mmol/L) 1.7 (1.3–2.1) 1.2 (0.9–1.7) 0.016b
Fasting glucose (mmol/L) 5.3 (4.9–5.8) 5.1 (4.7–5.5) 0.003
HbA1c (%) 5.9 (5.6–6.2) 5.7 (5.4–6.0) <0.001
hs-CRP (mg/L) 3.9 (1.7–8.9) 2.2 (0.9–5.5) <0.001
Creatinine (␮mol/L) 88 (80–97) 80 (71–88) <0.001
Vitamin B12 (pg/mL) 337 (240–476) 328 (238–440) 0.470
Folate (ng/mL) 7.4 (5.1–9.4) 7.1 (5.3–9.4) 0.914
tHcy (␮mol/L) 17.8 (14.1–23.0) 14.8 (13.0–18.0) <0.001
Abbreviations: HDL, high density lipoprotein; hs-CRP, high-sensitivity C-reactive protein; LDL, low density lipoprotein; PAD, peripheral arterial disease; tHcy,
total homocysteine. Biochemical data are presented as median (25th–75th percentiles).
a Nonparametric Mann-Whitney U-test.
b p-Values not significant after Bonferroni correction for multiple comparisons.

Table 4
PLTP activity and PLTP mass in cases and controls
PLTP activity (␮mol/mL/h) PLTP mass (␮g/mL)
PAD patients Controls p-Valuea PAD patients Controls p-Valuea
Entire study sample
All PAD patients (n = 153) vs. controls (n = 208) 5.5 (4.6–6.4) 5.9 (5.1–6.9) 0.001 8.5 (7.3–9.5) 8.3 (6.9–9.7) 0.665
Exploratory subgroup analyses
PAD patients with CAD (n = 61) vs. controls (n = 208) 5.4 (4.5–6.2) 5.9 (5.1–6.9) 0.010 8.7 (7.5–9.7) 8.3 (6.9–9.7) 0.272
PAD patients without CAD (n = 92) vs. controls (n = 208) 5.5 (4.7–6.4) 5.9 (5.1–6.9) 0.007 8.3 (7.3–9.3) 8.3 (6.9–9.7) 0.815
PAD patients with LLM (n = 36) vs. controls (n = 208) 5.4 (4.5–6.0) 5.9 (5.1–6.9) 0.006 8.1 (7.0–8.9) 8.3 (6.9–9.7) 0.292
PAD patients without LLM (n = 117) vs. controls (n = 208) 5.5 (4.7–6.5) 5.9 (5.1–6.9) 0.008 8.7 (7.6–9.6) 8.3 (6.9–9.7) 0.304
Abbreviations: PAD, peripheral arterial disease; CAD, coronary artery disease; LLM, lipid-lowering medication; PLTP, phospholipid transfer protein. PLTP
activity and PLTP mass values are presented as median (25th–75th percentiles).
a p-Values for all groups of patients vs. controls, respectively, calculated with nonparametric Mann–Whitney U-test.

ease and PAD in 25 patients (16%). Furthermore, 47 (31%) age of 72 years, 44 control subjects (21%) exhibited carotid
of the patients with PAD exhibited internal carotid stenosis plaques as a sign of early atherosclerosis. Table 2 shows
50% or greater. Thirty-nine patients with PAD had under- that, expectedly, the PAD group received more intensive anti-
gone percutaneous transluminal angioplasty (PTA) with or hypertensive, antiaggregatory and lipid-lowering treatment.
without stenting of leg arteries, 37 had undergone vascular Table 3 shows that LDL-cholesterol and triglycerides were
surgery, and 4 patients had undergone minor amputations. higher and HDL-cholesterol tended to be lower in cases. The
None of the controls suffered from manifest CAD or CVD, relatively weak relationship of lipoproteins to PAD proba-
had an ABI < 1 and an internal carotid stenosis of 50% or bly reflects the difference in lipid-lowering treatment (24%
greater. However, as expected in probands with a median in PAD, none in controls). In addition, median values of

