P-selectin is an adhesion molecule expressed on activated platelets and endothelium. It is known to play an important role in atherosclerosis. P-selectin also circulates in plasma in a soluble form (sP-selectin), which induces procoagulant microparticle formation. We investigated the role of platelet versus endothelial P-selectin in generating sP-selectin and in the formation of atherosclerotic lesions in the apolipoprotein E (apoE)–deficient mouse model. For this we transplanted apoE−/−P-selectin−/− and apoE−/−P-selectin+/+ lethally irradiated mice with bone marrow of either genotype. Seven months after transplantation, we determined from the chimeric animals that the majority of circulating sP-selectin was of endothelial origin. Thus, in atherosclerosis, the procoagulant sP-selectin reflects endothelial rather than platelet activation. We found that endothelial P-selectin was crucial for the promotion of atherosclerotic lesion growth because in its absence only relatively small lesions developed. However, platelet P-selectin also contributed to the lesion development because lesions in wild-type recipients receiving transplants with wild-type platelets were 30% larger than those receiving P-selectin-deficient platelets (P < .008) and were more frequently calcified (80% versus 44%). In comparison with P-selectin wild-type animals, absence of either endothelial or platelet P-selectin inhibited migration of smooth muscle cells into the lesion. Thus, in addition to endothelium, platelets and their P-selectin also actively promote advanced atherosclerotic lesion development.

Atherosclerosis and other inflammatory diseases are characterized by the infiltration of leukocytes into foci of inflammation. In atherosclerosis, the cells recruited are primarily monocytes.1 P-selectin, an adhesion receptor found in storage granules of platelets and endothelial cells,2mediates rolling of monocytes on activated endothelium,3the first step in the cell adhesion cascade.4,5 This interaction allows chemokines, such as monocyte chemoattractant protein 1 (MCP-1) or RANTES, presented on the surface of endothelial cells, to activate the monocytes, which then can bind to other endothelial cell adhesion molecules such as intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1). The monocytes thereby attach firmly to the endothelium and finally transmigrate into the arterial intima. Targeted disruption of the P-selectin gene in the mouse results in marked inhibition of leukocyte rolling along a stimulated vessel wall and in delayed recruitment of monocytes into sites of inflammation.6,7 Furthermore, absence of P-selectin in atherosclerosis-prone apolipoprotein E (apoE)-deficient mice delays and reduces atherosclerotic lesion formation.8,9 However, P-selectin is not only expressed on activated endothelial cells but also on activated platelets and it mediates rosetting of the platelets with monocytes and neutrophils.10 This interaction could contribute to monocyte recruitment11 and could bring platelets, with all their biologic activities, into lesions. Quite some time ago, Ross and Harker proposed the “response to injury” model of atherosclerosis and suggested platelet involvement in this process. However, a direct contribution of platelets to atherosclerotic lesion growth or maturation has not been demonstrated experimentally except when arteriosclerosis was induced by mechanical12 or chemical damage to the endothelium.13 

P-selectin, released from the cell surface, circulates as a soluble molecule in the plasma.14 It was recently shown that soluble P-selectin (sP-selectin) can exert procoagulant activity15 and it might therefore play an important role in thrombosis and acute coronary events. sP-selectin is often used as a marker of platelet activation,16 although its origin has not been established with certainty.

We performed bone marrow transplantation experiments using apoE-deficient and apoE/P-selectin double-deficient mice to evaluate both the contribution of endothelial and platelet P-selectin to the development of atherosclerotic lesions and to the production of sP-selectin.

Mice and bone marrow transplantation

P-selectin–deficient (P-sel−/−) mice were backcrossed 4 times to C57BL/6J and interbred with apolipoprotein E–deficient (apoE−/−) mice that themselves were backcrossed 8 times to the C57BL/6J background.17Littermates from the F2 generation of the intercross were used to establish apoE−/−P-sel+/+ and apoE−/−P-sel−/− matings.8 For the preparation of bone marrow, donor mice were killed; femur and tibia bones were isolated under sterile conditions and the bone marrow cells were harvested by flushing the bones with medium (RPMI, 5% fetal bovine serum [FBS]). The cells were washed and resuspended in Hanks balanced salt solution (HBSS). Recipient 6-week-old male mice were lethally irradiated by exposure to a cesium source, 2 doses of 6 Gy, 3 hours apart. Immediately after the second dose of irradiation, the mice were injected with 1 × 107 freshly prepared donor bone marrow cells. For the first 4 weeks after transplantation, recipient mice were kept in a sterile unit and given sterile water and sterile food. Mice were kept on a normal chow diet containing 5% fat (wt/wt; Harlan/Teklad, Madison, WI) until 30 weeks after transplantation. Thirty-six-week-old, nonirradiated apoE−/−P-sel+/+ and apoE−/−P-sel−/− mice were used for reference. Before being killed, mice were fasted overnight and blood was collected through puncture of the retro-orbital venous plexus. Experimental procedures were approved by the Animal Care and Use Committee of the Center for Blood Research.

