Platelet/endothelial cell adhesion molecule-1 (PECAM-1) is a 130-kD member of the Ig gene superfamily that is expressed on the surface of circulating platelets, monocytes, neutrophils, and selective T-cell subsets. It is also a major component of the endothelial cell intercellular junction. Previous studies have shown that cross-linking PECAM-1 on the surface of leukocytes results in the activation of adhesion molecules of both the β1 and β2integrin family. In addition, the process of leukocyte transendothelial migration appears to be mediated, at least in part, by homophilic adhesive interactions that take place between leukocyte and endothelial cell junctional PECAM-1 molecules. However, little is known about the functional role of this membrane glycoprotein in human platelets. In the present study, we examined the effects of PECAM-1 engagement on integrin-mediated platelet-extracellular matrix or platelet-platelet interactions. Bivalent, but not monovalent, anti–PECAM-1 monoclonal antibodies (MoAbs) specific for membrane-proximal Ig-homology domain 6 significantly augmented platelet deposition (increased surface coverage) and aggregation (increased average size) onto extracellular matrix, under both oscillatory or defined low shear flow conditions (200 s−1) in a modified cone and plate viscometer. Moreover, bivalent anti-domain 6 MoAbs were capable of serving as costimulatory agonists to markedly enhance both adenosine diphosphate (ADP)- and platelet activating factor (PAF)-induced platelet aggregation responses. These antibodies appeared to act via outside-in signal transduction through PECAM-1, as evidenced by the fact that their binding (1) led to conformational changes in the αIIbβ3 integrin complex, (2) induced surface expression of P-selectin, and (3) resulted in the tyrosine phosphorylation of PECAM-1. Together, these data support a role for PECAM-1 in cellular activation and suggest that PECAM-1 may serve as a costimulatory agonist receptor capable of modulating integrin function in human platelets during adhesion and aggregation.

PLATELET/ENDOTHELIAL cell adhesion molecule-1 (PECAM-1) is a 130-kD transmembrane glycoprotein that is expressed on the surface of circulating platelets, monocytes, neutrophils, and selected T-cell subsets. It is also a major constituent of the endothelial cell intercellular junction, where up to 106 PECAM-1 molecules concentrate after the formation of cell-cell contact (for a review, see Newman1). The 574 amino acid extracellular domain of PECAM-1 is organized into six Ig-like homology units,2 followed by a single-pass transmembrane domain, and a 118 amino acid cytoplasmic tail that contains specific sites for palmitoylation, phosphorylation, and assembly of cytosolic signaling molecules.3-6 Approximately 30% of the molecular mass of PECAM-1 is composed of carbohydrate residues whose influence on the adhesive properties of PECAM-1 is as yet unknown.

A great deal has been learned in the past several few years about the participation of PECAM-1 in the process of leukocyte transendothelial migration. The expression and/or distribution of PECAM-1 on the cell surface has been shown to be modulated in transmigrating leukocytes7-9 and on endothelial cells exposed to inflammatory cytokines.9-13 Moreover, Zocchi et al14 have shown that PECAM-1– but not ICAM-1–transfected fibroblasts support chemokine-independent transmigration of activated T lymphocytes.14 Finally, anti–PECAM-1 antibodies have been shown to inhibit leukocyte migration across an endothelial cell barrier both in vitro15 and in vivo,16-18 and the potential clinical relevance of interfering with PECAM-1 function has recently been shown in two different animal models of cardiac ischemia/reperfusion injury.19,20 

The precise mechanism by which PECAM-1 mediates cell-cell interactions is not clear. Several studies have shown that PECAM-1 is capable of interacting with other PECAM-1 molecules expressed on the cell surface21-23; a homophilic process that is mediated by N-terminal Ig-homology domains 1 and 2.22,23 The integrin αvβ324,25 and a yet to be characterized 120-kD glycoprotein found on activated T lymphocytes26 have also been implicated as heterophilic counter receptors for PECAM-1. However, in addition to its intrinsic adhesive properties, a number of investigators have shown that engagement of PECAM-1 can upregulate the function of adhesion receptors other than PECAM-1, most notably members of the integrin family. Tanaka et al27 provided the first experimental evidence that PECAM-1 might be involved in transducing signals to other adhesion receptors. They showed that cross-linking of PECAM-1 on the surface of selected T-lymphocyte subsets resulted in the upregulation of β1 integrin function. T cells treated with certain bivalent anti–PECAM-1 monoclonal antibodies (MoAbs) exhibited increased adherence to plastic wells coated with fibronectin (via α5β1) or vascular cell adhesion molecule-1 (VCAM-1; via α4β1). Additional cross-linking of the anti–PECAM-1 MoAb using goat antimouse IgG further augmented adhesion, but monovalent Fab fragments were ineffective, suggesting that dimerization of PECAM-1 on the cell surface might be required for integrin affinity modulation. Similarly, Leavesley et al28 reported that the binding of a bivalent anti–PECAM-1 MoAb to CD34+ hematopoietic progenitor cells enhanced their adhesion to VCAM-1–transfected CHO cells,28a process presumably mediated by α4β1. Modulation of integrin affinity does not appear to be limited to β1 integrins, because several groups have reported that ligation of PECAM-1 also leads to upregulation of β2integrin function in lymphokine-activated killer cells,29monocytes and neutrophils,30 and natural killer cells.31 Together, these data suggest a mechanism by which dimerization or oligomerization of PECAM-1 on the cell surface results in the generation of specific signals that are capable of modulating integrin affinity.

Human platelets offer an attractive model system to further examine the relationship between PECAM-1 engagement and cellular activation. Platelets normally exist in a resting, nonadhesive state, but can be stimulated by multiple agonists, under well-controlled conditions, to undergo a series of easily measurable cell biologic and biochemical changes, including protein-tyrosine phosphorylation, α-granule release, cell-extracellular matrix interactions, and cell-cell interactions (platelet aggregation). Moreover, platelets coexpress approximately 10,000 copies of PECAM-132,33 and 80,000 copies of the well-characterized integrin, αIIbβ3.34 Finally, there are specific MoAbs that are capable of detecting subtle conformational changes in this integrin that report its conformational (affinity) state, as well as MoAbs that specifically and sensitively measured the exposure of the α-granule-specific membrane protein, P-selectin, on the platelet surface. The purpose of the present investigation, therefore, was to test the hypothesis that PECAM-1 could serve as a costimulatory agonist receptor whose engagement modulates downstream cellular responses, including platelet adhesion, platelet aggregation, and integrin affinity.

MoAbs.

