Cerebrovascular and cardiovascular diseases are a major cause of morbidity and mortality. Soluble P-selectin (sP-selectin) is a biomarker for platelet/endothelial activation and is considered a risk factor for vascular disease. sP-selectin enhances procoagulant activity by inducing leukocyte-derived microparticle production and promotes activation of leukocyte integrins. However, it is not known whether it directly contributes to vascular complications. We investigated the effect of increased levels of sP-selectin on blood-brain barrier (BBB) function, stroke outcome, and atherosclerosis by comparing wild-type mice with P-selΔCTCT mice in which the endogenous P-selectin gene was replaced with a mutant that produces abnormally high plasma levels of sP-selectin. P-selΔCTCT mice presented several abnormalities, including (1) higher BBB permeability, with 25% of the animals showing differential permeability between the right and left hemispheres; (2) altered social behavior with increased aggression; (3) larger infarcts in the middle cerebral artery occlusion ischemic stroke model; and (4) increased susceptibility to atherosclerotic, macrophage-rich lesion development in both male and female mice on the apoE−/− genetic background. Thus, elevated sP-selectin is not only a biomarker for vascular disease, but also may contribute directly to atherosclerosis and cerebrovascular complications.

P-selectin is a cell surface adhesion molecule stored in the membranes of platelets' α-granules and in the Weibel-Palade bodies of endothelial cells.1,2  It is expressed on the cell surface upon granule exocytosis and plays an essential role in the initial recruitment of leukocytes to the sites of injury during inflammation.3  P-selectin consists of an N-terminal lectin domain, followed by an epidermal growth factor–like motif, a series of short consensus repeats, a transmembrane domain, and a short cytoplasmic tail.4  Lectin and epidermal growth factor domains are required to bind P-selectin glycoprotein ligand-1 (PSGL-1) on leukocytes, an interaction critical for leukocyte rolling and capable of leukocyte activation.5  The cytoplasmic domain of P-selectin is essential for its storage in Weibel-Palade bodies and its internalization from the plasma membrane.6  The membrane form of P-selectin has been shown to be involved in both inflammation and thrombosis.7  P-selectin–deficient mice are defective in leukocyte rolling, extravasation, and hemostasis.3,8  P-selectin deficiency delays and reduces the formation of atherosclerotic plaques, suggesting that P-selectin is involved in atherosclerosis, a chronic inflammatory disease.9 

A soluble form of P-selectin is derived either from alternative mRNA splicing that generates an isoform that lacks the transmembrane domain and/or from proteolytic cleavage of the membrane-bound form.10  Soluble P-selectin (sP-selectin) has been proposed as a useful biomarker in various pathologic states in which platelets and/or endothelial cells are activated. It is elevated in several conditions, including hyperlipidemia,11  hypertension,12  ischemic heart disease,13  atherosclerosis,14  and in patients with vascular disorders such as peripheral arterial occlusive disease15  and postangioplasty restenosis.16  Ridker et al17  showed that in apparently healthy women, elevated baseline levels of sP-selectin were associated with an increased risk of future myocardial infarction, stroke, and cardiovascular death.

Previously, Hartwell et al18  made genetically engineered knockin mice that express P-selectin without the cytoplasmic domain (P-selΔCTCT). The deletion of the cytoplasmic domain of P-selectin did not affect the level of P-selectin, neither on the endothelial cells' surface nor in the α-granules of platelets, but it severely compromised the storage and the regulated secretion of P-selectin from endothelial cells. The ΔCT P-selectin is proteolytically shed from the plasma membrane, which leads to several-fold increased levels of sP-selectin in the plasma.18  This increase in sP-selectin in P-selΔCTCT mice is associated with enhanced generation of tissue factor–expressing microparticles, resulting in shorter plasma-clotting time and increased fibrin deposition on platelet thrombi reflecting a procoagulant phenotype.19  Recently, the increase in fibrin deposition in the brain in a mouse model of Alzheimer disease was shown to be associated with increased blood-brain barrier (BBB) permeability.20 

In this study, taking advantage of the genetic model P-selΔCTCT, we investigated the potential impact of elevated sP-selectin and the procoagulant phenotype of these mice on BBB permeability and infarct size in experimental stroke. We further investigated whether elevated sP-selectin levels influence inflammation/atherosclerosis when crossed onto atherosclerosis-susceptible apoE−/− background.

Animals

Except for the atherosclerosis study, the mice were siblings produced by crosses of P-selΔCT/WT mice on B6/129 background. Factor V Leiden (FVL) mice21  were obtained from The Jackson Laboratory (Bar Harbor, ME). The mice used for BBB and behavioral studies were 4 to 5 months old, both male and female. For atherosclerosis studies, P-selΔCTCT mice on C57BL/6J were crossed to apoE−/− mice on C57BL/6J to get P-selΔCTCT/apoE−/−. ApoE−/− controls were from The Jackson Laboratory. Mice were maintained on a 12-hour light/dark cycle and on chow diet for 4 months. Animals were bred and housed at the Immune Disease Institute, and all experimental procedures were approved by its Animal Care and Use Committee.

BBB permeability assay

The 2% Evans blue dye (EBD) in phosphate-buffered saline (PBS) was injected intraperitoneally (50 μg/g body weight). Three hours later, mice were killed; skin, spleen, and brain were harvested and placed in formamide for 72 hours (Sigma-Aldrich, St Louis, MO). EBD permeability was expressed as the optical density per gram of tissue (OD/g).

Localization of BBB permeability defects

To visualize the BBB damage, 2% EBD (50 μg/g body weight) was injected retroorbitally. After 6 hours, animals were anesthetized and perfused with 2000 kD fluorescein isothiocyanate (FITC)-dextran/PBS (Sigma-Aldrich) to outline the vasculature. Fluorescence in 50-μm frozen sections was visualized with a laser-scanning confocal microscope (Zeiss-Bio-Rad radiance 2000MP confocal/multiphoton system; Carl Zeiss, Gottingen, Germany). Four sagittal sections from each animal were used for the visualization of the EBD extravasation. Right and left hemispheres of the cerebrum were observed.