Determination of bone marrow reconstitution

Platelet-rich plasma (PRP) was obtained from ACD-anticoagulated blood (38 mM citric acid, 75 mM trisodium citrate, 100 mM dextrose; 10% final concentration) by centrifugation at 200g for 7 minutes and recentrifugation of the plasma and buffy coat for another 5 minutes at 180g. Platelets were washed with washing buffer (129 mM NaCl, 13.6 mM trisodium citrate, 11.1 mM dextrose, 1.6 mM KH2PO4, pH 6.8), centrifuged at 850gfor 12 minutes, and resuspended in resuspension buffer (137 mM NaCl; 4 mM KCl; 0.5 mM MgCl2; 0.5 mM sodium phosphate, 11.1 mM dextrose; 10 mM HEPES [N-2-hydroxyethylpiperazine-N′-2-ethanesulfonic acid]; pH 7.4). Platelets were activated with 0.5 U/mL thrombin (Sigma, St Louis, MO) for 10 minutes at 37°C, incubated for 40 minutes at room temperature with fluorescein isothiocyanate (FITC)-labeled antimouse P-selectin antibody clone RB40.34 (Pharmingen, San Diego, CA), and analyzed by fluorescence-activated cell sorting (FACS) on a Becton Dickinson FACSCalibur (Franklin Lakes, NJ). A complete blood cell count of EDTA (ethylenediaminetetraacetic acid)-anticoagulated blood was performed using an automatic cell counter (Coulter, Miami, FL).

Cholesterol determination

Total cholesterol concentrations in plasma were measured using an enzymatic assay kit (Infinity Cholesterol Reagent, Sigma).

Quantification of aortic sinus lesions

Mice were deeply anesthetized by intraperitoneal injection of avertin. After cutting open the right atrium, the heart and the vascular system were perfused in situ for 5 to 10 minutes with phosphate-buffered saline (PBS) until the outflow from the right atrium was clear. The hearts were collected, placed immediately in optimum cutting temperature (OCT) compound, and frozen on dry ice. Cryostat sections (10 μm) were cut at the level of the aortic sinus, collected on Superfrost microscopic glass slides, and stored at −20°C until further use. Sections were stained with oil red-O and hematoxylin (Sigma) and counterstained with light green (Sigma). Within the aortic sinus, lesions from 5 sections, each 80 μm apart, were measured by a blinded observer, using a Leica Q500MC image analysis program. Values reported represent the mean lesion area (± SEM) for each animal. Presence or absence of calcifications was evaluated for each section.

Immunohistochemical analysis

Sections 10-μm thick were fixed in cold acetone for 6 minutes. Sections were incubated for 10 minutes with 3% hydrogen peroxide to block endogenous peroxidase. After washing with PBS, unspecific binding sites were blocked by 30 minutes of incubation with 10% normal rabbit serum (for monoclonal) or goat serum (for polyclonal first antibodies) just prior to incubation with the following primary antibodies: antihuman smooth muscle α-actin, horseradish peroxidase (HRP) coupled (no. U7033; Dako, Carpinteria, CA); monoclonal rat antimouse platelet–endothelial cell adhesion molecule (PECAM), clone Mec13.3 (Pharmingen); monoclonal rat antimouse Mac-3 antigen, present on mouse mononuclear phagocytes, clone M3/84 (Pharmingen); polyclonal rabbit antimouse P-selectin (generous gift from Dr M. C. Berndt, Baker Medical Research Institute, Melbourne, Victoria, Australia); monoclonal rat antimouse glycoprotein (GP) IIbIIIa (generous gift from Dr A. K. Ng, University of Southern Maine, Portland). Biotinylated rabbit-antirat or goat antirabbit secondary antibodies and streptavidin-linked HRP (all from Vector Laboratories, Burlingame, CA) were used. 3-amino-9-ethylcarbazole (AEC; Vector Laboratories) served as substrate for HRP and sections were counterstained with hematoxylin (Sigma). For each animal smooth muscle cells (SMCs) were counted on the section immediately next to those that were used to quantify lesion size.

sP-selectin ELISA

Enzyme-linked immunosorbent assay (ELISA) plates were coated overnight with monoclonal antimouse P-selectin antibodies, clone RB40.34 (Pharmingen), at 4°C. Plates were washed 3 times with PBS/0.1% Triton X-100/0.5% bovine serum albumin (BSA) and incubated for 45 minutes with blocking solution (PBS/0.1% Triton X-100/1% BSA). After removing the blocking solution, plasma diluted 1:15 in “diluent solution” from the soluble P-selectin ELISA kit (BEE 6; R & D Systems, Minneapolis, MN), was applied and incubated for 30 minutes. The “sP-selectin conjugate,” containing a polyclonal antihuman P-selectin antibody cross-reacting with mouse P-selectin, was then added and incubated for an additional 80 minutes. Plates were washed 3 times with 300 μL “wash buffer” and incubated for 15 minutes with 100 μL “substrate.” The reaction was stopped by adding 100 μL “stop solution” (reagents in quotation marks were all from the BEE 6 kit). A DYNEX ELISA-reader (Thermo Labsystems, Helsinki, Finland), set to 450 nm, with wavelength correction set to 650 nm, determined the optical density (OD).

Statistical analysis

In most instances, several group of animals with transplants were compared; ANOVA and Fisher protected least significant difference (PLSD) post hoc tests were performed. When only 2 groups were compared (Figure 2B), unpaired t test was applied. Statview software (SAS Institute, Cary, NC) was used and data are presented as mean ± SEM.

Characteristics of apoE-deficient mice receiving bone marrow transplants

At age 6 weeks, male apoE−/−P-sel−/−and apoE−/−P-sel+/+ mice were lethally irradiated and reconstituted with bone marrow of either genotype to obtain 4 groups of animals: apoE−/−P-sel+/+bone marrow transplanted into apoE−/−P-sel+/+recipient mice (wt-wt), apoE−/−P-sel−/−bone marrow transplanted into apoE−/−P-sel+/+mice (ko-wt), apoE−/−P-sel+/+ bone marrow transplanted into apoE−/−P-sel−/− mice (wt-ko), and apoE−/−P-sel−/− bone marrow transplanted into apoE−/−P-sel−/− mice (ko-ko). Thus, for clarity, the nomenclature of the chimeric mice denotes first the P-selectin genotype of platelets and second that of endothelium. Characteristics of the different groups of mice, before death at the end of the study, are listed in Table1.