Well-characterized35,36 domain-specific MoAbs used in this study included PECAM-1.3 (directed against Ig-homology domain 1), PECAM-1.1 (domain 5), PECAM-1.2 (domain 6), and 4G6 (domain 6; kindly provided by Dr Steven Albelda, University of Pennsylvania School of Medicine, Philadelphia, PA). IV.3, a blocking MoAb specific for the human FcγIIa receptor,37 was kindly provided by Dr Clark Anderson (Ohio State University, Columbus, OH). The ligand-induced binding site (LIBS) antibody, D3, which specifically recognizes the active conformation of αIIbβ3,38 was kindly provided by Dr Lisa Jennings (University of Tennessee, Memphis, TN). The anti–P-selectin MoAb, S12,39 was kindly provided by Dr Rodger McEver (University of Oklahoma, Oklahoma City, OK). F(ab′)2 and Fab fragments were generated using immobilized pepsin or papain, respectively, according to the manufacturer's (Pierce, Rockford, IL) instructions. After overnight dialysis in phosphate-buffered saline (PBS), pH 7.4, all fragments were carefully analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) under both reducing and nonreducing conditions to ensure that no intact IgG remained. Before their use, the reactivity of all PECAM-1–specific antibodies was confirmed by enzyme-linked immunosorbent assay against an immobilized recombinant protein containing the complete extracellular domain of human PECAM-1. The functional integrity of IV.3 Fab fragments was determined by measuring their ability to block FcγIIa receptor-mediated platelet activation induced by human heparin-induced thrombocytopenia (HITP) antibodies.

Platelet deposition on extracellular matrix (ECM).

ECM-coated tissue culture wells were prepared according to the method of Gospodarowicz et al.40 Briefly, bovine corneal endothelial cells were grown to confluence in 16-mm diameter tissue culture plates (Nunc, Roskilde, Denmark), washed with PBS, pH 7.4, and dissolved by exposure to 0.5% Triton X-100 and 0.1 mol/L NH4OH, followed by extensive washing with distilled water. Two hundred fifty microliters of citrated whole blood was preincubated with the indicated antibodies for 15 minutes at 37°C, and the mixture was added to the ECM-coated plates, which were then inserted into a modified rotating Teflon cone and plate viscometer described in detail by Varon et al.41 The platelets were then subjected to either low (200 s−1) or high (1,300 s−1) shear rates for 2 minutes, thoroughly washed with PBS, fixed, and stained by May-Grünwald stain. Platelet interactions with extracellular matrix and with each other were analyzed using an inverted Olympus light microscope (Olympus Corp, Lake Success, NY). The image was captured by a video camera, digitized, and quantitated using a computer-assisted image analysis system.41 Data are expressed as the percentage of surface coverage, the average size of the aggregate, and the total object number. Statistical analysis of the data (mean ± SD) was performed using a paired Student's t-test for single measurements or repeated measures of variance for a series of measurements. Pvalues less than .05 were considered significant.

Platelet aggregation.

Blood from healthy, nonaspirinated volunteer donors was collected into plastic tubes containing 3.8% (wt/vol) trisodium citrate (9:1) or acid citrate dextrose, pH 4.6 (9:1) and mixed gently by inversion. Platelet-rich plasma (PRP) was prepared by centrifugation of the blood at 140g for 15 minutes and collecting the upper layer. Platelet-poor plasma (PPP) was prepared by centrifugation of the remaining lower layer at 2,000g for 10 minutes. The concentration of platelets in PRP was adjusted with PPP to 2 × 108/mL. Aggregation studies were performed using a four channel platelet aggregometer (PAP-4 Bio-Data, Horsham, PA). Preliminary studies showed that the anti–PECAM-1 MoAbs, by themselves, failed to induce platelet aggregation (not shown), similar to previous reports of the functional effects of other anti–PECAM-1 MoAbs.32,33,42 All aggregation studies were performed in duplicate on at least three separate occasions.

Flow cytometry.

Platelets were washed twice in Ringer's citrate dextrose (RCD; 108 mmol/L NaCl, 38 mmol/L KCl, 1.7 mmol/L NaHCO3, 21.2 mmol/L Na citrate, 27.8 mmol/L glucose, and 1.1 mmol/L MgCl2, pH 6.5) containing 50 ng/mL prostaglandin E1(PGE1), diluted to a final concentration of 5 × 106/mL, and then incubated with various agonists in a microtiter well for 1 hour at room temperature in the presence of 2 mmol/L CaCl2. Control agonists included buffer alone, normal mouse IgG1 (NM IgG), 5 mmol/L RGEW peptide, or 5 mmol/L RGDW peptide. Antibody agonists were used at 10 μg/mL final concentration and included PECAM-1.3, PECAM-1.1, PECAM-1.2, and 4G6. All antibodies were used as either intact IgGs, F(ab′)2 fragments, or monovalent Fab fragments. Platelets were washed, incubated with fluorescein isothiocyanate (FITC)-conjugated D3 or S12 for 30 minutes at room temperature, washed once more, transferred to 450 μL of RCD, pH 7.4, and analyzed on a FACScan (Becton Dickinson, San Jose, CA). At least 5,000 platelets per sample were examined. Fluorescence data were displayed as logarithmic contour plots, dot plots, or logarithmic histograms using LYSYS II software (Becton Dickinson). Mean fluorescence intensity of S12 or D3 binding (minus background fluorescence observed in the presence of buffer alone) from at least three separate experiments (±SD) was then plotted versus the agonist used.

Immunoprecipitation and antiphosphotyrosine immunoblots.

Washed human platelets (1 × 109/mL) were incubated under stirring conditions with buffer, 7 μmol/L thrombin receptor-activating peptide (TRAP; having the amino acid sequence SFLLRN), NM IgG1 F(ab′)2, or PECAM-1.2 F(ab′)2 and then lysed for 1 hour at 4°C in an equal volume of 2% Triton X-100, 10 mmol/L EGTA, 15 mmol/L HEPES, 145 mmol/L NaCl, 1 mmol/L phenylmethylsulfonyl fluoride, 20 μg/mL leupeptin, and 2 mmol/L sodium orthovanadate, pH 7.4. After centrifugation at 14,000 rpm for 5 minutes at 4°C, clarified supernatants were subjected to immunoprecipitation using 10 μg of NM IgG1 or PECAM-1.3. Immune complexes were collected on protein G Sepharose beads (Pharmacia Biotech AB, Uppsala, Sweden), resolved on a 12.5% SDS-polyacrylamide gel, and transferred electrophoretically to Immobilon polyvinylidene fluoride membrane (Millipore, Bedford, MA). Detection of tyrosine-phosphorylated PECAM-1 using the horseradish peroxidase (HRP)-conjugated antiphosphotyrosine MoAb, PY-20, was performed as previously described.43 

Engagement of PECAM-1 Ig-domain 6 promotes platelet-extracellular matrix and platelet-platelet interactions.

