Platelet responses to collagen are mediated by the combined actions of the integrin α2β1, which serves as a major collagen-binding receptor, and the GPVI/FcRγ-chain complex, which transmits collagen-specific activation signals into the cell interior through the action of an immunoreceptor tyrosine-based activation motif within the cytoplasmic domain of the FcRγ-chain. Despite much progress in identifying components of the signaling pathway responsible for collagen-induced platelet activation, virtually nothing is known about the regulatory elements that modulate this important hemostatic event. PECAM-1, a recently recognized member of the inhibitory receptor family, contains a functional immunoreceptor tyrosine-based inhibitory motif within its cytoplasmic domain that, when tyrosine phosphorylated, recruits and activates the protein–tyrosine phosphatase, SHP-2. To test the hypothesis that PECAM-1 functions to regulate GPVI/FcRγ-chain–mediated platelet activation, the responses of wild-type versus PECAM-1–deficient murine platelets to GPVI-specific agonists were compared. Four distinct GPVI/FcRγ-chain–dependent responses were found to be significantly exaggerated in platelets derived from PECAM-1–deficient mice, including Mg++-independent adhesion to immobilized fibrillar collagen, collagen-induced platelet aggregation, platelet aggregation induced by the GPVI-specific agonist collagen-related peptide, and GPVI/FcRγ-chain–induced dense granule secretion. Together, these data provide compelling evidence that PECAM-1 modulates platelet responses to collagen, and they implicate this novel member of the inhibitory receptor family in the regulation of primary hemostasis.

Platelet endothelial cell adhesion molecule-1 (PECAM-1, CD31) is a 130-kd member of the immunoglobulin (Ig) superfamily that is expressed on the surface of circulating platelets, endothelial cells, neutrophils, monocytes, and certain T-lymphocyte subsets. The extracellular domain of PECAM-1 is composed of 6 extracellular Ig-like homology units,1 the amino-terminal 2 of which mediates PECAM-1–PECAM-1 homophilic interactions.2,3 Antibodies to the extracellular domain have been shown to have profound physiologic and cell biologic effects, including delaying leukocyte transendothelial migration4-6and inhibiting angiogenesis.7 The extracellular domain of PECAM-1 also serves as a portal for entry into endothelial cells of certain strains of Plasmodium falciparum–infected erythrocytes.8 9 

The PECAM-1 cytoplasmic domain also plays a key biologic role because a large number of extracellular stimuli (reviewed in Newman10) have been shown to result in the phosphorylation of 2 key tyrosine residues, located at positions 663 and 686 of the cytoplasmic domain.11 The sequence surrounding each of these 2 tyrosine residues conforms to an immunoreceptor tyrosine-based inhibitory motif (ITIM) that, when phosphorylated, provides a major docking site for the src homology 2 (SH2) domain–containing protein tyrosine phosphatase (PTP), SHP-2.11-14 Based on the presence of its highly conserved ITIM and the similarity in its genomic organization to that of other ITIM-bearing transmembrane receptors, we have recently proposed10 that PECAM-1 is a member of a growing family of transmembrane PTP-binding proteins collectively known as inhibitory receptors.

There are good examples at the cell biological level that support a role for inhibitory receptors in attenuating the action of agonist receptors that contain one or more immunoreceptor tyrosine-based activation motifs (ITAMs).15,16Human platelets express 2 such ITAM-bearing receptors—FcγRIIa,17 which serves as a low-affinity receptor for IgG,18 and the GPVI-FcRγ–chain complex, which transmits collagen-specific activation signals into the cell interior through the action of an ITAM present within the cytoplasmic domain of the FcRγ-chain.19 Although a role forITIM-bearing PECAM-1 in regulating the actions of either of these 2 ITAM-bearing agonist receptors in platelets has not been examined, we have recently established a regulatory role for PECAM-1 in attenuating the action of the ITAM-bearing T-cell antigen receptor in Jurkat T cells.20 These studies suggested to us that PECAM-1 may similarly function to negatively regulate protein tyrosine kinase (PTK)-dependent signal transduction pathways initiated by ITAM-containing stimulatory receptors in other cell types. The recent production of PECAM-1–deficient mice21 has enabled us to use a genetic approach to evaluate the effect of PECAM-1 on platelet responses to collagen. The purpose of the current investigation, therefore, was to examine the ability of PECAM-1 to modulate GPVI/FcRγ-chain–mediated platelet activation by comparing the responses of wild-type versus PECAM-1–deficient murine platelets to GPVI-specific agonists. Our findings provide compelling evidence that PECAM-1 modulates platelet responses to collagen and implicate this novel member of the inhibitory receptor family in the regulation of primary hemostasis.

Materials

A suspension of acid-insoluble equine tendon type I collagen fibrils was purchased from Chrono-Log (Havertown, PA). Collagen-related peptide (CRP:GKP*[GPP*]10GKP*G; single amino acid code; P* represents hydroxyproline) was synthesized and cross-linked as previously described.22 An adenosine triphosphate (ATP) determination kit and calcine am were purchased from Molecular Probes (Eugene, OR). PECAM-1−/−mice21 were obtained from Dr Tak W. Mak (Amgen Institute, Toronto, ON, Canada) and were bred at the Animal Resource Facility of the Medical College of Wisconsin. Immulon-2 96-well microtiter plates were purchased from Dynex Technologies (Chantilly, VA). All other chemicals were purchased from Sigma Chemical (St Louis, MO).