Table 1.

Characteristics of the transplanted apoE−/−animals

P-selectin genotypePltsECsCholesterol level, mg/dLWBC count, × 103 cells/μLPlt count, × 103 cells/μLSpleen weight, g
wt-wt wt wt 507.3 ± 38.8 9.6 ± 1.7 1389 ± 81 0.087 ± 0.012 
ko-wt ko wt 512.8 ± 65 8.5 ± 1.2 1216 ± 112 0.138 ± 0.006 
wt-ko wt ko 411.1 ± 17 5.4 ± 1.9 1159 ± 77 0.094 ± 0.013 
ko-ko ko ko 474.2 ± 48.1 6.0 ± 1.1 1152 ± 143 0.178 ± 0.022 
P-selectin genotypePltsECsCholesterol level, mg/dLWBC count, × 103 cells/μLPlt count, × 103 cells/μLSpleen weight, g
wt-wt wt wt 507.3 ± 38.8 9.6 ± 1.7 1389 ± 81 0.087 ± 0.012 
ko-wt ko wt 512.8 ± 65 8.5 ± 1.2 1216 ± 112 0.138 ± 0.006 
wt-ko wt ko 411.1 ± 17 5.4 ± 1.9 1159 ± 77 0.094 ± 0.013 
ko-ko ko ko 474.2 ± 48.1 6.0 ± 1.1 1152 ± 143 0.178 ± 0.022 

There were no significant differences due to the P-selectin genotype in serum total cholesterol, white blood cell (WBC) or platelet counts among the 4 groups of transplanted animals.

In contrast, the genotype of the chimeric animals affected splenic weights. Platelet P-selectin appeared to be the most important factor determining the weight of the organ (P determined by ANOVA and Fisher PLSD post-hoc test: wt-wt, ko-ko < .0001; wt-wt, ko-wt < .02; wt-ko, ko-wt < .04; wt-wt, wt-ko = .74; ko-wt, ko-ko < .06). Thus, the presence of platelet P-selectin appears to prevent the appearance of the enlarged spleen seen in apoE−/−P-sel−/− mice.8 

Plts indicates platelets; ECs, endothelial cells; WBC, white blood cell; n = 6-10.

Cholesterol values, WBC count, and platelet count were not significantly different among transplanted animals. ApoE−/−P-sel−/− animals were previously described as displaying splenomegaly with a large infiltrate of macrophages.8 In this study, the ko-ko animals had larger spleens than wt-wt mice (Table 1). The transplantation procedure did not affect spleen size significantly when compared to age-matched controls without transplants. Interestingly, reconstituting the P-sel–deficient animals with wild-type platelets (wt-ko) appeared to normalize the splenic weights so that they became similar to those of wt-wt mice (P = .74) and smaller than those of ko-ko mice (P < .0002). At this time, it is not clear how platelet P-selectin prevents splenomegaly nor whether it interferes with recruitment/retention of macrophages in the spleen.

Flow cytometer analysis of P-selectin expression on activated platelets at 36 weeks showed that more than 95% of the platelets in all mice chimeric for P-selectin were of donor origin, indicating that the bone marrow transplantation was successful (Figure1). ApoE−/−P-sel−/− mice bred poorly and showed significantly higher mortality than apoE−/−mice.8,18 Thus, the mice receiving bone marrow transplants presented in this study were generated in many small groups over a period of 2 years avoiding cage effects of housing.

Fig. 1.

Platelets in mice that received bone marrow transplants were of donor genotype.

Platelets isolated from the mice receiving transplants were activated by thrombin, incubated with FITC-labeled anti P-selectin antibodies, and analyzed by flow cytometry. A representative histogram for each set of chimeric animals is shown; ko-wt (A), wt-ko (B). The shaded curve represents P-selectin expression on activated platelets from an animal receiving a bone marrow transplant, whereas the nonshaded curve was obtained by staining activated platelets of a control mouse without transplant of the same genotype as the recipient.

Fig. 1.

Platelets in mice that received bone marrow transplants were of donor genotype.

Platelets isolated from the mice receiving transplants were activated by thrombin, incubated with FITC-labeled anti P-selectin antibodies, and analyzed by flow cytometry. A representative histogram for each set of chimeric animals is shown; ko-wt (A), wt-ko (B). The shaded curve represents P-selectin expression on activated platelets from an animal receiving a bone marrow transplant, whereas the nonshaded curve was obtained by staining activated platelets of a control mouse without transplant of the same genotype as the recipient.