To investigate the effects of PECAM-1 engagement on the process of platelet adhesion and spreading, citrated whole blood was exposed to extracellular matrix in a modified cone and plate viscometer at a shear rate of 200 s−1, as previously described.41 Under these conditions, platelets adhere to the matrix but do not form platelet aggregates. Application of higher shear rate of 1,300 s−1 or the addition of other agonists is accompanied by more extensive adhesion as well as the formation of platelet aggregates. As shown in Fig 1, preincubation of platelets with the anti–PECAM-1 Ig-domain 6-specific antibody, PECAM-1.2, before their exposure to extracellular matrix coated surfaces increased by nearly 50% both surface coverage (from 14.47 ± 3.84 to 22.50 ± 5.66,P < .017) and the average size of the platelet aggregates (from 26.10 ± 2.05 to 38.42 ± 7.12, P < .002). The MoAb 4G6, which also maps to PECAM-1 Ig-domain 6,35 showed similar effects (not shown). NM IgG1 or the anti–PECAM-1 Ig-domain 1-specific MoAb, PECAM-1.3, failed to augment platelet deposition on extracellular matrix, suggesting that this stimulatory effect was transmitted into the cell in a PECAM-1 domain-specific manner. In addition, PECAM-1.2 significantly enhanced both adenosine diphosphate (ADP)- and platelet activating factor (PAF)-induced platelet-platelet interactions, as measured by platelet aggregometry using platelet-rich plasma (Fig 2A). This effect was not due to activation of the platelet FcγIIa receptor, because preincubation of platelets with saturating amounts of a blocking antibody to FcγRII (IV.3) did not abolish the costimulatory effect of PECAM-1.2 (Fig 2B). The effect of PECAM-1.2 on both ADP- (2 μmol/L) and PAF- (40 nmol/L) induced activation of platelets was dose-dependent at PECAM-1.2 concentrations ranging from 0.2 to 5 μg/mL (Fig 2C and D). Together, these data suggest that engagement of PECAM-1 through Ig domain 6 lowers the threshold for platelet stimulation by a variety of agonists, including ADP, PAF, and ECM under conditions of shear.

Fig. 1.

Anti–PECAM-1 antibodies promote platelet deposition on extracellular matrix. Two hundred fifty microliters of citrated whole blood was preincubated for 15 minutes at 37°C with either buffer alone or 5 μg/mL of normal mouse IgG1, PECAM-1.3, or PECAM-1.2. After 8 minutes of exposure to ECM-covered plates under conditions of low shear (200 s−1), samples were washed and stained and adherent platelets and aggregates were evaluated by image analysis as described in the Materials and Methods. Data are expressed as the percentage of ECM coverage (A) as well as average size (in square micrometers) of the aggregates formed (B). Note that both parameters were significantly increased (P = .017 andP = .02, respectively) in the presence of PECAM-1.2, but not PECAM-1.3.

Fig. 1.

Anti–PECAM-1 antibodies promote platelet deposition on extracellular matrix. Two hundred fifty microliters of citrated whole blood was preincubated for 15 minutes at 37°C with either buffer alone or 5 μg/mL of normal mouse IgG1, PECAM-1.3, or PECAM-1.2. After 8 minutes of exposure to ECM-covered plates under conditions of low shear (200 s−1), samples were washed and stained and adherent platelets and aggregates were evaluated by image analysis as described in the Materials and Methods. Data are expressed as the percentage of ECM coverage (A) as well as average size (in square micrometers) of the aggregates formed (B). Note that both parameters were significantly increased (P = .017 andP = .02, respectively) in the presence of PECAM-1.2, but not PECAM-1.3.

Close modal
Fig. 2.

Anti–PECAM-1 MoAbs act as costimulatory agonists in ADP- and PAF-induced platelet aggregation. PRP was preincubated with (1) buffer, (2) normal mouse IgG1, (3) PECAM-1.3, or (4) PECAM-1.2. All antibodies were used at a final concentration of 5 μg/mL. Note that PECAM-1.2 augmented low-dose (1.25 μmol/L) ADP-induced platelet aggregation, both in the absence (A) and presence (B) of the MoAb IV.3 (a blocking MoAb specific for FcγRIIa). The degree of potentiation of aggregation induced by PECAM-1.2 varied somewhat from experiment to experiment (compare [A] with [B]), and this variation was not attributable to FcγRIIa blockade. Incubation of PRP with increasing concentrations of PECAM-1.2 (shown in micrograms per milliliter), followed by the addition of either 2 μmol/L ADP (C) or 40 nmol/L PAF (D), showed that the effects of antibody-mediated dimerization of PECAM-1 are dose-dependent.

Fig. 2.

Anti–PECAM-1 MoAbs act as costimulatory agonists in ADP- and PAF-induced platelet aggregation. PRP was preincubated with (1) buffer, (2) normal mouse IgG1, (3) PECAM-1.3, or (4) PECAM-1.2. All antibodies were used at a final concentration of 5 μg/mL. Note that PECAM-1.2 augmented low-dose (1.25 μmol/L) ADP-induced platelet aggregation, both in the absence (A) and presence (B) of the MoAb IV.3 (a blocking MoAb specific for FcγRIIa). The degree of potentiation of aggregation induced by PECAM-1.2 varied somewhat from experiment to experiment (compare [A] with [B]), and this variation was not attributable to FcγRIIa blockade. Incubation of PRP with increasing concentrations of PECAM-1.2 (shown in micrograms per milliliter), followed by the addition of either 2 μmol/L ADP (C) or 40 nmol/L PAF (D), showed that the effects of antibody-mediated dimerization of PECAM-1 are dose-dependent.

Close modal
Dimerization of PECAM-1 on the platelet surface leads to conformational changes in αIIbβ3 and exposure of P-selectin on the platelet surface.

To investigate the molecular mechanism by which PECAM-1 Ig-domain 6 engagement promotes increased platelet adhesion (Fig 1) and aggregation (Fig 2), we examined the ability of PECAM-1 antibodies, in either monovalent or bivalent form, to induce (1) conformational changes in the platelet integrin αIIbβ3 and (2) α-granule secretion. As shown in Fig 3, incubation of platelets with PECAM-1.2 (directed against Ig-domain 6), but not PECAM-1.1 (Ig-domain 5) or PECAM-1.3 (Ig-domain 1), led to activation of the αIIbβ3 complex, as reported by the binding of the conformationally sensitive LIBS antibody, D3. This effect was dependent on PECAM-1 dimerization, because PECAM-1.2 F(ab′)2 fragments induced D3 binding, whereas monovalent Fab fragments did not (Fig 4A). A second Ig-domain 6-specific MoAb, 4G6, had similar effects. In addition, incubation of resting platelets with either of these two MoAbs, either as intact IgGs or F(ab′)2 fragments, led to exposure of the α-granule membrane-specific protein, P-selectin, on the platelet surface (Fig4B), indicating that PECAM-1 cross-linking also results in some degree of cellular activation. These events do not appear to be due to ADP release or thromboxane A2 generation, because, as shown in Table 1, the addition of PGE1, apyrase, indomethicin, or SQ 29.5 (a thromboxane receptor antagonist) before stimulation with PECAM-1.2 had little to no effect on D3 exposure.

Fig. 3.