Preparation of mouse platelets

Blood (1.0-1.5 mL) from the inferior vena cava of tribromoethanol-anesthetized mice was drawn into 0.13 M sodium-citrate solution (0.1 mL). Blood pooled from 3 to 4 animals was diluted with an equal volume of modified Tyrode–HEPES buffer (134 mM NaCl, 0.36 mM NaH2PO4, 13.8 mM NaHCO3, 2.5 mM KCl, 20 mM HEPES, 5.5 mM glucose, 2.5 mg/mL bovine serum albumin, pH 7.4) and centrifuged at 200g for 10 minutes at room temperature. Platelets were collected by centrifuging the platelet-rich plasma at 500g for 20 minutes in the presence of 10 mM EDTA and 1 μmol/L prostaglandin E1. The sedimented platelets were resuspended in modified Tyrode–HEPES buffer to a density of 2.5 × 108 platelets/mL, supplemented with 1 mM CaCl2, and allowed to rest for 30 minutes at room temperature before experimentation.

Adhesion assays

Immulon-2 microtiter 96-well plates were coated with varying concentrations of fibrillar collagen without Mg2+ or with a constant concentration of poly L-lysine (0.01%) overnight at 4°C. Washed wild-type and PECAM-1−/− platelets, prepared as described above, were loaded with calcine am (5 μmol/L) for 30 minutes at 37°C. The loaded wild-type and PECAM-1−/− platelets were rewashed in modified Tyrode–HEPES buffer and suspended at a concentration of 1.2 × 108 platelets/mL. Calcine am-loaded platelets (100 μL) were added to the matrix-coated wells and incubated for the indicated amount of time at 37°C. Nonadherent platelets were removed by washing, and the extent of platelet adherence was determined by measuring the fluorescence emitted by adhered platelets at 520 nm after excitation at 490 nm. The assay was performed in triplicate, and each data point represented the mean ± SD of triplicate measurements.

Platelet aggregation and secretion

Platelets were suspended in 200 μL at a final concentration of 2.5 × 108 platelets/mL, and aggregometry was performed at 37°C in a Bio/Data PAP-4 channel aggregometer (Horsham, PA) with continuous stirring (1000 rpm). ATP release was determined by removing 10-μL aliquots at selected time points, centrifuging them at 500g for 1 minute at room temperature in the presence of 1 mM EDTA to prevent further platelet activation, and adding the supernatant to a 100-μL reaction mixture containing luciferase (0.5 mM) and luciferin (1.25 μg/mL). The luminescence generated in the presence of platelet-released ATP was read in a luminometer (TD-20e; Turner, Sunnyvale, CA) and compared to an ATP standard curve to determine the amount of ATP released.

Platelets derived from PECAM-1–deficient mice exhibit exaggerated GPVI-mediated adhesion and aggregation responses

Platelet activation by collagen is thought to proceed by a 2-step mechanism involving the initial attachment of collagen fibrils to surface-expressed integrin α2β1, followed by an obligatory signal transduction cascade mediated, at least in part, by the GPVI/FcRγ-chain complex.19 The latter operates through tyrosine-phosphorylated ITAMs within the cytoplasmic domain of the FcRγ-chain dimer.23 Despite much progress in identifying components of the signaling pathway responsible for initiating and sustaining collagen-induced platelet activation,24-30 virtually nothing is known about the regulatory elements that modulate this important hemostatic event. Others and we have recently found that PECAM-1 becomes strongly tyrosine phosphorylated on its ITIM after exposure of platelets to collagen,31 32 leading us to speculate that agonist induction simultaneously activates a feedback inhibitory pathway mediated by PECAM-1.

To test the hypothesis that PECAM-1 normally functions to attenuate GPVI-mediated platelet activation, we first compared the ability of platelets derived from wild-type versus PECAM-1–deficient mice to bind to immobilized fibrillar collagen in the absence of Mg2+—conditions that have previously been shown to result in GPVI-specific adhesion, independent of α2β1.24 33 As shown in Figure1, though wild-type and PECAM-1−/− platelets exhibited similar levels of adhesion after 15 minutes of exposure (Figure 1, top panel), PECAM-1−/− platelets bound significantly and reproducibly better at 30 (Figure 1, middle panel) and 60 (Figure 1, lower panel) minutes than did wild-type platelets. This effect was specific; adhesion to poly-L-lysine–coated wells was statistically indistinguishable between the 2 experimental groups.

Fig. 1.

PECAM-1−/− platelets demonstrate increased adhesion to immobilized fibrillar collagen in the absence of Mg2+.

Calcine am-loaded platelets derived from wild-type (open symbols) and PECAM-1−/− (closed symbols) mice were allowed to adhere, in the absence of Mg2+, to plates coated with increasing concentrations of fibrillar collagen (circles) or to plates coated with a constant concentration of polyL-lysine (squares) for 15, 30, or 60 minutes at 37°C. Wild-type and PECAM-1−/− platelets were labeled to a similar extent with calcine am (not shown). After removal of nonadherent platelets, fluorescence associated with adherent platelets was measured as described in “Materials and methods.” Each assay was performed in triplicate, and each data point represents the mean of triplicate measurements ± SD. Similar results were obtained from 3 independent experiments. Note that wild-type and PECAM-1−/− platelets bound to a similar extent to polyL-lysine–coated plates, whereas at later time points, higher levels of adhesion to fibrillar collagen, in the absence of Mg2+, was observed in PECAM-1−/− platelets relative to the wild-type platelets.