Middle cerebral artery occlusion model

We induced transient middle cerebral artery occlusion (MCAO) in males 10 to 12 weeks old. Mice were anesthetized with isoflurane in a mixture of 30% oxygen. A 7.0 silicon-precoated monofilament nylon suture was introduced into the internal carotid artery to occlude the right middle cerebral artery to produce transient focal cerebral ischemia. The filament was withdrawn after 90 minutes to allow reperfusion. A laser Doppler flowmeter (PERIMED System 5000, Stockholm, Sweden) was used to confirm induction of ischemia and reperfusion. Mouse body temperature (37° ± 1°C) was controlled throughout the experiment. Twenty-four hours after ischemia, the brains were removed, cut into 1-mm-thick coronal sections, and stained with 2% 2,3,5-triphenyl tetrazolium chloride (TTC). The infarct size in each section was determined with a computerized image analysis program (ImageJ software, National Institutes of Health, Bethesda, MD; http://rsbweb.nih.gov/ij/).

Social recognition test

Individual adult mice (4-5 months old, males and females) were tested in their home cage twice (I trial and II trial). Each trial began when a stimulus mouse (3-week-old juvenile mouse) was introduced into the home cage of 1 adult mouse. During the first exposure (I trial), a juvenile was exposed to the adult animal, and the duration of investigatory behavior of the adult toward the juvenile was recorded (Canon GL2 MiniDV digital camcorder; Canon, Kanagawa, Japan) on videotape for 2 minutes. The juvenile was then removed and returned to a holding cage. After 3 hours, the same juvenile was re-exposed to the same adult for 2 minutes (II trial). The duration of the investigatory behavior of the adult toward the juvenile was recorded and measured in both trials. Grooming, aggressive posturing, and sexual behaviors including mounting were not included in measures of investigation. Aggressive behavior such as chasing and attack was also analyzed. When an attack occurred, the juvenile was removed and the behavior of the adult mice was defined as aggressive.

Measurements of plasma levels of sP-selectin

Plasma levels of sP-selectin were measured by immunoassays (Quantikine; R&D Systems, Minneapolis, MN). Ethylenediaminetetraacetic acid (EDTA) was used as an anticoagulant, and plasma samples were diluted 1/50 before analysis.

Atherosclerotic lesion analysis

Mice were perfused with ice-cold PBS and 10% formalin. Hearts and aortas were carefully dissected, fixed in 10% formalin overnight, and infiltrated with 30% sucrose for 24 hours at 4°C. Hearts were embedded in optimal cooling temperature compound and sectioned (10 μm). Sections of the aortic sinus were stained with Oil Red-O and counterstained with hematoxylin. For each animal, 5 sections, 80 μm apart, were analyzed without knowledge of the genotype to determine atherosclerotic lesion size. Values represent the mean lesion area from 5 sections from each animal plus or minus SEM. The same sections were analyzed for the number of plaques, and the percentage of aortic sinus circumference covered by plaques was determined using a Zeiss Axioplan digital imaging microscope system with Zeiss AxioCam HRc camera and 10×/0.30 NA objective. AxioVision 4.6.3 software was used for image processing.

Quantification of plaque macrophage content

Macrophage staining in the aortic sinus was performed with a rat anti-mouse Mac-3 antibody (BD Biosciences, Franklin Lakes, NJ). Primary antibody was detected with a biotin-labeled anti-rat antibody (BD Biosciences). Sections were incubated with avidin-biotin complex reagent (Vectastain Elite ABC kit; Vector Laboratories, Burlingame, CA), and color was developed using diaminobenzidine reagent (Vector Laboratories). Four sections per mouse were examined under high magnification field microscopy, and the number of macrophages with a visible nucleus was counted. Values represent mean plus or minus SEM.

Plasma lipids and lipoprotein measurements

Blood samples were collected to heparinized Eppendorf tubes by retro-orbital venous plexus puncture after a 4-hour fast. Plasma was separated by centrifugation for analysis. Total cholesterol, unesterified cholesterol, triglycerides, and phospholipid levels were determined by using enzymatic colorimetric assays (Wako Chemicals, Richmond, VA). Lipoproteins were isolated from plasma by fast protein liquid chromatography (FPLC), as previously described.22 

Statistical analysis

The values are presented as mean plus or minus SEM. Differences between means were determined by using the Student 2-tailed t test when 2 groups were compared, and analysis of variance test followed by Bonferroni posttest. P values of .05 or less were regarded as statistically significant.

BBB permeability is increased in P-selΔCTCT mice

To determine the role of procoagulant state induced by increased levels of sP-selectin on the cerebrovasculature, we examined BBB permeability in the P-selΔCTCT mice. In this assay, EBD injected intraperitoneally enters the circulation, where it binds to albumin and therefore does not readily cross the BBB. Three hours after injection, the amounts of EBD in the blood, brain, spleen, and skin were measured. The amounts of EBD in the blood of wild-type (WT) and P-selΔCTCT mice were essentially identical (0.21 ± 0.04 and 0.23 ± 0.08 OD units/g, respectively, P = .6), suggesting that the mutant P-selectin did not alter EBD's transport to and gross clearance from the circulation. However, Figure 1A shows that EBD transfer into the cerebrum and cerebellum was significantly greater in P-selΔCTCT mice than in their WT littermate controls (2.7- and 1.4-fold, respectively). Additional analyses of the brains from these animals showed that all of the WT (17 of 17) and 19 of 31 of the P-selΔCTCT mice exhibited comparable, low levels of EBD leakage that were similar for the right and left cerebrum hemispheres (Figure 1B). Four P-selΔCTCT animals exhibited mild to substantial elevations of EBD leakage that were similar in both hemispheres. Strikingly, in 8 (25%) of the P-selΔCTCT animals, one hemisphere (left or right) exhibited substantially greater permeability than the other (indicated by ovals in Figure 1B). No statistical differences in EBD vascular permeability were observed in the skin (WT, 0.6 ± 0.04; P-selΔCTCT, 0.8 ± 0.1 OD/g, P = .1) or spleen (WT, 14 ± 0.5, P-selΔCTCT, 15 ± 0.7 OD/g, P = .1). These results suggest that in P-selΔCTCT mice there was an organ-specific increase in vascular permeability, and that it most likely resulted in an increased probability of events (breaks) in the BBB in some, but not all, animals at a particular point in time. Such events in brain vessels were not distributed evenly throughout, suggesting that they might be focal in nature.

Figure 1

Increased BBB permeability in P-selΔCTCT mice. (A) Quantification of EBD accumulation in the brain of P-selΔCTCT mice compared with WT. Data represent EBD extravasation calculated from A620 OD/g. n = 17 for WT and n = 31 for P-selΔCTCT mice. (B) Comparison of EBD accumulation between the right (RH, ▴) and left hemisphere (LH, ▵) in each animal. Ovals indicate the animals with a difference between RH and LH. (C) Representative images from confocal microscopy in the cortex of 4- to 5-month-old mice injected with EBD (red) 6 hours before perfusion with large dextran (green) at the time of sacrifice. Top panels show section of cerebrum from a WT; bottom panels are from P-selΔCTCT mice. Arrow points to the accumulation of EBD outside of the vessel in the left hemisphere of the P-selΔCTCT mice. Data represent mean ± SEM.