Engagement of PECAM-1 Ig-domain 6 leads to conformational changes in the integrin αIIbβ3. Washed human platelets were preincubated with the FcγRIIa-specific antibody, IV.3 (to prevent possible Fc-receptor activation), before the addition of 10 μg/mL of normal mouse IgG (NM IgG), PECAM-1.1, PECAM-1.2, or PECAM-1.3. Buffer or RGDW peptide was used as negative and positive controls, respectively. After the addition of FITC-conjugated D3 for 30 minutes at room temperature, platelets were washed and transferred to 450 μL of RCD buffer, pH 7.4, and analyzed by flow cytometry. Note that PECAM-1.2 binding resulted in the exposure of the D3 epitope to almost the same extent as that induced by RGD peptide, a known modulator of integrin conformational change.62 The ability of PECAM-1.2 to induce conformational changes in αIIbβ3 varied somewhat from experiment to experiment; sometimes PECAM-1.2 was more effective than RGDW in inducing D3 binding (see Fig 4).

Fig. 3.

Engagement of PECAM-1 Ig-domain 6 leads to conformational changes in the integrin αIIbβ3. Washed human platelets were preincubated with the FcγRIIa-specific antibody, IV.3 (to prevent possible Fc-receptor activation), before the addition of 10 μg/mL of normal mouse IgG (NM IgG), PECAM-1.1, PECAM-1.2, or PECAM-1.3. Buffer or RGDW peptide was used as negative and positive controls, respectively. After the addition of FITC-conjugated D3 for 30 minutes at room temperature, platelets were washed and transferred to 450 μL of RCD buffer, pH 7.4, and analyzed by flow cytometry. Note that PECAM-1.2 binding resulted in the exposure of the D3 epitope to almost the same extent as that induced by RGD peptide, a known modulator of integrin conformational change.62 The ability of PECAM-1.2 to induce conformational changes in αIIbβ3 varied somewhat from experiment to experiment; sometimes PECAM-1.2 was more effective than RGDW in inducing D3 binding (see Fig 4).

Close modal
Fig. 4.

Cross-linking PECAM-1 induces conformational changes in αIIbβ3 and exposure of P-selectin on the platelet surface. Washed platelets (5 × 106/mL) were incubated with the indicated agonists as described in the Materials and Methods. All antibodies were used at a final concentration of 10 μg/mL and were used in the form of intact IgG (not shown), F(ab′)2 fragments, or as monovalent Fab fragments. After the addition of FITC-conjugated D3 (A) or FITC-conjugated S12 (B) for 30 minutes at room temperature, platelets were washed and transferred to 450 μL of RCD, pH 7.4, and analyzed by flow cytometry. Results are expressed as the mean ± SD of fluorescence intensity for three independent experiments. The observation that bivalent PECAM-1.2 or 4G6 F(ab′)2 fragments, but not their monovalent Fab counterparts, induced conformational changes in the αIIbβ3 integrin complex and exposure of P-selectin on the platelet surface suggests that receptor dimerization is required for outside/in signal transduction mediated by PECAM-1.

Fig. 4.

Cross-linking PECAM-1 induces conformational changes in αIIbβ3 and exposure of P-selectin on the platelet surface. Washed platelets (5 × 106/mL) were incubated with the indicated agonists as described in the Materials and Methods. All antibodies were used at a final concentration of 10 μg/mL and were used in the form of intact IgG (not shown), F(ab′)2 fragments, or as monovalent Fab fragments. After the addition of FITC-conjugated D3 (A) or FITC-conjugated S12 (B) for 30 minutes at room temperature, platelets were washed and transferred to 450 μL of RCD, pH 7.4, and analyzed by flow cytometry. Results are expressed as the mean ± SD of fluorescence intensity for three independent experiments. The observation that bivalent PECAM-1.2 or 4G6 F(ab′)2 fragments, but not their monovalent Fab counterparts, induced conformational changes in the αIIbβ3 integrin complex and exposure of P-selectin on the platelet surface suggests that receptor dimerization is required for outside/in signal transduction mediated by PECAM-1.

Close modal
Table 1.

Effect of Platelet Inhibitors on LIBS Exposure by PECAM-1.2

Agonist Inhibitor
50 ng/mL PGE110 U/mL Apyrase 5 μmol/L Indomethicin 5 μmol/L SQ 29.5
2 mmol/L RGEW  0  9.6 2.5  0  
2 mmol/L RGDW  138.1  151.8  136.5  136.5 
NM IgG  6.6  17.4  0  0  
PECAM-1.1  5.5  1.3 2.0  2.1  
PECAM-1.3  0  0  4.5  0  
PECAM-1.2 33.5  46.2  36.6  25.6 
Agonist Inhibitor
50 ng/mL PGE110 U/mL Apyrase 5 μmol/L Indomethicin 5 μmol/L SQ 29.5
2 mmol/L RGEW  0  9.6 2.5  0  
2 mmol/L RGDW  138.1  151.8  136.5  136.5 
NM IgG  6.6  17.4  0  0  
PECAM-1.1  5.5  1.3 2.0  2.1  
PECAM-1.3  0  0  4.5  0  
PECAM-1.2 33.5  46.2  36.6  25.6 

Platelets were treated with IV.3 Fab fragments (10 μg/mL) before the addition of the indicated agonist or MoAb. MoAbs were used at a final concentration of 10 μg/mL. Numbers shown are the mean fluorescence intensity given by the binding of the LIBS antibody, D3, to platelets after platelet stimulation by the indicated agonist, in the presence of various inhibitors of platelet secondary metabolites or scavengers of ADP release. The mean fluorescence intensity of D3 binding in the presence of buffer alone has been subtracted. Data shown are representative of results obtained in three independent experiments.

Engagement of PECAM-1 induces tyrosine phosphorylation of its cytoplasmic domain.

Recent studies have shown that PECAM-1 becomes tyrosine phosphorylated during platelet aggregation in an integrin-dependent process that results in the association of the SH2 domain-containing protein-tyrosine phosphatase, SHP-2, with tyrosine residues 663 and 686 of the PECAM-1 cytoplasmic domain.43 To further examine the mechanism by which PECAM-1.2 is able to act as a costimulatory agonist to modulate integrin-mediated cellular adhesion and aggregation responses (Figs 1-3), we incubated resting platelets with PECAM-1.2 F(ab′)2 fragments, solubilized the cells, immunocaptured PECAM-1, and probed Western blots of the resulting immunoprecipitated proteins with the phosphotyrosine-specific MoAb, PY-20. As shown in Fig 5, dimerization of PECAM-1 induced by PECAM-1.2 F(ab′)2 fragments initiated outside/in signal transduction, resulting in tyrosine phosphorylation of the PECAM-1 cytoplasmic domain to an extent similar to that induced by the strong platelet agonist, TRAP. These data suggest that dimerization or oligomerization of PECAM-1 on the platelet surface promotes downstream cellular events, including increased adhesion and aggregation, via the creation of specific docking sites for SH2-containing signaling molecules such as SHP-2.

Fig. 5.