Fig. 1.

PECAM-1−/− platelets demonstrate increased adhesion to immobilized fibrillar collagen in the absence of Mg2+.

Calcine am-loaded platelets derived from wild-type (open symbols) and PECAM-1−/− (closed symbols) mice were allowed to adhere, in the absence of Mg2+, to plates coated with increasing concentrations of fibrillar collagen (circles) or to plates coated with a constant concentration of polyL-lysine (squares) for 15, 30, or 60 minutes at 37°C. Wild-type and PECAM-1−/− platelets were labeled to a similar extent with calcine am (not shown). After removal of nonadherent platelets, fluorescence associated with adherent platelets was measured as described in “Materials and methods.” Each assay was performed in triplicate, and each data point represents the mean of triplicate measurements ± SD. Similar results were obtained from 3 independent experiments. Note that wild-type and PECAM-1−/− platelets bound to a similar extent to polyL-lysine–coated plates, whereas at later time points, higher levels of adhesion to fibrillar collagen, in the absence of Mg2+, was observed in PECAM-1−/− platelets relative to the wild-type platelets.

Close modal

There is growing support for the notion that ITAM- and ITIM-bearing receptors act antagonistically when expressed in the same cell, possibly because of the potential for ITIM-associated PTPs to dephosphorylate ITAM-bearing receptors or their tyrosine-phosphorylated substrates.16,34 To determine whether the observed exaggerated platelet adhesion to immobilized collagen was specific for ITAM-bearing agonist receptors, we compared platelet aggregation responses to GPVI-specific agonists22with those obtained using thrombin, which acts through a heterotrimeric G-protein–coupled receptor.35 

Relative to wild-type platelets, PECAM-1–deficient platelets exhibited strikingly hyper-aggregation responses to subthreshold doses of fibrillar collagen (Figure 2A) or CRP (Figure 2C), though the aggregation responses were equivalent at higher concentrations of either agonist (Figure 2B,D). In contrast, platelet activation by either low-dose (Figure 2E) or high-dose (Figure 2F) thrombin was identical in wild-type and PECAM-1−/−platelets, indicating that PECAM-1 has no detectable effect on this G-protein–coupled signal transduction pathway, as predicted. These results demonstrate that, like platelet adhesion, platelet aggregation mediated by the ITAM-bearing GPVI/FcRγ-chain complex is enhanced in the absence of PECAM-1. The observation that higher concentrations of either GPVI-specific agonist can overcome the difference in aggregation between PECAM-1−/− and wild-type platelets suggests that PECAM-1 deficiency lowers the threshold for the induction of platelet aggregation after collagen binding to GPVI. Interestingly, mice heterozygous for PECAM-1 expression exhibited near-normal responses to low-dose collagen or low-dose CRP, suggesting that, like integrin αIIbβ3 defects, 50% receptor expression is sufficient to maintain normal function.

Fig. 2.

PECAM-1−/− platelets show hyper-aggregation in response to stimulation with GPVI-specific agonists.

Murine platelets were isolated from wild-type (WT) and PECAM-1−/− (KO) mice, washed, and resuspended at a concentration of 2.5 × 108 platelets/mL. Stirred platelets (200 μL) were exposed to fibrillar collagen in absence of Mg2+ at a concentration of 1 μg/mL (A) or 10 μg/mL (B), to CRP at a concentration of 0.5 μg/mL (C) or 5 μg/mL (D), or to thrombin at a concentration of 0.1 (E) or 1 (F) U/mL. Changes in light transmission of the stimulated platelet suspension were measured in an aggregometer. Similar results were obtained with 5 separate platelet preparations. Note that compared to wild-type platelets, PECAM-1−/− platelets showed hyper-aggregation in response to stimulation with low concentrations of GPVI-specific agonists but not in response to stimulation with high concentrations of the same agonists or thrombin.

Fig. 2.

PECAM-1−/− platelets show hyper-aggregation in response to stimulation with GPVI-specific agonists.

Murine platelets were isolated from wild-type (WT) and PECAM-1−/− (KO) mice, washed, and resuspended at a concentration of 2.5 × 108 platelets/mL. Stirred platelets (200 μL) were exposed to fibrillar collagen in absence of Mg2+ at a concentration of 1 μg/mL (A) or 10 μg/mL (B), to CRP at a concentration of 0.5 μg/mL (C) or 5 μg/mL (D), or to thrombin at a concentration of 0.1 (E) or 1 (F) U/mL. Changes in light transmission of the stimulated platelet suspension were measured in an aggregometer. Similar results were obtained with 5 separate platelet preparations. Note that compared to wild-type platelets, PECAM-1−/− platelets showed hyper-aggregation in response to stimulation with low concentrations of GPVI-specific agonists but not in response to stimulation with high concentrations of the same agonists or thrombin.