Figure 1

Increased BBB permeability in P-selΔCTCT mice. (A) Quantification of EBD accumulation in the brain of P-selΔCTCT mice compared with WT. Data represent EBD extravasation calculated from A620 OD/g. n = 17 for WT and n = 31 for P-selΔCTCT mice. (B) Comparison of EBD accumulation between the right (RH, ▴) and left hemisphere (LH, ▵) in each animal. Ovals indicate the animals with a difference between RH and LH. (C) Representative images from confocal microscopy in the cortex of 4- to 5-month-old mice injected with EBD (red) 6 hours before perfusion with large dextran (green) at the time of sacrifice. Top panels show section of cerebrum from a WT; bottom panels are from P-selΔCTCT mice. Arrow points to the accumulation of EBD outside of the vessel in the left hemisphere of the P-selΔCTCT mice. Data represent mean ± SEM.

Close modal

To evaluate further the BBB leakage at the microscopic level, mice were injected with EBD, and 6 hours later were perfused with FITC-labeled large dextran (2000 kDa). The large dextran does not permeate the blood vessels and was used to outline the vessels to separate them from the extravascular space of the brain. Using confocal microscopy of brain sections, we observed little or no EBD accumulation outside the vasculature (Figure 1C top panels) of all 5 WT mice. In contrast, in 4 of 6 P-selΔCTCT mice, there were large, uneven foci of EBD outside the vasculature, and in 2 P-selΔCTCT mice 1 hemisphere showed more leakage than the other hemisphere in the same animal (Figure 1C bottom panels). The uneven distribution of the BBB leakage most likely explains the differences in EBD levels between hemispheres of the P-selΔCTCT mice (Figure 1B).

To understand whether increased BBB leakage in P-selΔCTCT mice was due to inflammation or procoagulant state, we also evaluated Factor V Leiden (FVL) mice, a procoagulant mouse model.21  We injected EBD in these mice and measured the EBD levels in the cerebrum, skin, and spleen 3 hours later. Overall, EBD leakage in the cerebrum was 1.7-fold higher than in WT (P < .001, n = 9). Similarly to P-selΔCTCT mice, we have found that in 2 of 9 FVL mice, 1 hemisphere showed several-fold greater EBD leakage than the other. No significant increase was observed in EBD vascular permeability of the skin or spleen (data not shown). The similar BBB phenotype of the FVL to P-selΔCTCT mice supports our hypothesis that elevated procoagulant activity can induce breaks in the BBB that are not observed in WT mice.

Our results suggest that increased levels of sP-selectin in P-selΔCTCT mice have a negative effect on brain vasculature and may be implicated in the disruption of BBB.

P-selΔCTCT mice showed larger infarcts after transient MCAO

Because the results above might be a consequence of the propensity of the P-selΔCTCT mice to form small infarcts in the brain, we applied a stroke model in the P-selΔCTCT mice. Previously, P-selectin deficiency was shown to provide protection from stroke in the intraluminal MCAO model due to reduced neutrophil accumulation accompanied by increased postischemic cerebral reflow.23  In addition, high sP-selectin concentrations have been observed in patients with an acute ischemic stroke.24  To investigate the role of high sP-selectin in stroke, P-selΔCTCT and WT mice were subjected to MCAO for 90 minutes, followed by 22.5 hours of reperfusion. Doppler flowmetry was used in all mice to confirm ischemia and reperfusion. Only the mice that had blood flow less than 30% baseline of cerebral blood flow after MCAO and complete recovery after reperfusion were used. As shown in Figure 2A, infarction in the cortex and striatum was more severe in P-selΔCTCT mice than that observed in WT mice. The total infarct volume was significantly larger in the P-selΔCTCT (110 ± 9 mm3) than in WT (82 ± 9 mm3) mice (Figure 2C; P < .05). It is noteworthy that 1 of 9 P-selΔCTCT mice (Figure 2B arrow) had a spontaneous small infarct on the opposite side of the brain, the side that was not affected by the procedure. These results indicate that increased sP-selectin in P-selΔCTCT mice worsens the stroke outcome in the MCAO model of stroke.

Figure 2

Stroke infarct volume is increased in P-selΔCTCT mice. Intraluminal MCAO model of stroke on P-selΔCTCT mice and age-matched sibling WT controls. (A) Representative photographs of TTC staining of sections showing enhanced infarct volume in a P-selΔCTCT mouse brain, compared with a WT mouse brain. (B) White arrow indicates small spontaneous infarct present in the uninjured hemisphere of 1 P-selΔCTCT mouse. (C) Quantitative image analysis of the infarction area. n = 7 in each group. Error bars indicate SEM.

Figure 2

Stroke infarct volume is increased in P-selΔCTCT mice. Intraluminal MCAO model of stroke on P-selΔCTCT mice and age-matched sibling WT controls. (A) Representative photographs of TTC staining of sections showing enhanced infarct volume in a P-selΔCTCT mouse brain, compared with a WT mouse brain. (B) White arrow indicates small spontaneous infarct present in the uninjured hemisphere of 1 P-selΔCTCT mouse. (C) Quantitative image analysis of the infarction area. n = 7 in each group. Error bars indicate SEM.

Close modal

Decreased social investigation and increased aggression in P-selΔCTCT mice

Increased BBB permeability and susceptibility to stroke in the P-selΔCTCT mice suggest that procoagulant state and elevated sP-selectin caused changes in the brain vessels, which over time could impact brain function and thus affect the behavior of the mice. To evaluate this, we examined social behavior in 4- to 5-month-old animals. Each single adult mouse was kept in a home cage, and behavior trials began when a juvenile mouse was introduced. Investigatory behavior was recorded on videotape for 2 minutes (I trial). Three hours later, the same juvenile was introduced again with the same adult mouse, and behavior was recorded (II trial). Investigation time was defined as direct, active, olfactory exploration of the stimulus (juvenile mouse) by the subject (adult mouse) and consisted of nosing and sniffing of the head and anogenital regions, as well as close following of the juvenile. As shown in Figure 3A, P-selΔCTCT mice had severely impaired social behavior (reduced time of interaction) in both trials compared with their respective WT littermates.