Table 5
Results of logistic regression analysis of PAD risk factors (153 patients with PAD vs. 208 control subjects)
Change in risk factor Multivariate odds ratios of PADa p-Value
Male sex (vs. female) 1.21 (0.73–2.01) 0.459
Age (+5 years) 1.11 (0.98–1.26) 0.113
Arterial hypertension (vs. not) 2.69 (1.67–4.34) <0.001
LDL-cholesterol (+1 mmol/L) 1.61 (1.24–2.09) <0.001
Triglycerides (+1 mmol/L) 1.11 (0.83–1.49) 0.482
Fasting glucose (+1 mmol/L) 1.71 (1.17–2.49) 0.005
hs-CRP (+1 mg/L) 1.01 (1.00–1.02) 0.012
PLTP activity (+0.5 ␮mol/mL/h) 0.83 (0.76–0.92) <0.001
PLTP mass (+0.5 ␮g/mL) 1.04 (0.97–1.12) 0.233
Statistical model with an incremental approach for continuous variables.
Abbreviations: hs-CRP, high-sensitivity C-reactive protein; LDL, low density lipoprotein; PAD, peripheral arterial disease; PLTP, phospholipid transfer protein.
a Data are expressed as odds ratio (95% confidence interval); multivariate odds ratios were calculated by logistic regression analysis without variable selection

technique (all variables were included simultaneously into the model).


W. Schgoer et al. / Atherosclerosis 196 (2008) 219–226 223

fasting glucose, HbA1c , hs-CRP, creatinine and tHcy were inversely related to Fontaine stages (rs = −0.160; p = 0.048).
significantly higher in patients than in controls. When compared to controls, PLTP activity in PAD was con-
To examine the relationship between plasma PLTP activ- sistently decreased irrespective of the presence or absence
ity and PAD, we measured PLTP activity and PLTP mass of lipid-lowering treatment or concomitant clinical CAD
in all 361 subjects. Fig. 1 shows the distribution of the (Table 4).
PLTP activity and PLTP mass levels in patients with PAD In order to exclude interaction of confounding risk fac-
and control subjects. Plasma PLTP activity was significantly tors for PAD, we performed multivariate analysis (Table 5).
lower in patients with symptomatic PAD 5.5 ␮mol/mL/h Including risk factors such as sex, age, hypertension, LDL-
(4.6–6.4) when compared with controls 5.9 ␮mol/mL/h cholesterol, triglycerides, fasting glucose and hs-CRP, PLTP
(5.1–6.9; p = 0.001). In contrast, PLTP mass was similar activity was shown to be an independent and significant pre-
in patients with PAD 8.5 ␮g/mL (7.3–9.5) and in controls dictor of case-control status with an OR of 0.83 (95% CI,
8.3 ␮g/mL (6.9–9.7) (p = 0.665). Moreover, PLTP activity 0.76–0.92; p < 0.001), for an increment of 0.5 ␮mol/mL/h.
was positively associated with ABI (rs = 0.182; p < 0.001) and In this analysis, arterial hypertension (OR, 2.69; 95% CI,
1.67–4.34; p < 0.001), fasting glucose (OR, 1.71; 95% CI,
1.17–2.49; p = 0.005), LDL cholesterol (OR, 1.61; 95%
CI, 1.24–2.09; p < 0.001) and hs-CRP (OR, 1.01; 95%
CI, 1.00–1.02; p = 0.012) were also independently related
to symptomatic PAD. Stepwise logistic regression models
revealed the same predictors, with similar ORs compared
with the models without variable selection technique (data
not shown).
To test the relationship of the main biochemical mark-
ers with PLTP activity we used rank correlation analysis.
In cases, PLTP activity was significantly related to plasma
glucose (r = 0.191; p = 0.018) and tended to correlate with
total cholesterol (r = 0.14, p = 0.08) and plasma triglyc-
erides (r = 0.129, p = 0.11). In controls, however, the above
relationships were not observed. PLTP activity was sig-
nificantly correlated with PLTP mass in the entire study
sample (r = 0.24, p < 0.001; Pearson r = 0.268, p < 0.001)
(Fig. 2), reflecting positive relationships in both cases
(r = 0.213, p = 0.008; Pearson r = 0.299, p < 0.001) and in
controls (r = 0.262, p < 0.001; Pearson r = 0.259, p < 0.001).
PLTP mass was inversely correlated to TG in both cases
(r = −0.208, p = 0.01) and controls (r = −0.159, p = 0.022).