Close modal

Role of endothelial and platelet P-selectin in atherosclerotic lesion formation

After transplantation, mice were maintained on regular mouse chow for 30 weeks before collection of the hearts for analysis. We found that although the lesions at the aortic sinus were well developed at this stage, there were too few lesions in the rest of the aortic tree for comparative analysis. There are 2 likely reasons for this. First, in the absence of atherogenic diet, lesions in the aortic tree develop slowly in the apoE−/− mouse. We observed only 12% of the surface covered with lesions at 15 months and no lesions at 4 months of age.8 Second, it was reported that irradiation/bone marrow transplantation increases lesion growth in the aortic sinus while delaying lesion formation in the rest of the aorta.19 Absence of endothelial P-selectin alone produced a significant reduction (P < .0001) in lesion size (wt-ko: 0.334 ± 0.038 mm2, n = 8) compared with wt-wt lesions (wt-wt: 0.669 ± 0.047 mm2, n = 10). In the absence of endothelial P-selectin, the lesions were relatively small whether or not platelets were expressing P-selectin (ko-ko: 0.271 ± 0.032 mm2, n = 6; wt-ko: 0.334 ± 0.038 mm2, n = 8; P = .33), indicating that endothelial P-selectin is very important for lesion growth. Interestingly, the mean atherosclerotic lesions of ko-wt mice (0.513 ± 0.034 mm2, n = 9) were significantly smaller than the lesions of wt-wt mice (0.669 ± 0.047 mm2, n = 10; P < .008), demonstrating that, when endothelial P-selectin is present, platelets and platelet P-selectin also play a major role in mediating atherosclerotic lesion formation. Lesions of wt-wt animals (0.669 ± 0.047 mm2, n = 10) had a tendency to be slightly larger than those of apoE−/− mice without transplants (0.554 ± 0.041 mm2, n = 5;P = 0.14) as was reported for the aortic sinus lesions of irradiated low-density lipoprotein (LDL) receptor-deficient mice with transplants.19 Lesions of apoE−/−P-sel−/− mice without transplants (0.290 ± 0.018 mm2, n = 4) were much smaller than apoE−/−P-sel+/+ mice and similar in size to lesions of ko-ko animals (0.271 ± 0.032 mm2, n = 6;P = .68) likely due to P-selectin deficiency in these animals (Figure 2).

Fig. 2.

Endothelial and platelet P-selectin both affect the size of atherosclerotic lesions formed in apoE−/− animals.

Lesions at the aortic sinus were stained with oil red-O and measured. Dots represent the mean lesion size per animal. The bar indicates the mean lesion area ± SEM of the corresponding group. (A) Animals that received bone marrow transplants. Wt-wt and ko-wt animals had significantly larger lesions than wt-ko and ko-ko animals, establishing that endothelial P-selectin is crucial in atherosclerosis. Wt-wt animals had larger lesions than ko-wt animals, showing that also platelet P-selectin is important in atherosclerotic lesion growth. (B) Mice that did not receive transplants. The lesions in apoE−/−P-sel+/+ (wt) and apoE−/−P-sel−/− (ko) mice were not significantly different from those of wt-wt and ko-ko mice, respectively (shown in panel A).

Fig. 2.

Endothelial and platelet P-selectin both affect the size of atherosclerotic lesions formed in apoE−/− animals.

Lesions at the aortic sinus were stained with oil red-O and measured. Dots represent the mean lesion size per animal. The bar indicates the mean lesion area ± SEM of the corresponding group. (A) Animals that received bone marrow transplants. Wt-wt and ko-wt animals had significantly larger lesions than wt-ko and ko-ko animals, establishing that endothelial P-selectin is crucial in atherosclerosis. Wt-wt animals had larger lesions than ko-wt animals, showing that also platelet P-selectin is important in atherosclerotic lesion growth. (B) Mice that did not receive transplants. The lesions in apoE−/−P-sel+/+ (wt) and apoE−/−P-sel−/− (ko) mice were not significantly different from those of wt-wt and ko-ko mice, respectively (shown in panel A).

Close modal

Composition of atherosclerotic lesions of the mice with transplants

Lesions, stained with oil red-O and counterstained with light green, showed extensive lipid deposition (Figure3). Immunohistochemical staining for macrophages confirmed that most of the cells within the lesions were macrophages (> 90%). We also evaluated the presence of SMCs cells in the lesions with antibodies to α-actin. In all 4 groups of mice most of the lesions reached an early fibro-fatty stage, that is, contained SMCs and some lesions were calcified (Table2).

Fig. 3.

Photographs of the aortic sinus of the animals with bone marrow transplants depict the atherosclerotic lesions formed.

Representative sections of the 4 different sets of animals, stained with oil red-O and counterstained with hematoxylin and light green are as indicated in upper left corner. Lesions of all genotypes show extensive foam cell accumulation. Bar = 1 mm.

Fig. 3.

Photographs of the aortic sinus of the animals with bone marrow transplants depict the atherosclerotic lesions formed.

Representative sections of the 4 different sets of animals, stained with oil red-O and counterstained with hematoxylin and light green are as indicated in upper left corner. Lesions of all genotypes show extensive foam cell accumulation. Bar = 1 mm.

Close modal
Table 2.

Characteristics of lesions

Genotypewt-wtko-wtwt-koko-ko
% animals showing calcification 80 (10) 44.4 (9) 37.5 (8) 33.3 (6) 
SMCs/section 15.5 ± 3.5 (9) 10.2 ± 2.4 (8) 5.4 ± 1.6 (8) 1.9 ± 0.5 (6) 
SMC density SMCs/mm2 23.7 ± 6.2 (9) 20.0 ± 4.9 (8) 13.3 ± 3.3 (8) 9.2 ± 3.1 (6) 
Genotypewt-wtko-wtwt-koko-ko
% animals showing calcification 80 (10) 44.4 (9) 37.5 (8) 33.3 (6) 
SMCs/section 15.5 ± 3.5 (9) 10.2 ± 2.4 (8) 5.4 ± 1.6 (8) 1.9 ± 0.5 (6) 
SMC density SMCs/mm2 23.7 ± 6.2 (9) 20.0 ± 4.9 (8) 13.3 ± 3.3 (8) 9.2 ± 3.1 (6) 

Numbers in parentheses indicate number of animals analyzed.