Figure 3

Behavior of P-selΔCTCT mice in the social recognition test is severely impaired. (A) Total time(s) spent by adult mice investigating a juvenile. Males and females were analyzed separately in 2 trials (I and II). Error bars indicate SEM. (B) Aggressive behavior (adult mouse attacking juvenile) is shown as the percentage of mice that showed aggressive behavior toward the juvenile. n = 7-21 males; n = 4-8 females.

Figure 3

Behavior of P-selΔCTCT mice in the social recognition test is severely impaired. (A) Total time(s) spent by adult mice investigating a juvenile. Males and females were analyzed separately in 2 trials (I and II). Error bars indicate SEM. (B) Aggressive behavior (adult mouse attacking juvenile) is shown as the percentage of mice that showed aggressive behavior toward the juvenile. n = 7-21 males; n = 4-8 females.

Close modal

We also observed that adult P-selΔCTCT mice showed markedly increased aggressive behavior toward juveniles relative to their WT littermates. In P-selΔCTCT males, 40% of the animals showed aggressive behavior in the first trial and 75% in the second trial, whereas none of the WT mice showed aggression toward juveniles (Figure 3B). Similarly, in P-selΔCTCT females, aggressive behavior was observed in 50% of the animals in both trials compared with 0% in WT mice. These results suggest that increased levels of sP-selectin and procoagulant state in P-selΔCTCT mice can give rise to abnormal social behavior and increased aggression.

Enhanced atherosclerosis in P-selΔCTCT/apoE−/− mice

High levels of sP-selectin and hypercoagulability have been documented in a variety of cardiovascular disorders in humans, including atherosclerosis.25  Enhanced atherosclerosis has been also associated with procoagulant phenotype in FVL mice.21  To determine the effect of increased levels of sP-selectin in P-selΔCTCT mice on development of this inflammatory disease, we crossed P-selΔCTCT mice onto an apoE−/− background, known to be susceptible to atherosclerotic lesion development. To evaluate whether apoE deficiency influences sP-selectin plasma levels, we analyzed plasma samples from WT, P-selΔCTCT, P-sel+/+/apoE−/−, and P-selΔCTCT/apoE−/− mice (Table 1). Using ELISA, we found significantly increased sP-selectin levels in all groups compared with WT mice, and even observed slight elevation of sP-selectin in the apoE−/− genotype (Table 1). We then characterized levels of lipoproteins, known critical players in atherogenesis, in the plasma of P-sel+/+/apoE−/− and P-selΔCTCT/apoE−/− mice (Figure 4). We found that male, but not female, P-selΔCTCT/apoE−/− mice exhibited significantly greater plasma levels of total cholesterol, unesterified cholesterol, phospholipids, and triglycerides (Figure 4A). The lipoprotein cholesterol profiles suggest that much of the increase in cholesterol in male P-selΔCTCT/apoE−/− mice occurred in the intermediate density lipoprotein (IDL)/low density lipoprotein (LDL) and high density lipoprotein (HDL) size lipoprotein particles (Figure 4B). Body weights were not significantly different between the genotypes.

Table 1.

Comparison of sP-selectin levels in the plasma from WT, P-selΔCTCT, P-sel+/+/apoE−/−, and P-selΔCTCT/apoE−/− mice

GenotypensP-selectin, ng/mLP from WT
WT 12 127 ± 12  
P-selΔCTCT 1042 ± 68 < .0005 
P-sel+/+/apoE−/− 256 ± 37 < .03 
P-selΔCTCT/apoE−/− 13 1132 ± 83 < .0005 
GenotypensP-selectin, ng/mLP from WT
WT 12 127 ± 12  
P-selΔCTCT 1042 ± 68 < .0005 
P-sel+/+/apoE−/− 256 ± 37 < .03 
P-selΔCTCT/apoE−/− 13 1132 ± 83 < .0005 

Differences between groups were evaluated using analysis of variance test. Values are expressed as the mean ± SEM.

Figure 4

Plasma lipids and lipoprotein profiles in P-sel+/+/apoE−/− and P-selΔCTCT/apoE−/− mice. Plasma was collected from individual mice of both sexes, fasted for 4 hours. (A) Total cholesterol, unesterified cholesterol, phospholipid, and triglyceride levels in plasma from P-sel+/+/apoE−/− (■) and P-selΔCTCT/apoE−/− (□) mice. Data represent mean ± SEM (n = 6-9). Error bars indicate SEM. (B) Plasma samples from P-sel+/+/apoE−/− (●) and P-selΔCTCT/apoE−/− (○) individual animals were size fractionated by FPLC, and the total cholesterol content of each fraction was determined. The chromatograms are the average of multiple individually determined profiles (n = 4). Approximate elution positions of native very low density lipoprotein (VLDL), IDL/LDL, and HDL particles are indicated by brackets.

Figure 4

Plasma lipids and lipoprotein profiles in P-sel+/+/apoE−/− and P-selΔCTCT/apoE−/− mice. Plasma was collected from individual mice of both sexes, fasted for 4 hours. (A) Total cholesterol, unesterified cholesterol, phospholipid, and triglyceride levels in plasma from P-sel+/+/apoE−/− (■) and P-selΔCTCT/apoE−/− (□) mice. Data represent mean ± SEM (n = 6-9). Error bars indicate SEM. (B) Plasma samples from P-sel+/+/apoE−/− (●) and P-selΔCTCT/apoE−/− (○) individual animals were size fractionated by FPLC, and the total cholesterol content of each fraction was determined. The chromatograms are the average of multiple individually determined profiles (n = 4). Approximate elution positions of native very low density lipoprotein (VLDL), IDL/LDL, and HDL particles are indicated by brackets.

Close modal

Next, we determined the size of atherosclerotic lesions spontaneously produced in male and female 4-month-old mice fed a standard chow diet. We measured the areas of neutral lipid accumulation in cross-sections of the aortic sinus stained with Oil Red-O (Figure 5A,C). Compared with P-sel+/+/apoE−/− controls, there was a 2.6-fold increase in total aortic sinus lesion area in P-selΔCTCT/apoE−/− males (P < .05; Figure 5A left) and a 1.8-fold increase in females (P < .05; Figure 5A right). There was no significant difference in the number of plaques (data not shown) in the aortic sinus among the P-selΔCTCT/apoE−/− mice. However, the plaques in P-selΔCTCT/apoE−/− mice covered larger areas of the aortic sinus in both male and female groups compared with controls (P < .05; Figure 5B). To address whether macrophages/monocytes contribute to the increased atherosclerotic lesion burden, we quantified the macrophages in the lesions separately in male and female mice by immunohistochemistry. We observed increased macrophage numbers in the lesions of P-selΔCTCT/apoE−/− compared with P-sel+/+/apoE−/− (P < .05; Figure 5D).