Fig. 1. (a) Box-and-whisker plots of PLTP activity and PLTP mass for
patients with PAD (n = 153) and control subjects (n = 208). (b) In box-and-
whisker plots, central box represents values from lower to upper quartile,
middle line represents median; whiskers extend from minimum to maximum
value, excluding outside (>1.5 box-lengths from box) and far out values (>3
box-length from box) which are displayed as separate points. Median values
of plasma PLTP activity were significantly lower in PAD patients than in
control subjects (5.5 vs. 5.9 ␮mol/mL/h; p = 0.001); median PLTP mass was Fig. 2. Scatter plots for PLTP activity vs. PLTP mass in the entire study
not significantly different in PAD patients and in control subjects (8.5 vs. sample (n = 361). Spearman’s coefficient of rank correlation (rs ), 0.240
8.3 ␮g/mL; p = 0.665). (p < 0.001); Pearson r = 0.268 (p < 0.001).
224 W. Schgoer et al. / Atherosclerosis 196 (2008) 219–226

4. Discussion support previous observations, i.e. increased mass in older


subjects [18], correlation with PLTP activity when using
The relationship of lipid transfer proteins involved in exogenous substrate [18], and inverse correlation with VLDL
reverse cholesterol transport, like f.i. CETP, to atherosclerosis [18].
has turned out to be complex. Furthermore, results in exper- Our study clearly has a number of limitations. First, the
imental animals have been conflicting and have not always relationship of conventional risk factors to PAD is certainly
been borne out in the pathogenesis of human atherosclerosis not representative of PAD patients in general. In part, this
[38,39]. results from the study design, which for the reasons given
In the present paper, we undertook to analyze the role above chose to eliminate two of the major risk factors for
of PLTP, a functionally and genetically related LTP also PAD, i.e. smoking and diabetes. The weak relationship of
involved in RCT [1], in human atherosclerosis. PLTP is LDL to PAD in our cohort can be explained by the fact that
crucial for cellular cholesterol efflux [2,3], HDL matura- 24% of PAD patients were on lipid lowering drugs, predom-
tion, lipolysis of TGRLP [4–9], transfer of vitamin E and inantly statins.
lipopolysaccharide between lipoproteins and cells and VLDL Second, as any cross-sectional study, ours cannot estab-
secretion [10–12]. Which, if any, of these diverse pro- lish causality. Thus, while we fully believe that our findings
cesses are relevant for human atherosclerosis is currently reflect a protective role of PLTP in atherogenesis, we cannot
unknown. exclude the alternative explanation, i.e. that disease status
As a first step, we felt it would be worthwhile to exam- itself reduces PLTP activity. However, previous studies offer
ine whether PLTP activity in serum, a measure of circulating no support whatever to this contention. One possible expla-
enzymatically active PLTP [18], is related to clinically man- nation would be that lipid lowering therapy is responsible for
ifest atherosclerosis at all. Three previous papers on this decreased PLTP. Indeed, a recent study observed that PLTP
subject have reported differing results. Schlitt et al. [32] activity, which is abnormally increased in patients with type
have observed increased PLTP activity in CAD patients in 2 diabetes, decreases after treatment with low and high-dose
a cross-sectional design. In a prospective design, however, atorvastatin by 9% and 13%, respectively [42]. However,
PLTP activity failed to predict CAD events in patients with two previous studies clearly showed no effect of simvastatin
CAD at baseline [33]. Yatsuya et al. [34] have found PLTP treatment on PLTP activity in non-diabetics [43,44]. In our
mass to be decreased in Japanese CAD patients (PLTP activ- study, PLTP activity in PAD patients treated with statins did
ity was not assessed in this paper). A significant problem not differ from PAD patients without lipid lowering drugs
in establishing PLTP as an independent risk factor (or pro- and a significant decrease in PLTP compared to controls
tective factor) for atherosclerosis arises because at least two was found also in PAD patients not using statins. Thus, in
strong, established risk factors for atherosclerosis profoundly line with previous reports [32,43,44], lipid lowering therapy
affect active PLTP. First, smoking increases PLTP activity can be formally excluded as an explanation for low PLTP
in plasma [28,29]. Second, obesity [23–26], insulin resis- in PAD. Indeed, the relatively large percentage of our PAD
tance [24] and/or diabetes [30,31] have also repeatedly been group not treated with statins by their referring physicians,
documented to increase PLTP activity. As both smoking and although surprising to us, can be considered a strength of the
the insulin resistance syndrome promote atherosclerosis by present study. In any case, prospective studies on the rela-
a wide array of mechanisms independent of PLTP, inclusion tionship of PLTP to atherosclerosis are needed to establish
of smokers and people with insulin resistance has the poten- causality.
tial to spuriously associate PLTP with atherosclerosis in a Third, our study was not designed to study the
cross-sectional study. mechanism(s) by which active PLTP may protect from
Therefore, we chose to compare a cohort of non-smoking, atherosclerosis. Interestingly, we documented similar total
non-diabetic patients with clinically manifest PAD with non- PLTP mass in cases and controls. Human plasma contains two
smoking non-diabetic control subjects who were free not only forms of PLTP, a high-active form and a low-active form cir-
of clinical but also of relevant subclinical atherosclerosis. In culating in macromolecular complexes of differing size and
the present study, which is the first to analyze both PLTP composition [18,45,46]. Therefore, one potential explanation
activity and PLTP mass in relation to atherosclerosis, we of our findings is a decrease in plasma factors favouring dis-
report three major novel findings. First, PLTP activity was tribution of PLTP to high active as opposed to low-active
significantly decreased in our PAD patients by 10%, whereas complexes in PAD. Indeed, factors involved in modulation
PLTP mass was similar in cases and controls. Second, PLTP of PLTP distribution appear to be apolipoprotein E [47–49]
activity decreased progressively with increasing PAD stage and apolipoprotein A–I [47–49].
(from Fontaine stage IIa to IV). Third, in a multivariate HDL-cholesterol and LDL-cholesterol, as in previous
analysis PLTP activity remained independently associated studies in healthy subjects, were not significantly related to
as an atheroprotective factor with PAD. Interestingly, as has PLTP [18], thus offering no indication for a possible mech-
been reported by previous investigators [18,31,32,40,41], we anism. However, much more detailed structural analyses of
observed rather weak relationships of PLTP activity to plasma f.i. HDL-subfractions and analyses of HDL-kinetics will be
lipoprotein levels and glucose levels. Our data on PLTP mass necessary in order to rule out dysfunctional HDL as the
W. Schgoer et al. / Atherosclerosis 196 (2008) 219–226 225