Close modal

Effect of PECAM-1 deficiency on dense granule secretion

We have recently shown that co-ligation of PECAM-1 with the ITAM-bearing T-cell receptor on the surface of Jurkat T cells attenuates calcium mobilization from intracellular stores.20 The physiological consequences of this inhibition, however, have not been examined. Because platelet alpha and dense granule secretion also require the elevation of cytosolic calcium and because the GPVI-FcRγ-chain–mediated signaling acts through phospholipase Cγ2 (PLCγ2) to mobilize intracellular calcium ion stores,36 we hypothesized that, like adhesion and aggregation, the platelet release reaction should also be accentuated in PECAM-1–deficient platelets stimulated with GPVI-specific agonists.

To determine whether PECAM-1 deficiency has an effect on granule release, we compared the ability of wild-type and PECAM-1−/− platelets to secrete dense granule-derived ATP on stimulation of GPVI. As shown in Figure3, PECAM-1–deficient platelets released their granules at subthreshold doses of fibrillar collagen in the absence of Mg2+ (Figure 3A, left) and CRP (Figure 3B, left). ATP secretion was indistinguishable in response to either high- or low-dose thrombin (Figure 3C) or after the addition of higher concentrations of fibrillar collagen in the absence of Mg2+(Figure 3A, right) or CRP (Figure 3B, right). These data are consistent with the notion that PECAM-1 normally acts as a negative regulator of GPVI/FcRγ-chain–mediated granule release and that, in its absence, responses to such agonists are enhanced.

Fig. 3.

PECAM-1−/− platelets have a lower threshold for release of dense granular contents in response to stimulation through GPVI collagen receptor.

Platelets isolated from wild-type (open bars) and PECAM-1−/− (closed bars) mice were stimulated with varying concentrations of GPVI-specific agonists, including fibrillar collagen in absence of Mg2+ (A) and CRP (B), or with thrombin (C). The amount of ATP released was measured using the luciferin–luciferase assay as described in “Materials and methods.” The assays were performed in triplicate, and the results are expressed as the mean of triplicate measurements ± SD. Similar results were obtained from 3 independent experiments. Note that the release of dense granular contents in response to stimulation with low concentrations of GPVI-specific agonists was elevated in PECAM-1−/− platelets compared to the wild-type platelets.

Fig. 3.

PECAM-1−/− platelets have a lower threshold for release of dense granular contents in response to stimulation through GPVI collagen receptor.

Platelets isolated from wild-type (open bars) and PECAM-1−/− (closed bars) mice were stimulated with varying concentrations of GPVI-specific agonists, including fibrillar collagen in absence of Mg2+ (A) and CRP (B), or with thrombin (C). The amount of ATP released was measured using the luciferin–luciferase assay as described in “Materials and methods.” The assays were performed in triplicate, and the results are expressed as the mean of triplicate measurements ± SD. Similar results were obtained from 3 independent experiments. Note that the release of dense granular contents in response to stimulation with low concentrations of GPVI-specific agonists was elevated in PECAM-1−/− platelets compared to the wild-type platelets.

Close modal

After injury to the vascular endothelium, platelets adhere to and become activated by collagen fibers in the exposed subendothelium. Among the numerous platelet receptors for collagen that have been reported, the integrin α2β1 appears to play a major role in the initial adhesion to collagen,37 a concept that has been supported by the finding that antibodies specific for α2β1 block platelet adhesion to collagen38,39 and by the observation that α2β1-deficient patients have a mild bleeding disorder associated with defective collagen-induced platelet aggregation.40,41 The platelet-specific receptor GPVI has also been implicated in platelet responses to collagen because collagen-induced platelet activation and aggregation are also defective in GPVI-deficient patients.42 

GPVI is associated within the plane of the plasma membrane with the Fc-receptor γ-subunit,43 a 12-kd, disulfide-linked, ITAM-containing homodimer that normally functions as the signaling component of high-affinity cell surface receptors for IgG and IgE. In contrast to these immunoglobulin receptors, the GPVI–FcRγ-chain complex functions as an ITAM-containing adhesion receptor that supports platelet interactions with collagen and subsequently transduces signals that lead to platelet activation and aggregation.23,24,44-48 It has been proposed that the signaling pathways of PTK-binding, ITAM-containing activating receptors are regulated by ITIM-containing, PTP-binding inhibitory receptors.16 Because PECAM-1 is the only known ITIM-containing receptor in platelets,10 we used a genetic approach to investigate the possibility that PECAM-1 might regulate GPVI signaling. We observed that, in response to GPVI-specific interactions with collagen, platelets from PECAM-1−/−mice exhibited increased adhesion, hyper-aggregation, and a lowered threshold for dense granule release relative to wild-type platelets. These findings are consistent with the conclusion that the GPVI signal transduction pathway is under the control of an ITIM-containing inhibitory receptor and that the inhibitory receptor normally used is PECAM-1.