Figure 5

Atherosclerosis is accelerated in P-selΔCTCT/apoE−/− mice compared with P-sel+/+/apoE−/−. Animals were maintained on normal chow, and their aortic sinus lesions were evaluated at 16 weeks of age. (A) Quantitative image analysis of aortic sinus lesion area. Male group showed 2.6-fold increased size of the plaque area. Female mice had 1.8-fold larger lesions than control mice. (B) Measurement of percentage of the aortic sinus circumference covered by atherosclerotic plaques. (C) Representative images of Oil Red-O staining of the aortic sinus. Representative images, showing an increase in plaque area in P-selΔCTCT/apoE−/− mice compared with P-sel+/+/apoE−/− mice (female mice). (D) Quantitative image analysis of macrophage content in the aortic sinus atherosclerotic plaques. Data represent mean ± SEM.

Figure 5

Atherosclerosis is accelerated in P-selΔCTCT/apoE−/− mice compared with P-sel+/+/apoE−/−. Animals were maintained on normal chow, and their aortic sinus lesions were evaluated at 16 weeks of age. (A) Quantitative image analysis of aortic sinus lesion area. Male group showed 2.6-fold increased size of the plaque area. Female mice had 1.8-fold larger lesions than control mice. (B) Measurement of percentage of the aortic sinus circumference covered by atherosclerotic plaques. (C) Representative images of Oil Red-O staining of the aortic sinus. Representative images, showing an increase in plaque area in P-selΔCTCT/apoE−/− mice compared with P-sel+/+/apoE−/− mice (female mice). (D) Quantitative image analysis of macrophage content in the aortic sinus atherosclerotic plaques. Data represent mean ± SEM.

Close modal

There are no previous reports indicating that sP-selectin influences plasma lipoprotein cholesterol levels. However, because both female and male P-selΔCTCT/apoE−/− mice exhibited greater atherosclerosis than P-sel+/+/apoE−/− controls and only males had elevated cholesterol (Figure 4A), it seems unlikely that altered levels of plasma lipoproteins were the only reason for the increased lesion size in P-selΔCTCT/apoE−/− mice. Thus, a procoagulant and proinflammatory state resulting from increased levels of sP-selectin may lead to increased monocyte recruitment and enhanced formation of early atherosclerotic lesions in both sexes.

In vitro studies have shown that P-selectin, even in its soluble form, activates leukocytes to produce both procoagulant26-28  and proinflammatory activities.15,29-32  The central hypothesis of our study was that this might occur also in vivo, in animals that shed P-selectin at an increased rate, impacting their susceptibility to vascular disease.

Elevated plasma sP-selectin causes BBB permeability and affects murine behavior

Elevated levels of sP-selectin and hypercoagulability have been found in many vascular diseases,33  but their pathologic role is unclear. In our study, we found increased vessel permeability in the brains of P-selΔCTCT mice, but not in their skin or spleen, suggesting that this vessel damage is specific to the barrier function and specialized junctions of the brain endothelium. One possible mechanism could be fibrin deposition on the brain vessels of P-selΔCTCT mice. Previously, it has been shown that P-selΔCTCT mice have increased fibrin deposition on injured mesenteric blood vessels.19  Fibrin has been shown to disrupt vascular endothelial cell junctions in vitro.34  Furthermore, fibrin binds to vascular endothelial (VE)–cadherin, an adhesive molecule present at adherence junctions of endothelial cells, which mediates cell-cell contacts and regulates paracellular permeability.35  In the brain, the tight and adherence junctions of the cerebral capillary endothelium form the highly restrictive BBB. Recently, fibrin has been shown to provoke and accelerate neurovascular damage and neuroinflammation in an Alzheimer disease (AD) mouse model.20  These mouse models of AD have deficits in vascular integrity,36  as shown by areas with increased BBB permeability that colocalized with fibrin deposition.20  Spontaneous deposition of fibrin has been shown in the tissue of homozygous FVL mice.21  In our experiments, we have found increased BBB leakage in the FVL mice, thus supporting the hypothesis that elevated procoagulant activity and fibrin deposition might be responsible for the increased BBB leakage in the P-selΔCTCT mice.

We observed a difference in the BBB permeability between each of the 2 hemispheres of some individual P-selΔCTCT mice (Figure 1B) and FVL mice, suggesting local and uneven BBB disruption (also detected by microscopy) and/or small infarctions. Such cerebrovascular lesions, termed silent brain infarction (SBI), have been found in patients with obstructive sleep apnea,37  a disease associated with increased serum levels of sP-selectin,38  and increased cerebrovascular and cardiovascular morbidity and mortality.39  Occurrence of stroke in these patients is most likely preceded by SBI. Interestingly, the level of sP-selectin was higher in patients with obstructive sleep apnea who exhibited SBI compared with those without SBI.38 

We also found differences in social behavior and a profound increase in aggression in P-selΔCTCT mice. P-selΔCTCT mice consistently showed impairment in social interaction with other mice. Changes in BBB permeability could have an effect on behavior. It is possible that impaired BBB function, caused by increased sP-selectin, resulted in the leakage of plasma proteins into the brain, causing injury and behavioral changes in P-selΔCTCT mice over time.

Previously, we have linked other risk factors for atherosclerosis in mice, such as increased homocysteine, apoE deficiency, and old age, with elevated BBB permeability.40-42  These risk factors also influence neurodegenerative diseases such as Alzheimer disease.43  Now we show that increased sP-selectin may play an important role in atherosclerosis and cerebrovascular function, and thus perhaps also in neurodegenerative diseases.

Increased sP-selectin intensifies the impact of experimental stroke

Plasma P-selectin has been shown to be elevated after stroke, and P-selectin deficiency and P-selectin antagonism protect from cerebral injury in models of MCAO.23,24,44  We found that increased levels of sP-selectin lead to an increased infarct volume in a mouse model of MCAO. The increase in infarct volume in P-selΔCTCT mice is most likely due to the increased procoagulant activity of these mice with elevated soluble tissue factor.19  This may result in excessive platelet activation, endothelial P-selectin expression, and fibrin formation. Pathologic coagulopathy and thrombus formation were shown to be involved in enhancement of ischemic cerebral injury.45-47  Our study supports the hypothesis that increased sP-selectin is not only a biomarker and predictor of stroke, but that it actually promotes stroke and worsens the stroke outcome.