PECAM-1 dimerization results in its tyrosine phosphorylation. Washed platelets (1 × 109/mL) were incubated at 37°C with the following agonists in the presence of 2 mmol/L CaCl2, 1 mmol/L MgCl2, and 100 μg/mL fibrinogen: (1) buffer (in the presence of stirring), (2) 7 μmol/L TRAP for 5 minutes under stirring conditions (fully aggregated), (3) 10 μg/mL normal mouse IgG1 F(ab′)2fragments for 30 minutes (stirred), and (4) 10 μg/mL PECAM-1.2 F(ab′)2 for 30 minutes (stirred). After detergent lysis, immunoprecipitations (IP) were performed using either normal mouse IgG1 (NM, left panel) or PECAM-1.3 (right panel). Bound proteins were resolved by 12.5% SDS-PAGE and analyzed by immunoblotting using an HRP-conjugated antiphosphotyrosine antibody (PY-20). Arrows indicate the positions of tyrosine phosphorylated PECAM-1 and the heavy chain of IgG.

Fig. 5.

PECAM-1 dimerization results in its tyrosine phosphorylation. Washed platelets (1 × 109/mL) were incubated at 37°C with the following agonists in the presence of 2 mmol/L CaCl2, 1 mmol/L MgCl2, and 100 μg/mL fibrinogen: (1) buffer (in the presence of stirring), (2) 7 μmol/L TRAP for 5 minutes under stirring conditions (fully aggregated), (3) 10 μg/mL normal mouse IgG1 F(ab′)2fragments for 30 minutes (stirred), and (4) 10 μg/mL PECAM-1.2 F(ab′)2 for 30 minutes (stirred). After detergent lysis, immunoprecipitations (IP) were performed using either normal mouse IgG1 (NM, left panel) or PECAM-1.3 (right panel). Bound proteins were resolved by 12.5% SDS-PAGE and analyzed by immunoblotting using an HRP-conjugated antiphosphotyrosine antibody (PY-20). Arrows indicate the positions of tyrosine phosphorylated PECAM-1 and the heavy chain of IgG.

Close modal

In addition to serving as a homophilic cell-cell adhesion molecule,22 recent studies have shown that antibody-mediated engagement or dimerization of PECAM-1 on the cell surface can result in a number of downstream cellular events, including synthesis and release of hydrogen peroxide44 and proinflammatory cytokines45 by human monocytes, cell spreading and cytoskeletal rearrangement of natural killer cells,46 tyrosine phosphorylation of multiple cytoplasmic proteins in human T lymphocytes,47 and differentiation of cord blood progenitor cells.48 In addition, cross-linking PECAM-1 on the surface of leukocytes has been shown to result in the activation of adhesion molecules of both the β1 and β2 integrin family.27-31 The purpose of the present investigation was to examine whether PECAM-1–mediated activation of integrins could be extended to the β3integrin subfamily and to begin to understand the mechanism by which PECAM-1 engagement leads to augmentation of integrin function.

Unlike that which has been shown in other cell types, numerous PECAM-1–specific antibodies have been examined for their effects on platelet function over the past 10 years, and not one has been found, by itself, to induce or inhibit in vitro platelet aggregation, adhesion, or granule secretion. In the present report, we found that certain anti–PECAM-1 MoAbs were able to potentiate the ability of human platelets to adhere both to ECM proteins (Fig 1) and to each other (Fig 2). The findings suggest that, at least in human platelets, PECAM-1 engagement is by itself insufficient to induce a measurable cellular response and requires costimulation by either shear (Fig 1) or another soluble agonist (Fig 2). We have extended these observations by further showing that the mechanism by which this occurs appears to be related to the generation of intracellular signals that follow dimerization or oligomerization of PECAM-1 on the cell surface. Thus, intact anti–PECAM-1 IgG as well as F(ab′)2fragments, but not monovalent Fab fragments, were shown to be capable of inducing conformational changes in the platelet integrin αIIbβ3 and exposing P-selectin on the platelet surface (Figs 3 and 4). Although we do not yet fully understand the signal transduction pathway that leads from PECAM-1 engagement to integrin activation, Levine et al47have recently shown that anti–PECAM-1 MoAbs induce tyrosine phosphorylation of multiple unidentified cellular substrates.47 Our finding that PECAM-1 itself becomes tyrosine phosphorylated after PECAM-1 antibody binding (Fig 5), coupled with the recent demonstration by Jackson et al43 that tyrosine phosphorylation of the PECAM-1 cytoplasmic domain creates docking sites for cytosolic SH2-containing signaling molecules, offers hints as to the molecular mechanism of PECAM-1/integrin cross-talk and may help explain the link between PECAM-1 dimerization and β1 and β2 integrin activation seen previously by other investigators.

Stockinger et al44,45 have clearly shown that, in some instances, cellular activation induced by anti–PECAM-1 MoAbs can be entirely explained by the secondary engagement of IgG Fc receptors. In fact, the contribution of Fc receptor activation to signal transduction events attributed to PECAM-1 engagement cannot be ruled out in several other investigations in which intact anti–PECAM-1 IgG and/or secondary antibodies were used to evoke a PECAM-1–specific cellular response.28,29,49 The Fc region of IgG of many animal species, when in the correct conformation, can serve as a potent stimulatory agonist, as exemplified by the often life-threatening clinical syndrome, heparin-induced thrombocytopenia, in which human antibodies specific for heparin/platelet factor 4 complexes bind, via the Fc region of the resulting immune complex, to the platelet Fc receptor (FcγRIIa), resulting in both platelet activation (thrombosis) and clearance (thrombocytopenia).50,51 The often-employed use of nonactivating, isotype-matched MoAbs alone is an insufficient control for specificity of Fc receptor activation, because unbound antibodies do not have nearly as high an affinity for the Fc receptor as do bound antibodies,37 and many MoAbs, even when bound via their Fab regions to their target antigens, are spatially oriented in such a manner that precludes Fc receptor activation (for a more detailed treatment of the critical role of antigen topography in the interaction of antibodies with Fc receptors, see Kumpel et al,52 Horsewood et al,53 and Tomiyama et al54). In the present investigation, we attempted to carefully exclude Fc receptor-mediated cellular activation by using F(ab′)2 antibody fragments and/or by preblocking platelet FcγRIIa (the only Fc receptor for IgG present in platelets55) with saturating levels of the blocking FcγRIIa-specific MoAb, IV.3. The fact that augmentation of platelet aggregation (Fig 2), LIBS and P-selectin exposure (Figs 3 and 4 and Table 1), and PECAM-1 tyrosine phosphorylation (Fig 5) all occurred even after these precautions had been taken strongly support the notion that PECAM-1 receptor dimerization, and not secondary Fc receptor engagement, is capable of leading directly to cellular activation.