We could not, a priori, have predicted whether PECAM-1 deficiency would have a stimulatory or an inhibitory effect on collagen-induced, GPVI-mediated signal transduction. This is because the ability of an immunoglobulin–ITIM-bearing membrane receptor to have a positive rather than a negative regulatory effect is influenced by a number of factors, including the target specificity of the phosphatase recruited and the identity of the phosphatase substrates that are proximal to the receptor–PTP complex. The situation with PECAM-1 is complicated by the observations that, although the PECAM-1 ITIM clearly provides a docking site for the cytoplasmic PTP SHP-2,11-14,49-51tyrosine-phosphorylated PECAM-1 has also been reported to associate with SHP-113,49-51 and with the 5′-inositol phosphatase SHIP.51 The situation is further complicated by the observations that, depending on whether the pathway is regulated, SHP-1 and SHP-2 appear to be capable of conveying either positive or negative regulatory signals.20,52-59 Because the identity of the phosphatase recruited by PECAM-1 in response to GPVI signaling, the target substrates for SHP-1 and SHP-2 in general, and the subcellular location of these substrates relative to that of PECAM-1 are completely unknown, there was no way to predict the physiological consequences of PECAM-1 deficiency. Interestingly, Pasquet et al60 have recently found that SHP-1 becomes tyrosine phosphorylated after platelet activation by GPVI and associates with other tyrosine-phosphorylated proteins of 28, 32, 50, 70, and 130 kd, and they suggested that SHP-1 plays a functional role in platelet responses to collagen. The 130-kd co-precipitating band was not, however, identified as PECAM-1. The identity of the receptor(s) that recruit and activate SHP-1 under these conditions remains to be determined.

The observation that PECAM-1−/− platelets are hyperresponsive to collagen stimulation represents the first demonstration of an in vivo cellular phenotype associated with PECAM-1 deficiency. Initial characterization of PECAM-1−/− mice by Duncan et al21 was carried out before the realization that PECAM-1 contained a functional ITIM and focused on its role as an adhesion rather than a signaling molecule. In models of leukocyte transendothelial migration, they found an accumulation of neutrophils at the perivascular basement membrane; however, the extent of leukocyte infiltration into the peritoneal cavity in response to an inflammatory stimulus was normal, as was T-cell homing to lymphoid organs and to the site of cutaneous hypersensitivity. Relevant to the current investigation, these authors found that platelet responses to adenosine diphosphate (ADP) are indistinguishable in PECAM-1 knockout versus wild-type mice. However, because ADP-induced platelet activation is mediated by G–protein-coupled rather than ITAM-coupled receptors,61,62 platelet responses to ADP would not be expected to be regulated by the PECAM-1/SHP-2 complex. The current study expands on the initial observations by showing that PECAM-1 deficiency affects platelet responses to the ITAM-containing GPVI-FcRγ-chain collagen receptor complex, but not to G–protein-coupled receptors for agonists such as thrombin or ADP. These findings are consistent with our recent observation that in vitro co-ligation of PECAM-1 with the ITAM-bearing T-cell antigen receptor (TCR) inhibits TCR-induced calcium-mobilization from intracellular stores.20 

Our finding that platelets from PECAM-1−/− mice show hyper-aggregation to collagen stimulation might lead one to predict that bleeding times in PECAM-1–deficient mice would be shorter than in wild-type mice. Interestingly, we have recently shown that tail vein bleeding times in PECAM-1−/− mice are mildlyprolonged.63 These apparently paradoxic findings were resolved by bone marrow transplantation studies showing that the prolonged tail vein bleeding time in PECAM-1−/−mice is caused by a vascular rather than a platelet function abnormality.63 It is likely that skin template bleeding time, which, unlike the tail vein bleeding time commonly studied in the mouse, solely measures platelet function, would be shorter in PECAM-1−/− mice than in wild-type mice.

Despite the rapid formation of the PECAM-1/SHP-2 signaling complex during platelet aggregation,12 virtually nothing is known about the physiologic consequences of this association. One potential mechanism by which a PECAM-1/PTP signaling complex might interfere with GPVI-dependent platelet activation may involve dephosphorylation of PTKs and PTK substrates that become recruited to the nearby ITAM-bearing GPVI-FcRγ-chain collagen receptor complex. Consistent with this potential mechanism, it is notable that the same members of the Src family of PTKs that become activated on GPVI stimulation are also responsible for tyrosine phosphorylation of PECAM-1.32 Signaling molecules that might represent candidate substrates might include those that are shared by GPVI and TCR signaling pathways; we have recently shown that PECAM-1 becomes tyrosine phosphorylated, forms a complex with SHP-2, and attenuates calcium release from intracellular stores in T cells on co-ligation of the TCR with PECAM-1.20 The SH2 domain–containing leukocyte protein of 76 kd (SLP-76), originally identified as a component of the TCR signal transduction pathway,64 is one such candidate substrate. It has been shown to be an essential intermediate between Syk phosphorylation and PLCγ2 activation in platelet responses to collagen.65,66 Similarly, the linker for activation of T cells (LAT), a 36- to 38-kd adaptor molecule in TCR signaling,67 also seems to be involved in signal transduction mediated by the GPVI-FcRγ-chain complex because platelets from LAT−/− mice exhibit decreased PLCγ2 tyrosine phosphorylation and are impaired in their ability to expose P-selectin and to bind fibrinogen in response to stimulation through the GPVI collagen receptor.68 Studies are in progress to determine whether components common to the GPVI and TCR signal transduction pathways serve as substrates for dephosphorylation by the PECAM-1/SHP-2 signaling complex. Elucidation of the signaling pathways affected by PECAM-1 is likely to be important in furthering our understanding of the role played by this novel member of the immunoglobulin–ITIM family in vascular cell biology.