Increased levels of sP-selectin accelerate atherosclerosis

A role for P-selectin in atherogenesis is supported by many studies.7,9,13,48,49  For example, P-selectin is up-regulated in the endothelium overlying the atherosclerotic plaque,48  P-selectin–deficient mice develop smaller, less complex atherosclerotic lesions,9  and anti-P-selectin antibodies inhibit monocyte rolling and attachment across the carotid endothelium.49  sP-selectin was shown to exhibit a consistent and independent association with atherosclerotic burden within a population of asymptomatic, nonsmoking, hypercholesterolemic men, suggesting that sP-selectin might be a useful biomarker of preclinical atherosclerosis.50  Our study is the first report demonstrating a causal role for increased levels of sP-selectin in the development of atherosclerosis. Adhesion and migration of inflammatory cells, in particular mononuclear cells, into the vascular wall are a hallmark of an atherosclerotic plaque. This process is fortified by the progressive deposition of lipids in the subendothelial layer.51  Presence of elevated sP-selectin in plasma of patients with peripheral arterial disease has been shown to induce the activation of leukocytes as detected by αMβ2 (Mac-1) activation, an integrin involved in leukocyte adhesion, and to enhance leukocyte adhesion on the fibrinogen or platelet monolayer in a shear-dependent manner.15  The signaling produced by sP-selectin binding to leukocyte PSGL-1 is similar to that produced by its membrane-bound form.15  Wang et al31  also found that sP-selectin partially restores impaired leukocyte adhesion and peritoneal accumulation in stimulated P-selectin–deficient mice. Therefore, sP-selectin can prime leukocyte integrin activation and leukocyte adhesion during inflammation,31  and this activity could explain the enhanced atherosclerotic lesion growth with elevated recruitment of monocytes to the lesion in the P-selΔCTCT/apoE−/− mice. In addition, elevated procoagulant activity was shown to promote atherosclerosis52,53  and may stimulate platelet activation that enhances lesion growth as well.54,55 

Our animal results are also in agreement with clinical studies showing that increased sP-selectin is associated with increased plasma LDL in hypercholesterolemic individuals.56  It is possible that cholesterol synthesis or absorption is affected by high levels of sP-selectin or by inflammation resulting from sP-selectin elevation. The molecular and cellular mechanisms explaining how elevated sP-selectin leads to an altered lipid profile in P-selΔCTCT mice remain to be clarified.

In conclusion, our studies demonstrate that sP-selectin has a profound negative impact on mouse vasculature. This results in a defect in BBB function and greater susceptibility to atherosclerosis. Similar to P-selectin on activated platelets,1  sP-selectin appears to promote the vicious circle of inflammation leading to endothelial dysfunction. In addition, the procoagulant activity of sP-selectin can aggravate the consequences of an ischemic event such as stroke. Therefore, elevated sP-selectin should be considered not only as a biomarker of inflammation/thrombosis, but also as an active player in these conditions. Targeting sP-selectin in disease states where it is elevated could reduce future cerebrovascular and cardiovascular events.

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 USC section 1734.

We thank Lesley Cowan for assistance in preparing the manuscript and Stephen Cifuni for technical assistance.

This work was supported by grants HL056949 (to D.D.W.) and HL066105 (to M.K. and D.D.W.) from the National Heart, Lung, and Blood Institute of the National Institutes of Health (Bethesda, MD).

National Institutes of Health

Contribution: J.K., A.K.C., and D.D.W. designed the study. J.K., A.K.C., B.-Q.Z., I.S.P., and A.Y. obtained data. J.K. wrote the paper. All authors were involved in the interpretation of the results and read, commented on, and approved the final version of the manuscript.

Conflict-of-interest disclosure: D.D.W. has consulted for Hoffmann-LaRoche and Archemix, and M.K. has consulted for Roche. The remaining authors declare no competing financial interests.

The current address for J.K. is Department of Biology, Massachusetts Institute of Technology, Cambridge, MA. The current address for A.K.C. is Department of Internal Medicine, Division of Hematology/Oncology, University of Iowa, Iowa City, IA.

Correspondence: Dr Denisa D. Wagner, Immune Disease Institute, 3 Blackfan Circle, 3rd Floor, Boston, MA 02115; e-mail: wagner@idi.harvard.edu.

1
Wagner
 
DD
Frenette
 
PS
The vessel wall and its interactions.
Blood
2008
, vol. 
111
 (pg. 
5271
-
5281
)
2
McEver
 
RP
Beckstead
 
JH
Moore
 
KL
Marshall-Carlson
 
L
Bainton
 
DF
GMP-140, a platelet α-granule membrane protein, is also synthesized by vascular endothelial cells and is localized in Weibel-Palade bodies.
J Clin Invest
1989
, vol. 
84
 (pg. 
92
-
99
)
3
Mayadas
 
TN
Johnson
 
RC
Rayburn
 
H
Hynes
 
RO
Wagner
 
DD
Leukocyte rolling and extravasation are severely compromised in P selectin-deficient mice.
Cell
1993
, vol. 
74
 (pg. 
541
-
554
)
4
Johnston
 
GI
Bliss
 
GA
Newman
 
PJ
McEver
 
RP
Structure of the human gene encoding granule membrane protein-140, a member of the selectin family of adhesion receptors for leukocytes.
J Biol Chem
1990
, vol. 
265
 (pg. 
21381
-
21385
)
5
McEver
 
RP
Cummings
 
RD
Role of PSGL-1 binding to selectins in leukocyte recruitment.
J Clin Invest
1997
, vol. 
100
 (pg. 
97
-
103
)
6
Disdier
 
M
Morrissey
 
JH
Fugate
 
RD
Bainton
 
DF
McEver
 
RP
Cytoplasmic domain of P-selectin (CD62) contains the signal for sorting into the regulated secretory pathway.
Mol Biol Cell
1992
, vol. 
3
 (pg. 
309
-
321
)
7
Wagner
 
DD
Burger
 
PC
Platelets in inflammation and thrombosis.
Arterioscler Thromb Vasc Biol
2003
, vol. 
23
 (pg. 
2131
-
2137
)
8
Subramaniam
 
M
Frenette
 
PS
Saffaripour
 
S
Johnson
 
RC
Hynes
 
RO
Wagner
 
DD
Defects in hemostasis in P-selectin-deficient mice.
Blood
1996
, vol. 
87
 (pg. 
1238
-
1242
)
9
Dong
 
ZM
Brown
 
AA
Wagner
 
DD
Prominent role of P-selectin in the development of advanced atherosclerosis in ApoE-deficient mice.
Circulation
2000
, vol. 
101
 (pg. 
2290
-
2295
)
10
Ishiwata
 