potential pathophysiological defect mediating increased risk Acknowledgments


of atherosclerosis due to subnormal PLTP activity. Alterna-
tively, subnormal PLTP activity may promote atherosclerosis Miss Sari Nuutinen is thanked for expert technical assis-
directly in the vessel wall, f.i. by compromising choles- tance in PLTP mass assays. This study was supported by grant
terol efflux from macrophage foam cells [19–21,50] without P16121-B07 of the Austrian Fonds zur Förderung der wis-
appreciably altering HDL-subfraction distribution or HDL- senschaftlichen Forschung (FWF), by grant Nr. 9890 of the
kinetics in plasma. Clearly, the above analyses were beyond Jubiläumsfonds der Österreichischen Nationalbank (ÖNB),
the scope of the present investigation. and by grant Nr. 48 of the Fonds zur Förderung der Forschung
Fourth, in the present study we analyzed patients with an den Universitätskliniken Innsbruck (MFF)(all to B.F.).
PAD, and, thus, cannot extrapolate our findings to other
vascular territories. Indeed, substantial differences in the rel-
ative importance of risk factors for cervical, coronary and References
peripheral atherosclerosis have been well documented. In
[1] Huuskonen J, Olkkonen VM, Jauhiainen M, Ehnholm C. The impact of
fact, a large previous study by Schlitt et al. [32] reported phospholipid transfer protein (PLTP) on HDL metabolism. Atheroscle-
an increase in PLTP activity in CAD patients compared to rosis 2001;155:269–81.
healthy controls. However, these data have been somewhat [2] Wolfbauer G, Albers JJ, Oram JF. Phospholipid transfer pro-
controversial as another study group has reported low PLTP tein enhances removal of cellular cholesterol and phospholipids
in CAD patients [34] and in a prospective study by Schlitt et by high-density lipoprotein apolipoproteins. Biochim Biophys Acta
1999;1439:65–76.
al. PLTP failed to predict CAD events [33]. In our subgroup of [3] Oram JF, Wolfbauer G, Vaughan AM, Tang C, Albers JJ. Phospho-
patients with both PAD and CAD we found a similar decrease lipid transfer protein interacts with and stabilizes ATP-binding cassette
in PLTP as in our PAD patients without CAD. An obvious dif- transporter A1 and enhances cholesterol efflux from cells. J Biol Chem
ference between our study and the study by Schlitt et al. [32] is 2003;278:52379–85.
that they studied a substantial number of patients immediately [4] Tall AR, Abreu E, Shuman J. Separation of a plasma phospholipid
transfer protein from cholesterol ester/phospholipid exchange protein.
after an acute coronary syndrome (25%). It is well known that J Biol Chem 1983;258:2174–80.
PLTP behaves as an acute phase protein, which, accordingly, [5] Rye KA, Barter PJ. Changes in the size and density of human high-
would be expected to be increased after an acute coronary density lipoproteins promoted by a plasma-conversion factor. Biochim
syndrome [51]. In contrast, our study evaluates a chronic, Biophys Acta 1986;875:429–38.
stable phase of atherosclerosis. Another obvious difference [6] Jauhiainen M, Metso J, Pahlman R, Blomqvist S, van Tol A, Ehnholm
C. Human plasma phospholipid transfer protein causes high density