We thank Drs Gordon S. Duncan and Tak W. Mak (Amgen Institute, Toronto, ON, Canada) for providing PECAM-1–deficient mice.

Supported by National Institutes of Health grant P01 HL44612 (D.K.N., P.J.N.). S.P. is a recipient of a Northwestern Mutual Life Fellowship in Biomedicine awarded to the Medical College of Wisconsin.

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

1
Newman
PJ
Berndt
MC
Gorski
J
et al
PECAM-1 (CD31) cloning and relation to adhesion molecules of the immunoglobulin gene superfamily.
Science.
247
1990
1219
1222
2
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
18570
3
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
11098
4
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
1064
5
Muller
WA
Weigl
SA
Deng
X
Phillips
DM
PECAM-1 is required for transendothelial migration of leukocytes.
J Exp Med.
178
1993
449
460
6
Vaporciyan
AA
DeLisser
HM
Yan
H-C
et al
Involvement of platelet endothelial cell adhesion molecule-1 in neutrophil recruitment in vivo.
Science.
262
1993
1580
1582
7
Zhou
Z
Christofidou-Solomidou
M
Garlanda
C
DeLisser
HM
Antibody against murine PECAM-1 inhibits tumor angiogenesis in mice.
Angiogenesis.
3
1999
181
188
8
Treutiger
CJ
Heddini
A
Fernandez
V
Muller
WA
Wahlgren
M
PECAM-1/CD31, an endothelial receptor for binding Plasmodium falciparuminfected erythrocytes.
Nat Med.
3
1997
1405
1408
9
Chen
Q
Heddini
A
Barragan
A
Fernandez
V
Pearce
SF
Wahlgren
M
The semiconserved head structure of Plasmodium falciparum erythrocyte membrane protein 1 mediates binding to multiple independent host receptors.
J Exp Med.
192
2000
1
10
10
Newman
PJ
Switched at birth: a new family for PECAM-1.
J Clin Invest.
103
1999
5
9
11
Jackson
DE
Kupcho
KR
Newman
PJ
Characterization of phosphotyrosine binding motifs in the cytoplasmic domain of platelet/endothelial cell adhesion molecule-1 (PECAM-1) that are required for the cellular association and activation of the protein-tyrosine phosphatase, SHP-2.
J Biol Chem.
272
1997
24868
24875
12
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
6993
13
Sagawa
K
Kimura
T
Swieter
M
Siraganian
RP
The protein-tyrosine phosphatase SHP-2 associates with tyrosine-phosphorylated adhesion molecule PECAM-1 (CD31).
J Biol Chem.
272
1997
31086
31091
14
Masuda
M
Osawa
M
Shigematsu
H
Harada
N
Fujiwara
K
Platelet endothelial cell adhesion molecule-1 is a major SH-PTP2 binding protein in vascular endothelial cells.
FEBS Lett.
408
1997
331
336
15
Cambier
JC
Inhibitory receptors abound?
Proc Natl Acad Sci U S A.
94
1997
5993
5995
16
Vely
F
Vivier
E
Conservation of structural features reveals the existence of a large family of inhibitory cell surface receptors and noninhibitory/activatory counterparts.
J Immunol.
159
1997
2075
2077
17
Reth
M
Antigen receptor tail clue [letter].
Nature.
338
1989
383
384
18
Ravetch
JV
Fc receptors.
Curr Opin Immunol.
9
1997
121
125
19
Watson
SP
Gibbins
J
Collagen receptor signalling in platelets: extending the role of the ITAM.
Immunol Today.
19
1998
260
264
20
Newton-Nash
DK
Newman
PJ
A new role for platelet-endothelial cell adhesion molecule-1 (CD31): inhibition of TCR-mediated signal transduction.
J Immunol.
163
1999
682
688
21
Duncan
GS
Andrew
DP
Takimoto
H
et al
Genetic evidence for functional redundancy of platelet/endothelial cell adhesion molecule-1 (PECAM-1): CD31 deficient mice reveal PECAM-1 dependent and PECAM-1 independent functions.
J Immunol.
162
1999
3022
3030
22
Morton
LF
Hargreaves
PG
Farndale
RW
Young
RD
Barnes
MJ
Integrin α2β1-independent activation of platelets by simple collagen-like peptides: collagen tertiary (triple helical) and quaternary (polymeric) structures are sufficient alone for α2β1-independent platelet reactivity.
Biochem J.
306
1995
337
344
23
Gibbins
JM
Okuma
M
Farndale
R
Barnes
M
Watson
SP
Glycoprotein VI is the collagen receptor in platelets which underlies tyrosine phosphorylation of the Fc receptor gamma-chain.
FEBS Lett.
413
1997
255
259
24
Asselin
J
Gibbins
JM
Achison
M
et al
A collagen-like peptide stimulates tyrosine phosphorylation of syk and phospholipase Cγ2 in platelets independent of integrin α2β1.
Blood.
89
1997
1235
1242
25
Gibbins
JM
Briddon
S
Shutes
A
et al