It is not clear why some anti–PECAM-1 MoAbs are able to augment integrin function and cellular activation, whereas others are not, or why, in the present study, both potentiating anti–PECAM-1 antibodies, PECAM-1.2 and 4G6, map to Ig-domain 6, the Ig-homology domain closest to the membrane. Several previous observations may be relevant. First, Ig-domain 6 contains two divalent cation binding sites,56although the structural and functional consequences of cation occupancy are not yet known. Second, Sun et al22 recently showed that the homophilic adhesive properties of PECAM-1–containing proteoliposomes could be selectively increased by Fab fragments of the same two antibodies, PECAM-1.2 and 4G6 (anti–PECAM-1 Fabs specific for other Ig homology domains did not augment adhesion), and proposed that engagement of domain 6 might induce LIBS-like long-range conformational changes in the PECAM-1 molecule. Finally, another domain 6-specific antibody, LYP21, as well as an Ig-domain 6 peptide corresponding to amino acid residues 551-574 have been shown to inhibit T-cell responses57 and delay the onset of graft-versus-host disease.58 Together with the data presented here on the effect of these antibodies on integrin activation and P-selectin exposure, it is tempting to speculate that Ig-domain 6 may play a regulatory role in PECAM-1 function, mediating both outside-out as well as outside-in signal transduction.

However, other explanations for the seemingly selective action of domain 6 antibodies on cell function are possible. It could be that MoAbs specific for Ig-domain 6 happen to bind with a topographical orientation that preferentially favors PECAM-1/PECAM-1 interactions on the cell surface. Receptor dimerization, in turn, may be all that is required to induce conformational changes in the molecule, leading to its activation and initiating intracellular tyrosine phosphorylation, augmenting integrin function, or increasing the homophilic binding properties of PECAM-1 proteoliposomes. It is notable that augmentation of β1 and β2 integrin function in leukocytes does not appear to be limited to Ig-domain 6-specific MoAbs27,35; perhaps the relative receptor density or its lateral mobility within the plane of the membrane is sufficiently different from that in platelets to permit PECAM-1 dimerization by a less-selective set of anti–PECAM-1 MoAbs. Further studies using fixed monomeric and dimeric membrane-bound forms of PECAM-1 are planned to distinguish between these two models of PECAM-1–mediated cellular activation.

Independent of the mechanism by which PECAM-1 MoAbs are exerting their effects, it is becoming clear that PECAM-1 can both initiate, as well as respond to, changes in cellular function. Osawa et al59have recently shown that PECAM-1 becomes tyrosine phosphorylated in endothelial cells subjected to mechanical shear stress and suggested that PECAM-1 may be one of the junctional receptors responsible for sensing and then communicating changes in fluid flow. Whether mechanical shear contributes to the tyrosine phosphorylation of platelet PECAM-1 that occurs in aggregating platelets43 is not known. In addition, Sagawa et al60 showed that aggregation of the high-affinity IgE receptor in rat basophilic leukemia cells also results in the tyrosine phosphorylation of PECAM-1. Finally, the recent studies of Lu et al61 suggest that the phosphorylation state of PECAM-1 may also be sensitive to cell-cell and cell matrix interactions, because integrin engagement and/or cell migration of cultured endothelial cells were shown to result in tyrosine dephosphorylation of PECAM-1. Taken together, these data implicate PECAM-1 as one of a growing number of cell surface receptors that are able to mediate bidirectional signal transduction, serving both as an agonist receptor as well as an adhesion molecule in blood and vascular cells. Studies defining the cytosolic signaling molecules that link these two interrelated cellular functions of PECAM-1 represent important areas of future investigation.

The authors thank Kevin Kupcho for technical support in performing flow cytometric measurements, Dr Gian Visentin for his assistance in HITP antibody-induced activation of platelets, and Dr William Campbell for providing indomethicin and SQ 29.5.

D.V. and D.E.J. contributed equally to this work.

Supported by Grants No. HL-44612 and HL-40926 (to P.J.N.) from the National Institutes of Health and a postdoctoral fellowship award to D.E.J. (#F96F-Post-34) from the Wisconsin Affiliate of the American Heart Association. D.V. and N.S. were supported by grants from the National Council for Research and Development, Israel and Deutsche Forschungsanstalt, Fuer Luft und Raumfahrt. P.J.N. is an Established Investigator of the American Heart Association.

Presented in abstract form at the 1996 European Granulocyte and Platelet Meeting in Helsinki, Finland.

Address reprint requests to Peter J. Newman, PhD, Blood Research Institute, The Blood Center of Southeastern Wisconsin, 638 N 18th St, Milwaukee, WI 53233-2121.