Interestingly, the total number of SMCs in the lesions was a function of both platelet and endothelial P-selectin, with highest SMC numbers in lesions expressing P-selectin on both cell types (15.5 ± 3.5 SMCs/lesion, n = 9), medium number in lesions expressing P-selectin on one cell type only (7.8 ± 1.5 SMCs/lesion, n = 16), and the lowest in animals without P-selectin (1.9 ± 0.5 SMCs/lesion, n = 6; Figure 4). The density of the SMCs in the lesions also apparently depended on both platelet and endothelial P-selectin, although this did not reach statistical significance (Table 2). Compared with wt-wt mice, the percentage of animals showing calcifications in lesions was markedly reduced in chimeras missing either endothelial or platelet P-selectin (Table 2). Immunohistochemical staining for GPIIbIIIa showed that platelets were present in atherosclerotic lesions. However, numbers of platelets were low and no significant differences among the 4 groups could be established. Staining for PECAM-1 indicated that the observed platelets were not within atherosclerotic neo-vessels.

Fig. 4.

Immunohistochemical staining for α-actin shows positive SMCs in atherosclerotic lesions.

(A) wt-wt; (B) ko-wt; (C) ko-ko animals; bar = 0.2 mm; (D) α-actin–positive SMCs within lesions were counted on 5 aortic sinus sections 80 μm apart. Dots represent the mean number of SMCs per section for each animal. Black dots (●) correspond to animals that lack endothelial P-selectin (ie, wt-ko, ko-ko). Bars show the mean ± SEM for each group. The atherosclerotic lesions of the chimeric animals appear to have an intermediate content of SMCs.

Fig. 4.

Immunohistochemical staining for α-actin shows positive SMCs in atherosclerotic lesions.

(A) wt-wt; (B) ko-wt; (C) ko-ko animals; bar = 0.2 mm; (D) α-actin–positive SMCs within lesions were counted on 5 aortic sinus sections 80 μm apart. Dots represent the mean number of SMCs per section for each animal. Black dots (●) correspond to animals that lack endothelial P-selectin (ie, wt-ko, ko-ko). Bars show the mean ± SEM for each group. The atherosclerotic lesions of the chimeric animals appear to have an intermediate content of SMCs.

Close modal

Hematopoietic progenitor cells, released from the bone marrow, were shown to participate in angiogenesis20,21 and could potentially also replace endothelium over damaged atherosclerotic areas. By immunochemistry, we did not detect P-selectin–positive endothelial cells in the aortic sinus section of wt-ko animals. In addition, the fact that there was no significant difference in lesion size between wt-ko and ko-ko animals indicates that bone marrow–derived endothelialization did not play a significant role in promoting atherosclerosis in the apoE−/− mice. We cannot conclude that no angioblasts were recruited to the lesion from the transplanted bone marrow, only that the large majority of the endothelium within and covering the lesion was of the recipient genotype.

Origin of soluble P-selectin in plasma of apoE−/− mice

P-selectin on the cell surface of activated platelets or endothelial cells can be shed and circulates in the soluble form in plasma. Because sP-selectin has procoagulant activity that could play an important role during plaque rupture, we were interested in establishing the cellular origin of this molecule. We measured sP-selectin by a sandwich ELISA. As in clinical studies,22we did not ultracentrifuge the plasma and therefore we did not differentiate between microparticle-associated and free-in-plasma P-selectin.14,23 Measurements of plasma from all ko-ko animals gave background OD readings of zero. When we considered the mean OD reading of wt-wt animals as 100%, soluble P-selectin levels in ko-wt animals represented 57.6% of this value, whereas levels of wt-ko animals represented only 15.1%. The small increment of sP-selectin coming from the platelets of wt-ko mice did not reach statistical significance when compared with mice not expressing P-selectin (ko-ko). This indicates that the majority of the soluble P-selectin in the apoE−/− mice was of endothelial origin (Figure5).

Fig. 5.

sP-selectin in apoE−/− mice originates predominantly from endothelium.

Plasma sP-selectin levels of mice that received transplants were measured by ELISA at the end of the study. OD readings from plasma of wt-wt animals were considered as 100%. Bars represent the mean OD ± SEM.

Fig. 5.

sP-selectin in apoE−/− mice originates predominantly from endothelium.

Plasma sP-selectin levels of mice that received transplants were measured by ELISA at the end of the study. OD readings from plasma of wt-wt animals were considered as 100%. Bars represent the mean OD ± SEM.

Close modal

It is becoming increasingly clear that P-selectin is a pivotal component in cardiovascular disease and therefore is a potential therapeutic target. The absence of P-selectin delays and reduces atherosclerotic lesion formation in mice.8,9,24,25 In several animal models of restenosis, deficiency or blocking of P-selectin with antibodies or recombinant soluble P-selectin glycoprotein ligand 1 (PSGL-1) inhibits neointimal growth after arterial injury.18,26-29 P-selectin mediates monocyte rolling ex vivo on dissected atherosclerotic arteries3 and thus could be involved in leukocyte recruitment into the lesions. Although the role of endothelial P-selectin in monocyte recruitment to the growing atherosclerotic lesion seems intuitive,30until now it was not directly demonstrated as the gene targeting and inhibition studies described affect both endothelial and platelet P-selectin. Similarly, it has not been shown whether platelets with their P-selectin contribute to lesion development. In the case of plaque rupture, soluble P-selectin may help to promote thrombus formation and fibrin deposition,15 eventually leading to a myocardial infarction. In the present study we have evaluated the separate contributions of platelets versus endothelial P-selectin in the development of atherosclerosis and in the generation of plasma sP-selectin.