The p85 subunit of phosphatidylinositol 3-kinase associates with the Fc receptor gamma-chain and linker for activator of T cells (LAT) in platelets stimulated by collagen and convulxin.
J Biol Chem.
273
1998
34437
34443
26
Gross
BS
Lee
JR
Clements
JL
et al
Tyrosine phosphorylation of SLP-76 is downstream of Syk following stimulation of the collagen receptor in platelets.
J Biol Chem.
274
1999
5963
5971
27
Inoue
K
Ozaki
Y
Satoh
K
et al
Signal transduction pathways mediated by glycoprotein Ia/IIa in human platelets: comparison with those of glycoprotein VI.
Biochem Biophys Res Commun.
256
1999
114
120
28
Briddon
SJ
Melford
SK
Turner
M
Tybulewicz
V
Watson
SP
Collagen mediates changes in intracellular calcium in primary mouse megakaryocytes through syk-dependent and -independent pathways.
Blood.
93
1999
3847
3855
29
Pasquet
JM
Gross
B
Quek
L
et al
LAT is required for tyrosine phosphorylation of phospholipase Cγ2 and platelet activation by the collagen receptor GPVI.
Mol Cell Biol.
19
1999
8326
8334
30
Oda
A
Ikeda
Y
Ochs
HD
et al
Rapid tyrosine phosphorylation and activation of Bruton's tyrosine/Tec kinases in platelets induced by collagen binding or CD32 cross-linking.
Blood.
95
2000
1663
1670
31
Wang
R
Kupcho
KR
Armstrong
MJ
Sun
Q-H
Paddock
C
Newman
PJ
PECAM-1-mediated signal transduction can be initiated by cell adhesion to extracellular matrix [abstract].
Blood.
92
1998
29a
32
Cicmil
M
Thomas
JM
Sage
T
et al
Collagen, convulxin, and thrombin stimulate aggregation-independent tyrosine phosphorylation of cd31 in platelets: evidence for the involvement of Src family kinases.
J Biol Chem.
275
2000
27339
27347
33
Nakamura
T
Jamieson
GA
Okuma
M
Kambayashi
J
Tandon
NN
Platelet adhesion to native type I collagen fibrils: role of GPVI in divalent cation-dependent and -independent adhesion and thromboxane A2 generation.
J Biol Chem.
273
1998
4338
4344
34
Neel
BG
Role of phosphatases in lymphocyte activation.
Curr Opin Immunol.
9
1997
405
420
35
Vu
T-KH
Hung
DT
Wheaton
VI
Coughlin
SR
Molecular cloning of a functional thrombin receptor reveals a novel proteolytic mechanism of receptor activation.
Cell.
64
1991
1057
1068
36
Watson
SP
Gibbins
JM
Collagen receptor signaling in platelets: extending the role of the ITAM.
Immunol Today.
19
1998
260
265
37
Santoro
SA
Identification of a 160,000 dalton platelet membrane protein that mediates the initial divalent cation-dependent adhesion of platelets to collagen.
Cell.
46
1986
913
920
38
Kunicki
TJ
Nugent
DJ
Staats
SJ
Orchekowski
RP
Wayner
EA
Carter
WG
The human fibroblast class II extracellular matrix receptor mediates platelet adhesion to collagen and is identical to the platelet glycoprotein Ia-IIa complex.
J Biol Chem.
263
1988
4516
4519
39
Coller
BS
Beer
JH
Scudder
LE
Steinberg
MH
Collagen-platelet interactions: evidence for a direct interaction of collagen with platelet GPIa/IIa and an indirect interaction with platelet GPIIb/IIIa mediated by adhesive proteins.
Blood.
74
1989
182
192
40
Nieuwenhuis
HK
Akkerman
JWN
Houdijk
WPM
Sixma
JJ
Human blood platelets showing no response to collagen fail to express surface glycoprotein Ia.
Nature.
318
1985
470
472
41
Kehrel
B
Balleisen
L
Kokott
R
et al
Deficiency of intact thrombospondin and membrane glycoprotein Ia in platelets with defective collagen-induced aggregation and spontaneous loss of disorder.
Blood.
71
1988
1074
1078
42
Moroi
M
Jung
SM
Okuma
M
Shinmyozu
K
A patient with platelets deficient in glycoprotein VI that lack both collagen-induced aggregation and adhesion.
J Clin Invest.
84
1989
1440
1445
43
Tsuji
M
Ezumi
Y
Arai
M
Takayama
H
A novel association of Fc receptor gamma-chain with glycoprotein VI and their co-expression as a collagen receptor in human platelets.
J Biol Chem.
272
1997
23528
23531
44
Ezumi
Y
Shindoh
K
Tsuji
M
Takayama
H
Physical and functional association of the Src family kinases Fyn and Lyn with the collagen receptor glycoprotein VI-Fc receptor gamma chain complex on human platelets.
J Exp Med.
188
1998
267
276
45
Briddon
SJ
Watson
SP
Evidence for the involvement of p59fyn and p53/56lyn in collagen receptor signalling in human platelets.
Biochem J.
338
1999
203
209
46
Gibbins
JM
Asselin
J
Farndale
R
Barnes