between our study and the study by Schlitt et al. [32] is the lipoprotein conversion. J Biol Chem 1993;268:4032–6.
exclusion of smokers (25.8% in cases versus 12.6% in con- [7] Tu AY, Nishida HI, Nishida T. High density lipoprotein conversion
trols) and diabetics (32.8% in cases versus 9.9% in controls) mediated by human plasma phospholipid transfer protein. J Biol Chem
[32]. Therefore, we think that differences in study design 1993;268:23098–105.
and, potentially, in analytical methodology are more likely to [8] von Eckardstein A, Jauhiainen M, Huang Y, et al. Phospholipid
transfer protein mediated conversion of high density lipoproteins
account for the apparent discrepancy than the atherosclerotic generates pre beta 1-HDL. Biochim Biophys Acta 1996;1301:255–
territory involved. 62.
Fifth, one of the major aims of the present study was to [9] Foger B, Santamarina-Fojo S, Shamburek RD, et al. Plasma phos-
study the relationship of PLTP to atherosclerosis, as far as pholipid transfer protein. Adenovirus-mediated overexpression in mice
possible, independently of the insulin resistance syndrome. leads to decreased plasma high density lipoprotein (HDL) and enhanced
hepatic uptake of phospholipids and cholesteryl esters from HDL. J Biol
Thus, whether our findings are pertinent to people with pro- Chem 1997;272:27393–400.
nounced obesity or diabetes is unclear. [10] Jiang XC, Qin S, Qiao C, et al. Apolipoprotein B secretion and
In summary, PLTP activity in plasma is decreased atherosclerosis are decreased in mice with phospholipid-transfer pro-
in elderly patients with symptomatic PAD, compared to tein deficiency. Nat Med 2001;7:847–52.
controls free of clinical and relevant subclinical arterioscle- [11] Desrumaux C, Deckert V, Athias A, et al. Plasma phospholipid
transfer protein prevents vascular endothelium dysfunction by deliv-
rotic disease. In contrast, total PLTP mass is similar in ering alpha-tocopherol to endothelial cells. FASEB J 1999;13:883–
patients with PAD and controls indicating disturbed dis- 92.
tribution of PLTP between high-activity and low-activity [12] Levels JH, Marquart JA, Abraham PR, et al. Lipopolysaccha-
macromolecular complexes. In multivariate logistic regres- ride is transferred from high-density to low-density lipoproteins by
sion, PLTP remains strongly and independently associated lipopolysaccharide-binding protein and phospholipid transfer protein.
Infect Immun 2005;73:2321–6.
with PAD. Our data strongly support an atheroprotective [13] van Haperen R, van Tol A, van Gent T, et al. Increased risk of atheroscle-
role of PLTP in the pathogenesis of peripheral vascular rosis by elevated plasma levels of phospholipid transfer protein. J Biol
disease. Chem 2002;277:48938–43.
[14] Yang XP, Yan D, Qiao C, et al. Increased atherosclerotic lesions in
apoE mice with plasma phospholipid transfer protein overexpression.
Arterioscler Thromb Vasc Biol 2003;23:1601–7.
Financial disclosure [15] Jiang XC, Bruce C, Mar J, et al. Targeted mutation of plasma phospho-
lipid transfer protein gene markedly reduces high-density lipoprotein
The authors have no financial interest in this article. levels. J Clin Invest 1999;103:907–14.
226 W. Schgoer et al. / Atherosclerosis 196 (2008) 219–226