N
Takio
 
K
Katayama
 
M
, et al. 
Alternatively spliced isoform of P-selectin is present in vivo as a soluble molecule.
J Biol Chem
1994
, vol. 
269
 (pg. 
23708
-
23715
)
11
Sampietro
 
T
Tuoni
 
M
Ferdeghini
 
M
, et al. 
Plasma cholesterol regulates soluble cell adhesion molecule expression in familial hypercholesterolemia.
Circulation
1997
, vol. 
96
 (pg. 
1381
-
1385
)
12
Lip
 
GY
Blann
 
AD
Zarifis
 
J
Beevers
 
M
Lip
 
PL
Beevers
 
DG
Soluble adhesion molecule P-selectin and endothelial dysfunction in essential hypertension: implications for atherogenesis? A preliminary report.
J Hypertens
1995
, vol. 
13
 (pg. 
1674
-
1678
)
13
Frijns
 
CJ
Kappelle
 
LJ
van Gijn
 
J
Nieuwenhuis
 
HK
Sixma
 
JJ
Fijnheer
 
R
Soluble adhesion molecules reflect endothelial cell activation in ischemic stroke and in carotid atherosclerosis.
Stroke
1997
, vol. 
28
 (pg. 
2214
-
2218
)
14
Blann
 
AD
Lip
 
GY
Beevers
 
DG
McCollum
 
CN
Soluble P-selectin in atherosclerosis: a comparison with endothelial cell and platelet markers.
Thromb Haemostasis
1997
, vol. 
77
 (pg. 
1077
-
1080
)
15
Woollard
 
KJ
Kling
 
D
Kulkarni
 
S
Dart
 
AM
Jackson
 
S
Chin-Dusting
 
J
Raised plasma soluble P-selectin in peripheral arterial occlusive disease enhances leukocyte adhesion.
Circ Res
2006
, vol. 
98
 (pg. 
149
-
156
)
16
Ishiwata
 
S
Tukada
 
T
Nakanishi
 
S
Nishiyama
 
S
Seki
 
A
Postangioplasty restenosis: platelet activation and the coagulation-fibrinolysis system as possible factors in the pathogenesis of restenosis.
Am Heart J
1997
, vol. 
133
 (pg. 
387
-
392
)
17
Ridker
 
PM
Buring
 
JE
Rifai
 
N
Soluble P-selectin and the risk of future cardiovascular events.
Circulation
2001
, vol. 
103
 (pg. 
491
-
495
)
18
Hartwell
 
DW
Mayadas
 
TN
Berger
 
G
, et al. 
Role of P-selectin cytoplasmic domain in granular targeting in vivo and in early inflammatory responses.
J Cell Biol
1998
, vol. 
143
 (pg. 
1129
-
1141
)
19
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
2000
, vol. 
97
 (pg. 
13835
-
13840
)
20
Paul
 
J
Strickland
 
S
Melchor
 
JP
Fibrin deposition accelerates neurovascular damage and neuroinflammation in mouse models of Alzheimer's disease.
J Exp Med
2007
, vol. 
204
 (pg. 
1999
-
2008
)
21
Cui
 
J
Eitzman
 
DT
Westrick
 
RJ
, et al. 
Spontaneous thrombosis in mice carrying the Factor V Leiden mutation.
Blood
2000
, vol. 
96
 (pg. 
4222
-
4226
)
22
Rigotti
 
A
Trigatti
 
BL
Penman
 
M
Rayburn
 
H
Herz
 
J
Krieger
 
M
A targeted mutation in the murine gene encoding the high density lipoprotein (HDL) receptor scavenger receptor class B type I reveals its key role in HDL metabolism.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 (pg. 
12610
-
12615
)
23
Connolly
 
ES
Winfree
 
CJ
Prestigiacomo
 
CJ
, et al. 
Exacerbation of cerebral injury in mice that express the P-selectin gene: identification of P-selectin blockade as a new target for the treatment of stroke.
Circ Res
1997
, vol. 
81
 (pg. 
304
-
310
)
24
Nadar
 
SK
Lip
 
GY
Blann
 
AD
Platelet morphology, soluble P selectin and platelet P-selectin in acute ischaemic stroke: the West Birmingham Stroke Project.
Thromb Haemostasis
2004
, vol. 
92
 (pg. 
1342
-
1348
)
25
Cambien
 
B
Wagner
 
DD
A new role in hemostasis for the adhesion receptor P-selectin.
Trends Mol Med
2004
, vol. 
10
 (pg. 
179
-
186
)
26
Celi
 
A
Pellegrini
 
G
Lorenzet
 
R
, et al. 
P-selectin induces the expression of tissue factor on monocytes.
Proc Natl Acad Sci U S A
1994
, vol. 
91
 (pg. 
8767
-
8771
)
27
Hrachovinova
 
I
Cambien
 
B
Hafezi-Moghadam
 
A
, et al. 
Interaction of P-selectin and PSGL-1 generates microparticles that correct hemostasis in a mouse model of hemophilia A.
Nat Med
2003
, vol. 
9
 (pg. 
1020
-
1025
)
28
del Conde
 
I
Nabi
 
F
Tonda
 
R
Thiagarajan
 
P
López
 
JA
Kleiman
 
NS
Effect of P-selectin on phosphatidylserine exposure and surface-dependent thrombin generation on monocytes.
Arterioscler Thromb Vasc Biol
2005
, vol. 
5
 (pg. 
1065
-
1070
)
29
Damle
 
NK
Klussman
 
K
Dietsch
 
MT
, et al. 
GMP-140 (P-selectin/CD62) binds to chronically stimulated but not resting CD4+ T lymphocytes and regulates their production of proinflammatory cytokines.
Eur J Immunol
1992
, vol. 
22
 (pg. 
1789
-
1793
)
30
Evangelista
 
V
Manarini
 
S
Sideri
 
R
, et al. 
Platelet/Polymorphonuclear leukocyte interaction: P-selectin triggers protein-tyrosine phosphorylation-dependent CD11b/CD18 adhesion: role of PSGL-1 as a signaling molecule.
Blood
1999
, vol. 
93
 (pg. 
876
-
885
)
31
Wang
 
HB
Wang
 
JT
Zhang
 
L
, et al. 
P-selectin primes leukocyte integrin activation during inflammation.
Nat Immunol
2007
, vol. 
8
 (pg. 
882
-
892
)
32
Woollard
 
KJ
Suhartoyo
 
A
Harris
 
EE
, et al. 
Pathophysiological levels of soluble P-selectin mediate adhesion of leukocytes to the endothelium through Mac-1 activation.
Circ Res
2008
, vol. 
103
 (pg. 
1128
-
1138
)
33
Woollard
 