M
Law
C-L
Watson
SP
Tyrosine phosphorylation of the Fc receptor γ-chain in collagen-stimulated platelets.
J Biol Chem.
271
1996
18095
18099
47
Melford
SK
Turner
M
Briddon
SJ
Tybulewicz
VL
Watson
SP
Syk and Fyn are required by mouse megakaryocytes for the rise in intracellular calcium induced by a collagen-related peptide.
J Biol Chem.
272
1997
27539
27542
48
Polgar
J
Clemetson
JM
Kehrel
BE
et al
Platelet activation and signal transduction by convulxin, a C-type lectin from Crotalus durissus terrificus (tropical rattlesnake) venom via the p62/GPVI collagen receptor.
J Biol Chem.
272
1997
13576
13583
49
Hua
CT
Gamble
JR
Vadas
MA
Jackson
DE
Recruitment and activation of SHP-1 protein-tyrosine phosphatase by human platelet endothelial cell adhesion molecule-1 (PECAM-1).
J Biol Chem.
273
1998
28332
28340
50
Cao
MY
Huber
M
Beauchemin
N
Famiglietti
J
Albelda
SM
Veillette
A
Regulation of mouse PECAM-1 tyrosine phosphorylation by the Src and Csk families of protein-tyrosine kinases.
J Biol Chem.
273
1998
15765
15772
51
Pumphrey
NJ
Taylor
V
Freeman
S
et al
Differential association of cytoplasmic signalling molecules SHP-1, SHP-2, SHIP and phospholipase Cγ1 with PECAM-1/CD31.
FEBS Lett.
450
1999
77
83
52
Neel
BG
Tonks
NK
Protein tyrosine phosphatases in signal transduction.
Curr Opin Cell Biol.
9
1997
193
204
53
Stein-Gerlach
M
Wallasch
C
Ullrich
A
SHP-2, SH2-containing protein tyrosine phosphatase-2.
Int J Biochem Cell Biol.
30
1998
559
566
54
Feng
GS
SHP-2 tyrosine phosphatase: signaling one cell or many.
Exp Cell Res.
253
1999
47
54
55
Somani
AK
Bignon
JS
Mills
GB
Siminovitch
KA
Branch
DR
Src kinase activity is regulated by the SHP-1 protein-tyrosine phosphatase.
J Biol Chem.
272
1997
21113
21119
56
Brockdorff
J
Williams
S
Couture
C
Mustelin
T
Dephosphorylation of ZAP-70 and inhibition of T cell activation by activated SHP-1.
Eur J Immunol.
29
1999
2539
2550
57
Symes
A
Stahl
N
Reeves
SA
et al
The protein tyrosin phosphatase SHP-2 negatively regulates ciliary neurotrophic induction of gene expression.
Curr Biol.
7
1997
697
700
58
Shi
Z-Q
Lu
W
Feng
G-S
The SHP-2 tyrosine phosphatase has opposite effects in mediating the activation of extracellular signal-regulated and c-Jun NH2-terminal mitogen-activated protein kinases.
J Biol Chem.
273
1998
4904
4908
59
You
M
Yu
DH
Feng
GS
Shp-2 tyrosine phosphatase functions as a negative regulator of the interferon-stimulated Jak/STAT pathway.
Mol Cell Biol.
19
1999
2416
2424
60
Pasquet
J
Quek
L
Pasquet
S
et al
Evidence for a role of SHP-1 in platelet activation by the collagen receptor GPVI.
J Biol Chem.
275
2000
28526
28531
61
Ayyanathan
K
Webbs
TE
Sandhu
AK
Athwal
RS
Barnard
EA
Kunapuli
SP
Cloning and chromosomal localization of the human P2Y1 purinoceptor 35.
Biochem Biophys Res Commun.
218
1996
783
788
62
Daniel
JL
Dangelmaier
C
Jin
J
Ashby
B
Smith
JB
Kunapuli
SP
Molecular basis for ADPinduced platelet activation, I: evidence for three distinct ADP receptors on human platelets.
J Biol Chem.
273
1998
2024
2029
63
Mahooti
S
Graesser
D
Patil
S
et al
PECAM-1 (CD31) expression modulates bleeding time in vivo.
Am J Pathol.
157
2000
75
81
64
Jackman
JK
Motto
DG
Sun
Q
et al
Molecular cloning of SLP-76, a 76-kDa tyrosine phosphoprotein associated with Grb2 in T cells.
J Biol Chem.
270
1995
7029
7032
65
Gross
BS
Lee
JR
Clements
JL
et al
Tyrosine phosphorylation of SLP-76 is downstream of syk following stimulation of the collagen receptor in platelets.
J Biol Chem.
274
1999
5963
5971
66
Clements
JL
Lee
JR
Gross
B
et al
Fetal hemorrhage and platelet dysfunction in SLP-76–deficient mice.
J Clin Invest.
103
1999
19
25
67
Zhang
W
Sloan-Lancaster
J
Kitchen
J
Trible
RP
Samelson
LE
LAT: the ZAP-70 tyrosine kinase substrate that links T cell receptor to cellular activation.
Cell.
92
1998
83
92
68
Pasquet
JM
Gross
B
Quek
L
et al
LAT is required for tyrosine phosphorylation of phospholipase Cγ2 and platelet activation by the collagen receptor GPVI.
Mol Cell Biol.
19
1999
8326
8334

Author notes

Peter J. Newman, Blood Research Institute, The Blood Center of Southeastern Wisconsin, 638 N. 18th St, PO Box 2178, Milwaukee, WI 53233; e-mail: pjnewman@bcsew.edu.

Sign in via your Institution