[16] Jiang XC, Tall AR, Qin S, et al. Phospholipid transfer protein deficiency [34] Yatsuya H, Tamakoshi K, Hattori H, et al. Serum phospholipid transfer
protects circulating lipoproteins from oxidation due to the enhanced protein mass as a possible protective factor for coronary heart diseases.
accumulation of vitamin E. J Biol Chem 2002;277:31850–6. Circ J 2004;68:11–6.
[17] Navab M, Ananthramaiah GM, Reddy ST, et al. The oxidation hypoth- [35] Ruhling K, Lang A, Richard F, et al. Net mass transfer of plasma
esis of atherogenesis: the role of oxidized phospholipids and HDL. J cholesteryl esters and lipid transfer proteins in normolipidemic patients
Lipid Res 2004;45:993–1007. with peripheral vascular disease. Metabolism 1999;48:1361–6.
[18] Janis MT, Siggins S, Tahvanainen E, et al. Active and low-active forms [36] Mueller T, Marschon R, Dieplinger B, et al. Factor V Lei-
of serum phospholipid transfer protein in a normal Finnish population den, prothrombin G20210A, and methylenetetrahydrofolate reductase
sample. J Lipid Res 2004;45:2303–9. C677T mutations are not associated with chronic limb ischemia:
[19] Valenta DT, Ogier N, Bradshaw G, et al. Atheroprotective potential the Linz Peripheral Arterial Disease (LIPAD) study. J Vasc Surg
of macrophage-derived phospholipid transfer protein in low-density 2005;41:808–15.
lipoprotein receptor-deficient mice is overcome by apolipoprotein AI [37] Damen J, Regts J, Scherphof G. Transfer of [14 C]phosphatidylcholine
overexpression. Arterioscler Thromb Vasc Biol 2006;26:1572–8. between liposomes and human plasma high density lipoprotein. Partial
[20] Vikstedt R, Ye D, Metso J, et al. Macrophage phospholipid transfer purification of a transfer-stimulating plasma factor using a rapid transfer
protein contributes significantly to total plasma phospholipid transfer assay. Biochim Biophys Acta 1982;712:444–52.
activity and its deficiency leads to diminished atherosclerotic lesion [38] Ritsch A, Patsch JR. Cholesteryl ester transfer protein: gathering
development. Arterioscler Thromb Vasc Biol 2007;27:578–86. momentum as a genetic marker and as drug target. Curr Opin Lipidol
[21] Liu R, Hojjati MR, Devlin CM, Hansen IH, Jiang XC. Macrophage 2003;14:173–9.
phospholipid transfer protein deficiency and ApoE secretion: impact [39] Foger B, Chase M, Amar MJ, et al. Cholesteryl ester transfer pro-
on mouse plasma cholesterol levels and atherosclerosis. Arterioscler tein corrects dysfunctional high density lipoproteins and reduces aortic
Thromb Vasc Biol 2007;27:190–6. atherosclerosis in lecithin cholesterol acyltransferase transgenic mice.
[22] Tahvanainen E, Jauhiainen M, Funke H, et al. Serum phospholipid J Biol Chem 1999;274:36912–20.
transfer protein activity and genetic variation of the PLTP gene. [40] Oka T, Yamashita S, Kujiraoka T, et al. Distribution of human plasma
Atherosclerosis 1999;146:107–15. PLTP mass and activity in hypo- and hyperalphalipoproteinemia. J
[23] Dullaart RP, Sluiter WJ, Dikkeschei LD, Hoogenberg K, Van Tol Lipid Res 2002;43:1236–43.
A. Effect of adiposity on plasma lipid transfer protein activities: a [41] Cheung MC, Knopp RH, Retzlaff B, et al. Association of plasma
possible link between insulin resistance and high density lipoprotein phospholipid transfer protein activity with IDL and buoyant LDL:
metabolism. Eur J Clin Invest 1994;24:188–94. impact of gender and adiposity. Biochim Biophys Acta 2002;1587:
[24] Riemens SC, van Tol A, Sluiter WJ, Dullaart RP. Plasma phospholipid 53–9.
transfer protein activity is related to insulin resistance: impaired acute [42] Geesje M, Dallinga-Thie AVT. Apolipoprotein E is a determinant of
lowering by insulin in obese Type II diabetic patients. Diabetologia PLTP activity in plasma from Type 2 diabetes mellitus patients: effects
1998;41:929–34. of atorvastatin treatment. Diabetes Suppl 2005;54:A1.