sP-selectin, either endogenously generated through proteolytic cleavage of surface-expressed P-selectin or infused intravenously in its recombinant form, stimulates the release of procoagulant microparticles in mice, some of which contain tissue factor.15 Thus high levels of sP-selectin induce a procoagulant state. This observation may explain why high levels of sP-selectin in apparently healthy women are associated with a higher risk of future cardiovascular events.22 The origin of sP-selectin in blood remains an open question. Positive correlation of sP-selectin levels with the platelet count31 and with released platelet proteins, as well as lack of correlation with von Willebrand factor, an endothelial cell activation marker,32 led to the assumption that sP-selectin originates predominantly from platelets. Because high levels of sP-selectin might contribute to the severity of many thrombotic and cardiovascular diseases such as unstable angina,33 we felt it would be clinically relevant to determine the cellular origin of sP-selectin to eventually devise ways to inhibit its generation in patients. Our bone marrow transplantation experiment provided an excellent opportunity to establish the origin of sP-selectin. To our surprise we observed that, in atherosclerotic apoE−/− mice, endothelial cells released almost 4 times more sP-selectin than platelets (Figure 5). Thus, sP-selectin levels reflected more endothelial than platelet activation and we therefore question the use of sP-selectin as a platelet activation marker in the context of atherosclerosis. High levels of fibrinogen and coagulation factors are well-known risk factors for coronary artery disease.34,35 Because sP-selectin through its procoagulant activity could stimulate deposition of fibrin, it could also promote atherosclerosis. Because platelets contributed relatively little sP-selectin in this model of atherosclerosis, we believe that their significant contribution to lesion development extends beyond their share of sP-selectin.

Our study documents that endothelial P-selectin plays a prominent role in atherosclerosis. ApoE−/− mice in which endothelial P-selectin was present (wt-wt and ko-wt) had significantly larger lesions than mice that were deficient for endothelial P-selectin (ko-ko and wt-ko). Platelets release many growth factors, chemokines, and small molecules promoting hemostasis. Whether platelets are involved in atherosclerosis before plaque rupture occurs was not clear. In this study, we provide evidence that platelets play an important role in atherosclerotic lesion formation because we observed that the lesions of wt-wt animals were 30% larger than lesions of ko-wt animals (Figure2). How could platelets expressing P-selectin promote atherosclerosis? There are several possibilities, which are not mutually exclusive: (1) P-selectin expressed on activated platelets causes formation of platelet-neutrophil36 and platelet-monocyte aggregates in blood.37,38 Platelet microparticles, containing P-selectin, were also shown to support leukocyte-leukocyte interaction under flow.39 These associations could augment infiltration of monocytes into atherosclerotic lesions,11,28 similar to L-selectin promoting secondary capture of leukocytes at sites of inflammation.40 (2) It was recently reported that platelet-derived microparticles associate with PSGL-1–expressing hematopoietic stem cells and facilitate their homing to the bone marrow.41 By a similar mechanism, P-selectin on microparticles could coat monocytes and contribute to their recruitment into atherosclerotic lesions. Interestingly, monocytoid cells were shown to roll and adhere to endothelium in vitro at a much higher shear rate when complexed with activated platelets than alone.11 This could help homing of the monocytes in unfavorable shear stress conditions. (3) Platelets and platelet microparticles were shown to deposit chemokines, such as RANTES42 or other agents,43 on endothelium, which can attract and activate monocytes, thus possibly promoting their recruitment into the lesions. (4) Platelets or substances released from activated platelets were shown to induce in vitro macrophage foam cell formation and thereby could contribute to lesion growth.44-46 

P-selectin also plays a role in maturation of atherosclerotic lesions. It was previously shown that most (70%) of 4-month-old apoE−/− mice already had SMC-containing fibro-fatty lesions, whereas none of the apoE−/−P-sel−/− mice had SMCs in their lesions at that age.8 We now show that both endothelial and platelet P-selectin apparently contribute to lesion maturation. Lesions expressing either endothelial or platelet P-selectin showed an intermediate level of maturity in regard to SMC content, that is, they were less mature than lesions where both cell types expressed P-selectin but showed a tendency to be more mature than those where no P-selectin was expressed (Figure 4; Table 2). This indicates that the recruitment of SMCs might be influenced by the amount of P-selectin expressed. Contact with P-selectin, presented either by platelets or endothelium, might activate monocytes entering or within the atherosclerotic lesion to secrete chemokines that attract or stimulate proliferation of SMCs.47 Similarly, in comparison with wt-wt animals, we observed a striking reduction in the number of animals that showed calcification in their lesions if either endothelial or platelet P-selectin were missing (Table 2).

In conclusion, we have shown that most of the procoagulant sP-selectin in atherosclerotic apoE−/− mice came from endothelium rather than platelets. We further demonstrated that endothelial P-selectin is crucial for atherosclerotic lesion growth and more unexpectedly that platelets and platelet P-selectin also play an important role in atherosclerotic lesion progression. Thus, agents inhibiting P-selectin or preventing platelet activation could become effective tools in reducing atherosclerotic lesion development and preventing acute cardiovascular events.