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

1
Newman
PJ
The biology of PECAM-1.
J Clin Invest
99
1997
3
2
Newman
PJ
Berndt
MC
Gorski
J
White
GC
Lyman
S
Paddock
C
Muller
WA
PECAM-1 (CD31) cloning and relation to adhesion molecules of the immunoglobulin gene superfamily.
Science
247
1990
1219
3
Newman
PJ
Hillery
CA
Albrecht
R
Parise
LV
Berndt
MC
Mazurov
AV
Dunlop
LC
Zhang
J
Rittenhouse
SE
Activation-dependent changes in human platelet PECAM-1: Phosphorylation, cytoskeletal association, and surface membrane redistribution.
J Cell Biol
119
1992
239
4
Zehnder
JL
Hirai
K
Shatsky
M
McGregor
JL
Levitt
LJ
Leung
LLK
The cell adhesion molecule CD31 is phosphorylated after cell activation. Down-regulation of CD31 in activated T lymphocytes.
J Biol Chem
267
1992
5243
5
Watt
SM
Williamson
J
Genevier
H
Fawcett
J
Simmons
DL
Harzfield
A
Nesbitt
SA
Coombe
DR
The heparin binding PECAM-1 adhesion molecule is expressed by CD34+ hematopoietic precursor cells with early myeloid and B-lymphoid cell phenotypes.
Blood
82
1993
2649
6
(abstr, suppl 1)
Jackson
DE
Ward
CM
Wang
R
Newman
PJ
The protein-tyrosine phosphatase, SHP-2, binds PECAM-1 and forms a distinct signaling complex during platelet aggregation: Evidence for a mechanistic link between PECAM-1- and integrin-mediated signal transduction.
Blood
88
1996
438a
7
Bogen
SA
Baldwin
HS
Watkins
SC
Albelda
SM
Abbas
AK
Association of murine CD31 with transmigrating lymphocytes following antigenic stimulation.
Am J Pathol
141
1992
843
8
Johnson
BA
Haines
GK
Harlow
LA
Koch
AE
Adhesion molecule expression in human synovial tissue.
Arthritis Rheum
36
1993
137
9
Tang
Q
Hendricks
RL
Interferon gamma regulates platelet endothelial cell adhesion molecule 1 expression and neutrophil infiltration into Herpes Simplex virus-infected mouse corneas.
J Exp Med
184
1996
1435
10
Ioffreda
MD
Albelda
SM
Elder
DE
Radu
A
Leventhal
LC
Zweiman
B
Murphy
GF
TNFα induces E-selectin expression and PECAM-1 (CD31) redistribution in extracutaneous tissues.
Endothelium
1
1993
47
11
Romer
LH
McLean
NV
Yan
H-C
IFN-gamma and TNF-α induce redistribution of PECAM-1 (CD31) on human endothelial cells.
J Immunol
154
1995
6582
12
Stewart
RJ
Kashour
TS
Marsden
PA
Vascular endothelial platelet endothelial adhesion molecule-1 (PECAM-1) expression is decreased by TNF-alpha and IFN-gamma. Evidence for cytokine-induced destabilization of messenger ribonucleic acid transcripts in bovine endothelial cells.
J Immunol
156
1996
1221
13
Rival
Y
Maschio
AD
Rabiet
M-J
Dejana
E
Duperray
A
Inhibition of platelet endothelial cell adhesion molecule-1 synthesis and leukocyte transmigration in endothelial cell by the combined action of TNF-α and IFN-gamma.
J Immunol
157
1996
1233
14
Zocchi
MR
Ferrero
E
Leone
BE
Rovere
P
Bianchi
E
Toninelli
E
Pardi
R
CD31/PECAM-1-driven chemokine-independent transmigration of human T lymphocytes.
Eur J Immunol
26
1996
759
15
Muller
WA
Weigl
SA
Deng
X
Phillips
DM
PECAM-1 is required for transendothelial migration of leukocytes.
J Exp Med
178
1993
449
16
Vaporciyan
AA
DeLisser
HM
Yan
H-C
Mendiguren
II
Thom
SR
Jones
ML
Ward
PA
Albelda
SM
Involvement of platelet endothelial cell adhesion molecule-1 in neutrophil recruitment in vivo.
Science
262
1993
1580
17
Bogen
S
Pak
J
Garifallou
M
Deng
X
Muller
WA
Monoclonal antibody to murine PECAM-1 (CD31) blocks acute inflammation in vivo.
J Exp Med
179
1994
1059
18
Wakelin
MW
Sanz
M-J
Dewar
A
Albelda
SM
Larkin
SW
Boughton-Smith
N
Nourshargh
S
An anti-platelet-endothelial cell adhesion molecule-1 antibody inhibits leukocyte extravasation from mesenteric microvessels in vivo by blocking the passage through the basement membrane.
J Exp Med
184
1996
229
19
Murohara
T
Delyani
JA
Albelda
SM
Lefer
AM
Blockade of platelet endothelial cell adhesion molecule-1 protects against myocardial ischemia and reperfusion injury in cats.
J Immunol
156
1996
3550
20
Gumina
RJ
Schultz
JE
Yao
Z
Kenny
D
Warltier
DC
Newman
PJ
Gross
GJ
Antibody to platelet/endothelial cell adhesion molecule-1 reduces myocardial infarct size in a rat model of ischemia-reperfusion injury.
Circulation
94
1996
3327
21
Fawcett
J
Buckley
C
Holness
CL
Bird
IN
Spragg
JH
Saunders
J
Harris
A
Simmons
DL
Mapping the homotypic binding sites in CD31 and the role of CD31 adhesion in the formation of interendothelial cell contacts.
J Cell Biol
128
1995
1229
22
Sun
Q-H
DeLisser
HM
Zukowski
MM
Paddock
C
Albelda
SM
Newman
PJ
Individually distinct Ig homology domains in PECAM-1 regulate homophilic binding and modulate receptor affinity.
J Biol Chem
271
1996
11090
23
Sun
J
Williams
J
Yan
H-C
Amin
KM
Albelda
SM
DeLisser
HM
Platelet endothelial cell adhesion molecule-1 (PECAM-1) homophilic adhesion is mediated by immunoglobulin-like domains 1 and 2 and depends on the cytoplasmic domain and the level of surface expression.
J Biol Chem
271
1996
18561
24
Piali
L
Hammel
P
Uherek
C
Bachmann
F
Gisler
RH
Dunon
D
Imhof
BA
CD31/PECAM-1 is a ligand for αvβ3 integrin involved in adhesion of leukocytes to endothelium.
J Cell Biol
130
1995
451
25
Buckley
CD
Doyonnas
R
Newton
JP
Blystone
SD
Brown
EJ
Watt
SM
Simmons
DL
Identification of αvβ3 as a heterotypic ligand for CD31/PECAM-1.
J Cell Sci
109
1996
437
26
Prager
E
Sunder-Plassmann
R
Hansmann
C
Koch
C
Holter
W
Knapp
W
Stockinger
H
Interaction of CD31 with a heterophilic counterreceptor involved in downregulation of human T cell responses.
J Exp Med
184
1996
41
27
Tanaka
Y
Albelda
SM
Horgan
KJ
Van Seventer
GA
Shimizu
Y
Newman
W
Hallam
J
Newman
PJ
Buck
CA
Shaw
S
CD31 expressed on distinctive T cell subsets is a preferential amplifier of β1 integrin-mediated adhesion.
J Exp Med
176
1992
245
28
Leavesley
DI
Oliver
JM
Swart
BW
Berndt
MC
Haylock
DN
Simmons
PJ
Signals from platelet/endothelial cell adhesion molecule enhance the adhesive activity of the very late antigen-4 integrin of human CD34+ hemopoietic progenitor cells.
J Immunol
153
1994
4673
29
Piali
L
Albelda
SM
Baldwin
HS
Hammel
P
Gisler
RH
Imhof
BA
Murine platelet enbdothelial cell adhesion molecule (PECAM-1/CD31) modulates β2 integrins on lymphokine-activated killer cells.
Eur J Immunol
23
1993
2464
30
Berman
ME
Muller
WA
Ligation of platelet/endothelial cell adhesion molecule 1 (PECAM-1/CD31) on monocytes and neutrophils increases binding capacity of leukocyte CR3 (CD11b/CD18).
J Immunol
154
1995
299
31
Berman
ME
Xie
Y
Muller
WA