KJ
Soluble bio-markers in vascular disease: much more than gauges of disease?
Clin Exp Pharmacol Physiol
2005
, vol. 
32
 (pg. 
233
-
240
)
34
Kadish
 
JL
Butterfield
 
CE
Folkman
 
J
The effect of fibrin on cultured vascular endothelial cells.
Tissue Cell
1979
, vol. 
11
 (pg. 
99
-
108
)
35
Bach
 
TL
Barsigian
 
C
Yaen
 
CH
Martinez
 
J
Endothelial cell VE-cadherin functions as a receptor for the beta15-42 sequence of fibrin.
J Biol Chem
1998
, vol. 
273
 (pg. 
30719
-
30728
)
36
Ujiie
 
M
Dickstein
 
DL
Carlow
 
DA
Jefferies
 
WA
Blood-brain barrier permeability precedes senile plaque formation in an Alzheimer disease model.
Microcirculation
2003
, vol. 
10
 (pg. 
463
-
470
)
37
Minoguchi
 
K
Yokoe
 
T
Tazaki
 
T
, et al. 
Silent brain infarction and platelet activation in obstructive sleep apnea.
Am J Respir Crit Care Med
2007
, vol. 
175
 (pg. 
612
-
617
)
38
Robinson
 
GV
Pepperell
 
JC
Segal
 
HC
Davies
 
RJ
Stradling
 
JR
Circulating cardiovascular risk factors in obstructive sleep apnea: data from randomized controlled trials.
Thorax
2004
, vol. 
59
 (pg. 
777
-
782
)
39
Yaggi
 
HK
Concato
 
J
Kernan
 
WN
Lichtman
 
JH
Brass
 
LM
Mohsenin
 
V
Obstructive sleep apnea as a risk factor for stroke and death.
N Engl J Med
2005
, vol. 
353
 (pg. 
2034
-
2041
)
40
Kamath
 
AF
Chauhan
 
AK
Kisucka
 
J
, et al. 
Elevated levels of homocysteine compromise blood-brain barrier integrity in mice.
Blood
2006
, vol. 
107
 (pg. 
591
-
593
)
41
Methia
 
N
Andre
 
P
Hafezi-Moghadam
 
A
Economopoulos
 
M
Thomas
 
KL
Wagner
 
DD
ApoE deficiency compromises the blood brain barrier especially after injury.
Mol Med
2001
, vol. 
7
 (pg. 
810
-
815
)
42
Hafezi-Moghadam
 
A
Thomas
 
KL
Wagner
 
DD
ApoE deficiency leads to a progressive age-dependent blood-brain barrier leakage.
Am J Physiol
2007
, vol. 
292
 (pg. 
1256
-
1262
)
43
Anello
 
G
Gueant-Rodriguez
 
RM
Bosco
 
P
, et al. 
Homocysteine and methylenetetrahydrofolate reductase polymorphism in Alzheimer's disease.
NeuroReport
2004
, vol. 
15
 (pg. 
859
-
861
)
44
Zhang
 
R
Chopp
 
M
Zhang
 
Z
Jiang
 
N
Powers
 
C
The expression of P- and E-selectins in three models of middle cerebral artery occlusion.
Brain Res
1998
, vol. 
785
 (pg. 
207
-
214
)
45
Barnes
 
C
Deveber
 
G
Prothrombotic abnormalities in childhood ischemic stroke.
Thromb Res
2006
, vol. 
118
 (pg. 
67
-
74
)
46
Mocco
 
J
Choudhri
 
T
Huang
 
J
, et al. 
HuEP5C7 as a humanized monoclonal anti-E/P-selectin neurovascular protective strategy in a blinded placebo-controlled trial of nonhuman primate stroke.
Circ Res
2002
, vol. 
91
 (pg. 
907
-
914
)
47
Kleinschnitz
 
C
Pozgajova
 
M
Pham
 
M
Bendszus
 
M
Nieswandt
 
B
Stoll
 
G
Targeting platelets in acute experimental stroke: impact of glycoprotein Ib, VI, and IIb/IIIa blockade on infarct size, functional outcome, and intracranial bleeding.
Circulation
2007
, vol. 
115
 (pg. 
2323
-
2330
)
48
Johnson-Tidey
 
RR
McGregor
 
JL
Taylor
 
PR
Poston
 
RN
Increase in the adhesion molecule P-selectin in endothelium overlying atherosclerotic plaques: coexpression with intercellular adhesion molecule-1.
Am J Pathol
1994
, vol. 
144
 (pg. 
952
-
961
)
49
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
1999
, vol. 
84
 (pg. 
1237
-
1244
)
50
Chironi
 
G
Dosquet
 
C
Del-Pino
 
M
, et al. 
Relationship of circulating biomarkers of inflammation and hemostasis with preclinical atherosclerotic burden in nonsmoking hypercholesterolemic men.
Am J Hypertens
2006
, vol. 
19
 (pg. 
1025
-
1031
)
51
Libby
 
P
Fat fuels the flame: triglyceride-rich lipoproteins and arterial inflammation.
Circ Res
2007
, vol. 
100
 (pg. 
299
-
301
)
52
Eitzman
 
DT
Westrick
 
RJ
Bi
 
X
, et al. 
Homozygosity for Factor V Leiden leads to enhanced thrombosis and atherosclerosis in mice.
Circulation
2005
, vol. 
111
 (pg. 
1822
-
1825
)
53
Westrick
 
RJ
Bodary
 
PF
Xu
 
Z
Shen
 
YC
Broze
 
GJ
Eitzman
 
DT
Deficiency of tissue factor pathway inhibitor promotes atherosclerosis and thrombosis in mice.
Circulation
2001
, vol. 
103
 (pg. 
3044
-
3046
)
54
Burger
 
PC
Wagner
 
DD
Platelet P-selectin facilitates atherosclerotic lesion development.
Blood
2003
, vol. 
101
 (pg. 
2661
-
2666
)
55
Huo
 
Y
Schober
 
A
Forlow
 
SB
, et al. 
Circulating activated platelets exacerbate atherosclerosis in mice deficient in apolipoprotein E.
Nat Med
2003
, vol. 
9
 (pg. 
61
-
67
)
56
Davi
 
G
Romano
 
M
Mezzetti
 
A
, et al. 
Increased levels of soluble P-selectin in hypercholesterolemic patients.
Circulation
1998
, vol. 
97
 (pg. 
953
-
957
)
Sign in via your Institution