We thank Ali Hafezi-Moghadam for critical reading of the manuscript and Hongwei Wang for help with statistical analysis; Simin Saffaripour, Maria Economopoulos, Patrick André, and Christine Livingston for advice and help; and Lesley Cowan for assistance in preparing the manuscript. We also thank Dr M. C. Berndt, Baker Medical Research Institute, Melbourne, Victoria, Australia, and Dr A. K. Ng, University of Southern Maine, Portland, for providing antibodies.

Prepublished online as Blood First Edition Paper, December 12, 2002; DOI 10.1182/blood-2002-07-2209.

Supported by National Heart, Lung and Blood Institute grant R01 HL 53756 (D.D.W.). P.C.B. was supported by a stipend from the Swiss National Science Foundation.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Ross
R
Atherosclerosis—an inflammatory disease.
N Engl J Med.
340
1999
115
126
2
Vestweber
D
Blanks
JE
Mechanisms that regulate the function of the selectins and their ligands.
Physiol Rev.
79
1999
181
213
3
Ramos
CL
Huo
Y
Jung
U
et al
Direct demonstration of P-selectin- and VCAM-1-dependent mononuclear cell rolling in early atherosclerotic lesions of apolipoprotein E- deficient mice.
Circ Res.
84
1999
1237
1244
4
Butcher
EC
Leukocyte-endothelial cell recognition: three (or more) steps to specificity and diversity.
Cell.
67
1991
1033
1036
5
Springer
TA
Traffic signals for lymphocyte recirculation and leukocyte emigration: the multistep paradigm.
Cell.
76
1994
301
314
6
Mayadas
TN
Johnson
RC
Rayburn
H
Hynes
RO
Wagner
DD
Leukocyte rolling and extravasation are severely compromised in P selectin-deficient mice.
Cell.
74
1993
541
554
7
Subramaniam
M
Saffaripour
S
Watson
SR
Mayadas
TN
Hynes
RO
Wagner
DD
Reduced recruitment of inflammatory cells in a contact hypersensitivity response in P-selectin-deficient mice.
J Exp Med.
181
1995
2277
2282
8
Dong
ZM
Brown
AA
Wagner
DD
Prominent role of P-selectin in the development of advanced atherosclerosis in apoE-deficient mice.
Circulation.
101
2000
2290
2295
9
Collins
RG
Velji
R
Guevara
NV
Hicks
MJ
Chan
L
Beaudet
AL
P-selectin or intercellular adhesion molecule (ICAM)-1 deficiency substantially protects against atherosclerosis in apolipoprotein E-deficient mice.
J Exp Med.
191
2000
189
194
10
Larsen
E
Celi
A
Gilbert
GE
et al
PADGEM protein: a receptor that mediates the interaction of activated platelets with neutrophils and monocytes.
Cell.
59
1989
305
312
11
Theilmeier
G
Lenaerts
T
Remacle
C
Collen
D
Vermylen
J
Hoylaerts
MF
Circulating activated platelets assist THP-1 monocytoid/endothelial cell interaction under shear stress.
Blood.
94
1999
2725
2734
12
Ross
R
Harker
L
Hyperlipidemia and atherosclerosis.
Science.
193
1976
1094
1100
13
Harker
LA
Ross
R
Slichter
SJ
Scott
CR
Homocystine-induced arteriosclerosis: the role of endothelial cell injury and platelet response in its genesis.
J Clin Invest.
58
1976
731
741
14
Dunlop
LC
Skinner
MP
Bendall
LJ
et al
Characterization of GMP-140 (P-selectin) as a circulating plasma protein.
J Exp Med.
175
1992
1147
1150
15
Andre
P
Hartwell
D
Hrachovinova
I
Saffaripour
S
Wagner
DD
Pro-coagulant state resulting from high levels of soluble P-selectin in blood.
Proc Natl Acad Sci U S A.
97
2000
13835
13840
16
Blann
AD
Lip
GY
Hypothesis: is soluble P-selectin a new marker of platelet activation?
Atherosclerosis.
128
1997
135
138
17
Zhang
SH
Reddick
RL
Piedrahita
JA
Maeda
N
Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E.
Science.
258
1992
468
471
18
Manka
D
Collins
RG
Ley
K
Beaudet
AL
Sarembock
IJ
Absence of P-selectin, but not intercellular adhesion molecule-1, attenuates neointimal growth after arterial injury in apolipoprotein E- deficient mice.
Circulation.
103
2001
1000
1005
19
Schiller
NK
Kubo
N
Boisvert
WA
Curtiss
LK
Effect of gamma-irradiation and bone marrow transplantation on atherosclerosis in LDL receptor-deficient mice.
Arterioscler Thromb Vasc Biol.
21
2001
1674
1680
20
Asahara
T
Murohara
T
Sullivan
A
et al
Isolation of putative progenitor endothelial cells for angiogenesis.
Science.
275
1997
964
967
21
Hattori
K
Dias
S
Heissig
B
et al
Vascular endothelial growth factor and angiopoietin-1 stimulate postnatal hematopoiesis by recruitment of vasculogenic and hematopoietic stem cells.
J Exp Med.
193
2001
1005
1014
22
Ridker
PM
Buring
JE
Rifai
N
Soluble P-selectin and the risk of future cardiovascular events.
Circulation.
103
2001
491
495
23
Hartwell
DW
Mayadas
T
Frenette
PS
et al
Role of P-selectin cytoplasmic domain in granular targeting in vivo and in early inflammatory responses.