Roles of platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31) in natural killer cell transendothelial migration and β2 integrin activation.
J Immunol
156
1996
1515
32
Mazurov
AV
Vinogradov
DV
Kabaeva
NV
Antonova
AN
Romanov
YA
Vlasik
TN
Antonov
AS
Smirnov
VN
A monoclonal antibody, VM64, reacts with a 130 kDa glycoprotein common to platelets and endothelial cells: Heterogeneity in antibody binding to human aortic endothelial cells.
Thromb Haemost
66
1991
494
33
Metzelaar
MJ
Korteweg
J
Sixma
JJ
Nieuwenhuis
HK
Biochemical characterization of PECAM-1 (CD31 antigen) on human platelets.
Thromb Haemost
66
1991
700
34
Wagner
CL
Mascelli
MA
Neblock
DS
Weisman
HF
Coller
BS
Jordan
RE
Analysis of GPIIb/IIIa receptor number by quantification of 7E3 binding to human platelets.
Blood
88
1996
907
35
Yan
H-C
Pilewski
JM
Zhang
Q
DeLisser
HM
Romer
L
Albelda
SM
Localization of multiple functional domains on human PECAM-1 (CD31) by monoclonal antibody epitope mapping.
Cell Adhesion Commun
3
1995
45
36
Liao
F
Huynh
HK
Eiroa
A
Greene
T
Polizzi
E
Muller
WA
Migration of monocytes across endothelium and passage through extracellular matrix involve separate molecular domains of PECAM-1.
J Exp Med
182
1995
1337
37
Rosenfeld
SI
Looney
RJ
Leddy
JP
Phipps
DC
Abraham
GN
Anderson
CL
Human platelet Fc receptor for immunoglobulin G. Identification as a 40,000-molecular-weight membrane protein shared by monocytes.
J Clin Invest
76
1985
2317
38
Kouns
WC
Wall
CD
White
MM
Fox
CF
Jennings
LK
A conformation-dependent epitope of human platelet glycoprotein IIIa.
J Biol Chem
265
1990
20594
39
McEver
RP
Martin
MN
A monoclonal antibody to a membrane glycoprotein binds only to activated platelets.
J Biol Chem
259
1984
9799
40
Gospodarowicz
D
Greenburg
G
Foidart
JM
Savion
N
The production and localization of laminin in cultured vascular and corneal endothelial cells.
J Cell Physiol
107
1981
171
41
Varon
D
Dardik
R
Shenkman
B
Kotev-Emeth
S
Farzame
N
Tamarin
I
Savion
N
A new method for quantitative analysis of whole blood platelet interaction with extracellular matrix under flow conditions.
Throm Res
85
1997
283
42
Ohto
H
Maeda
H
Shibata
Y
Chen
R-F
Qzaki
Y
Higashihara
M
Takeuchi
A
Tohyama
H
A novel leukocyte differentiation antigen: Two monoclonal antibodies TM2 and TM3 define a 120-kd molecule present on neutrophils, monocytes, platelets, and activated lymphoblasts.
Blood
66
1985
873
43
Jackson
DE
Ward
CM
Wang
R
Newman
PJ
The protein-tyrosine phosphatase SHP-2 binds PECAM-1 and forms a distinct signaling complex during platelet aggregation. Evidence for a mechanistic link between PECAM-1 and integrin-mediated cellular signaling.
J Biol Chem
272
1997
6986
44
Stockinger
H
Gadd
SJ
Eher
R
Majdic
O
Schreiber
W
Kasinrerk
W
Strass
B
Schnabl
E
Knapp
W
Molecular characterization and functional analysis of the leukocyte surface protein CD31.
J Immunol
145
1990
3889
45
Chen
W
Knapp
W
Majdic
O
Stockinger
H
Bohmig
GA
Zlabinger
GJ
Co-ligation of CD31 and FcgammaRII induces cytokine production in human monocytes.
J Immunol
152
1994
3991
46
Poggi
A
Panzeri
MC
Moretta
L
Zocchi
MR
CD31-triggered rearrangement of the actin cytoskeleton in human natural killer cells.
Eur J Immunol
26
1996
817
47
Levine
BL
Ueda
Y
Faith
A
June
CH
Signal transduction properties of the T cell activation monoclonal antibody panel: Ubiquitous increase in cellular substrate tyrosine phosphorylation in the absence of detectable calcium mobilization.
Tissue Antigens
48
1996
319
48
El Marsafy
S
Carosella
ED
Agrawal
SG
Gluckman
E
Mansur
I-G
Elhabazi
A
Bensussan
A
Functional role of PECAM-1/CD31 molecule expressed on human cord blood progenitors.
Leukemia
10
1996
1340
49
Takada
Y
Hemler
ME
The primary structure of the VLA-2/collagen receptor alpha 2 subunit (platelet GPIa): Homology to other integrins and the presence of a possible collagen-binding domain.
J Cell Biol
109
1989
397
50
Kelton
JG
Sheridan
D
Santos
A
Smith
J
Steeves
K
Smith
C
Brown
C
Murphy
WG
Heparin-induced thrombocytopenia: Laboratory studies.
Blood
72
1988
925
51
Visentin
GP
Ford
SE
Scott
JP
Aster
RH
Antibodies from patients with heparin-induced thrombocytopenia/thrombosis are specific for platelet factor 4 complexed with heparin or bound to endothelial cells.
J Clin Invest
93
1993
81
52
Kumpel
BM
van de Winkel
JGJ
Westerdaal
NAC
Hadley
AG
Dugoujon
JM
Blancher
A
Antigen topography is critical for interaction of IgG2 anti-red-cell antibodies with Fcgamma receptors.
Br J Haematol
94
1996
175
53
Horsewood
P
Hayward
CPM
Warkentin
TE
Kelton
JG
Investigation of the mechanism of monoclonal antibody-induced platelet activation.
Blood
78
1991
1019
54
Tomiyama
Y
Kunicki
TJ
Zipf
TF
Ford
SB
Aster
RH
Response of human platelets to activating monoclonal antibodies: Importance of FcgammaRII (CD32) phenotype and level of expression.
Blood
80
1992
2261
55
Anderson
CL
Chacko
GW
Osborne
JM
Brandt
JT
The Fc receptor for immunoglobulin G (FcgammaRII) on human platelets.
Semin Thromb Hemost
21
1995
1
56
Jackson
DE
Loo
RO
Holyst
MT
Newman
PJ
Identification and characterization of functional cation coordination sites in platelet endothelial cell adhesion molecule-1 (PECAM-1).
Biochemistry
36
1997
9395
57
Zehnder
JL
Shatsky
M
Leung
LLK
Butcher
EC
McGregor
JL
Levitt
LJ
Involvement of CD31 in lymphocyte-mediated immune responses: Importance of the membrane-proximal immunoglobulin domain and identification of an inhibiting CD31 peptide.
Blood
85
1995
1282
58
Chen
Y
Schlegel
PG
Tran
N
Thompson
D
Zehnder
JL
Chao
NJ
Administration of a CD31-derived peptide delays the onset and significantly increases survival from lethal graft-versus-host disease.
Blood
89
1997
1452
59
Osawa
M
Masuda
M
Harada
N
Lopes
RB
Fujiwara
K
Tyrosine phosphorylation of platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31) in mechanically stimulated vascular endothelial cells.
Eur J Cell Biol
72
1997
229
60
Sagawa
K
Swaim
W
Zhang
J
Unsworth
E
Siraganian
RP
Aggregation of the hign affinity IgE receptor results in the tyrosine phosphorylation of the surface adhesion protein PECAM-1 (CD31).
J Biol Chem
272
1997
13412
61
Lu
TT
Yan
LG
Madri
JA
Integrin engagement mediates tyrosine dephosphorylation on platelet-endothelial cell adhesion molecule 1.
Proc Natl Acad Sci USA
93
1996
11808
62
Du
XP
Plow
EF
Frelinger
AL
O'Toole
TE
Loftus
JC
Ginsberg
MH
Ligands “activate” integrin alpha IIb beta 3 (platelet GPIIb-IIIa).
Cell
65
1991
409
Sign in via your Institution