[25] Kaser S, Laimer M, Sandhofer A, et al. Effects of weight loss on [43] Lagrost L, Athias A, Lemort N, et al. Plasma lipoprotein distribution
PLTP activity and HDL particle size. Int J Obes Relat Metab Disord and lipid transfer activities in patients with type IIb hyperlipidemia
2004;28:1280–2. treated with simvastatin. Atherosclerosis 1999;143:415–25.
[26] Murdoch SJ, Kahn SE, Albers JJ, Brunzell JD, Purnell JQ. PLTP activ- [44] Cheung MC, Wolfbauer G, Kennedy H, Brown BG, Albers JJ. Plasma
ity decreases with weight loss: changes in PLTP are associated with phospholipid transfer protein activity in patients with low HDL and
changes in subcutaneous fat and FFA but not IAF or insulin sensitivity. cardiovascular disease treated with simvastatin and niacin. Biochim
J Lipid Res 2003;44:1705–12. Biophys Acta 2001;1537:117–24.
[27] Huuskonen J, Ekstrom M, Tahvanainen E, et al. Quantification of [45] Karkkainen M, Oka T, Olkkonen VM, et al. Isolation and par-
human plasma phospholipid transfer protein (PLTP): relationship tial characterization of the inactive and active forms of human
between PLTP mass and phospholipid transfer activity. Atherosclerosis plasma phospholipid transfer protein (PLTP). J Biol Chem 2002;277:
2000;151:451–61. 15413–8.
[28] Dullaart RP, Hoogenberg K, Dikkeschei BD, van Tol A. Higher plasma [46] Oka T, Kujiraoka T, Ito M, et al. Distribution of phospholipid transfer
lipid transfer protein activities and unfavorable lipoprotein changes in protein in human plasma: presence of two forms of phospholipid trans-
cigarette-smoking men. Arterioscler Thromb 1994;14:1581–5. fer protein, one catalytically active and the other inactive. J Lipid Res
[29] Cheung MC, Brown BG, Marino Larsen EK, Frutkin AD, O’Brien 2000;41:1651–7.
KD, Albers JJ. Phospholipid transfer protein activity is associated with [47] Cheung MC, Albers JJ. Active plasma phospholipid transfer protein is
inflammatory markers in patients with cardiovascular disease. Biochim associated with apoA-I- but not apoE-containing lipoproteins. J Lipid
Biophys Acta 2006;1762:131–7. Res 2006;47:1315–21.
[30] Jonkers IJ, Smelt AH, Hattori H, et al. Decreased PLTP mass but ele- [48] Janis MT, Metso J, Lankinen H, et al. Apolipoprotein E activates the
vated PLTP activity linked to insulin resistance in HTG: effects of low-activity form of human phospholipid transfer protein. Biochem
bezafibrate therapy. J Lipid Res 2003;44:1462–9. Biophys Res Commun 2005;331:333–40.
[31] Colhoun HM, Scheek LM, Rubens MB, et al. Lipid transfer protein [49] Tan KC, Shiu SW, Wong Y, Wong WK, Tam S. Plasma apolipoprotein
activities in type 1 diabetic patients without renal failure and non- E concentration is an important determinant of phospholipid transfer
diabetic control subjects and their association with coronary artery protein activity in type 2 diabetes mellitus. Diabetes Metab Res Rev
calcification. Diabetes 2001;50:652–9. 2006;22:307–12.
[32] Schlitt A, Bickel C, Thumma P, et al. High plasma phospholipid transfer [50] Lee-Rueckert M, Vikstedt R, Metso J, et al. Absence of endoge-
protein levels as a risk factor for coronary artery disease. Arterioscler nous phospholipid transfer protein impairs ABCA1-dependent efflux of
Thromb Vasc Biol 2003;23:1857–62. cholesterol from macrophage foam cells. J Lipid Res 2006;47:1725–32.
[33] Schlitt A, Blankenberg S, Rupprecht HJ, et al. Prognostic value of clas- [51] Barlage S, Frohlich D, Bottcher A, et al. ApoE-containing high den-
sical and modern markers of lipoprotein-metabolism among elderly sity lipoproteins and phospholipid transfer protein activity increase
with CAD, only apoA-I is an independent predictor for event free in patients with a systemic inflammatory response. J Lipid Res
survival. Eur Heart J 2006;27(Abstract Suppl.):850. 2001;42:281–90.

Vous aimerez peut-être aussi