J Cell Biol.
143
1998
1129
1141
24
Johnson
RC
Chapman
SM
Dong
ZM
et al
Absence of P-selectin delays fatty streak formation in mice.
J Clin Invest.
99
1997
1037
1043
25
Nageh
MF
Sandberg
ET
Marotti
KR
et al
Deficiency of inflammatory cell adhesion molecules protects against atherosclerosis in mice.
Arterioscler Thromb Vasc Biol.
17
1997
1517
1520
26
Hayashi
S
Watanabe
N
Nakazawa
K
et al
Roles of P-selectin in inflammation, neointimal formation, and vascular remodeling in balloon-injured rat carotid arteries.
Circulation.
102
2000
1710
1717
27
Bienvenu
JG
Tanguay
JF
Theoret
JF
Kumar
A
Schaub
RG
Merhi
Y
Recombinant soluble P-selectin glycoprotein ligand-1-Ig reduces restenosis through inhibition of platelet-neutrophil adhesion after double angioplasty in swine.
Circulation.
103
2001
1128
1134
28
Smyth
SS
Reis
ED
Zhang
W
Fallon
JT
Gordon
RE
Coller
BS
Beta(3)-integrin-deficient mice but not P-selectin-deficient mice develop intimal hyperplasia after vascular injury: correlation with leukocyte recruitment to adherent platelets 1 hour after injury.
Circulation.
103
2001
2501
2507
29
Wang
K
Zhou
Z
Zhou
X
Tarakji
K
Topol
EJ
Lincoff
AM
Prevention of intimal hyperplasia with recombinant soluble P-selectin glycoprotein ligand-immunoglobulin in the porcine coronary artery balloon injury model.
J Am Coll Cardiol.
38
2001
577
582
30
Dong
ZM
Wagner
DD
Leukocyte-endothelium adhesion molecules in atherosclerosis.
J Lab Clin Med.
132
1998
369
375
31
Fijnheer
R
Frijns
CJ
Korteweg
J
et al
The origin of P-selectin as a circulating plasma protein.
Thromb Haemost.
77
1997
1081
1085
32
Blann
AD
Lip
GY
Beevers
DG
McCollum
CN
Soluble P-selectin in atherosclerosis: a comparison with endothelial cell and platelet markers.
Thromb Haemost.
77
1997
1077
1080
33
Ikeda
H
Takajo
Y
Ichiki
K
et al
Increased soluble form of P-selectin in patients with unstable angina.
Circulation.
92
1995
1693
1696
34
Meade
TW
Mellows
S
Brozovic
M
et al
Haemostatic function and ischaemic heart disease: principal results of the Northwick Park Heart Study.
Lancet.
2
1986
533
537
35
Folsom
AR
Wu
KK
Rosamond
WD
Sharrett
AR
Chambless
LE
Prospective study of hemostatic factors and incidence of coronary heart disease: the Atherosclerosis Risk in Communities (ARIC) Study.
Circulation.
96
1997
1102
1108
36
Ott
I
Neumann
FJ
Gawaz
M
Schmitt
M
Schomig
A
Increased neutrophil-platelet adhesion in patients with unstable angina.
Circulation.
94
1996
1239
1246
37
Rinder
HM
Bonan
JL
Rinder
CS
Ault
KA
Smith
BR
Dynamics of leukocyte-platelet adhesion in whole blood.
Blood.
78
1991
1730
1737
38
Michelson
AD
Barnard
MR
Krueger
LA
Valeri
CR
Furman
MI
Circulating monocyte-platelet aggregates are a more sensitive marker of in vivo platelet activation than platelet surface P-selectin: studies in baboons, human coronary intervention, and human acute myocardial infarction.
Circulation.
104
2001
1533
1537
39
Forlow
SB
McEver
RP
Nollert
MU
Leukocyte-leukocyte interactions mediated by platelet microparticles under flow.
Blood.
95
2000
1317
1323
40
Eriksson
EE
Xie
X
Werr
J
Thoren
P
Lindbom
L
Importance of primary capture and L-selectin-dependent secondary capture in leukocyte accumulation in inflammation and atherosclerosis in vivo.
J Exp Med.
194
2001
205
218
41
Janowska-Wieczorek
A
Majka
M
Kijowski
J
et al
Platelet-derived microparticles bind to hematopoietic stem/progenitor cells and enhance their engraftment.
Blood.
98
2001
3143
3149
42
von Hundelshausen
P
Weber
KS
Huo
Y
et al
RANTES deposition by platelets triggers monocyte arrest on inflamed and atherosclerotic endothelium.
Circulation.
103
2001
1772
1777
43
Barry
OP
Pratico
D
Savani
RC
FitzGerald
GA
Modulation of monocyte-endothelial cell interactions by platelet microparticles.
J Clin Invest.
102
1998
136
144
44
Curtiss
LK
Black
AS
Takagi
Y
Plow
EF
New mechanism for foam cell generation in atherosclerotic lesions.
J Clin Invest.
80
1987
367
373
45
Mendelsohn
ME
Loscalzo
J
Role of platelets in cholesteryl ester formation by U-937 cells.
J Clin Invest.
81
1988
62
68
46
Maor
I
Brook
GJ
Aviram
M
Platelet secreted lipoprotein-like particle is taken up by the macrophage scavenger receptor and enhances cellular cholesterol accumulation.
Atherosclerosis.
88
1991
163
174
47
Weyrich
AS
Elstad
MR
McEver
RP
et al
Activated platelets signal chemokine synthesis by human monocytes.
J Clin Invest.
97
1996
1525
1534

Author notes

Denisa D. Wagner, Center for Blood Research, 800 Huntington Ave, Boston, MA 02115; e-mail:wagner@cbr.med.harvard.edu.

Sign in via your Institution