当前位置: > 正文
编号:11371633
Critical roles for the COOH-terminal NITY and RGT sequences of the int
http://www.100md.com 《细胞学杂志》
     Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612

    Address correspondence to Xiaoping Du, Department of Pharmacology, College of Medicine, University of Illinois at Chicago, 835 South Wolcott Ave., Chicago, IL 60612. Tel.: (312) 355-0237. Fax: (312) 996-1225. E-mail: xdu@uic.edu

    Abstract

    Bidirectional signaling of integrin IIb?3 requires the ?3 cytoplasmic domain. To determine the sequence in the ?3 cytoplasmic domain that is critical to integrin signaling, cell lines were established that coexpress the platelet receptor for von Willebrand factor (vWF), glycoprotein Ib-IX, integrin IIb, and mutants of ?3 with truncations at sites COOH terminal to T741, Y747, F754, and Y759. Truncation at Y759 did not affect integrin activation, as indicated by vWF-induced fibrinogen binding, but affected cell spreading and stable adhesion. Thus, the COOH-terminal RGT sequence of ?3 is important for outside-in signaling but not inside-out signaling. In contrast, truncation at F754, Y747, or T741 completely abolished integrin activation. A point mutation replacing Y759 with alanine also abolished integrin activation. Thus, the T755NITY759 sequence of ?3, containing an NXXY motif, is critical to inside-out signaling, whereas the intact COOH terminus is important for outside-in signaling. In addition, we found that the calcium-dependent protease calpain preferentially cleaves at Y759 in a population of ?3 during platelet aggregation and adhesion, suggesting that calpain may selectively regulate integrin outside-in signaling.

    Key Words: integrin; platelet; calpain; signaling; platelet activation

    * Abbreviation used in this paper: vWf, von Willebrand factor.

    Introduction

    The prototype integrin, IIb?3, plays critical roles in platelet adhesion and aggregation. Normally, IIb?3 on circulating platelets is present in a "resting" form, with a low affinity for its ligands such as soluble fibrinogen and von Willebrand factor (vWF).* Upon vascular injury, exposure of platelets to soluble agonists, such as thrombin and ADP, or extracellular matrix adhesive proteins, such as collagen and vWF, induces "inside-out" signals activating the ligand-binding function of IIb?3 (Ginsberg et al., 1992; Shattil et al., 1998; Parise, 1999). Ligand binding to IIb?3 not only forms adhesive bonds between platelets and adhesive ligands, but also transmits "outside-in" signals to induce a series of cellular responses, such as protein phosphorylation (Ferrell and Martin, 1989; Golden et al., 1990; Lipfert et al., 1992; Clark et al., 1994; Law et al., 1996; Lerea et al., 1999), elevation of intracellular Ca2+ (Pelletier et al., 1992), and cytoskeleton reorganization (Phillips et al., 1980), leading to cell spreading, stabilization of cell adhesion, and secondary wave of platelet aggregation (Shattil et al., 1998; Parise, 1999).

    Inside-out and outside-in signaling of integrins have been suggested to involve the interaction between the cytoplasmic domains of integrins and intracellular signaling molecules. Interactions between the membrane-proximal regions of the integrin IIb and ?3 cytoplasmic domains are important in maintaining a resting conformation of the integrin, and inside-out signaling is associated with disruption of this interaction (Hughes et al., 1996; Vinogradova et al., 2002). The cytoplasmic domain of IIb?3 interacts with several intracellular proteins, including cytoskeletal proteins such as talin (Calderwood et al., 1999; Knezevic et al., 1996) and myosin (Phillips et al., 2001), calcium-binding protein CIB (Naik et al., 1997), phosphotyrosine-binding proteins SHC and GRB2 (Law et al., 1996), protein kinases such as integrin-linked protein kinase (Hannigan et al., 1996), and ?3 endonexin (Shattil et al., 1995). Binding of talin (Calderwood et al., 1999, 2002) and ?3 endonexin (Kashiwagi et al., 1997) to ?3 has been implicated in promoting integrin activation. Binding of phosphotyrosine-binding proteins and focal adhesion kinase has been suggested to play roles in outside-in signaling (Law et al., 1996). Also, the ?3 cytoplasmic domain can be chemically modified during platelet activation. For example, T753 (Lerea et al., 1999), Y747, and Y759 (Law et al., 1996) are phosphorylated by protein kinases, and the COOH-terminal region of ?3 is cleaved by calpain at the COOH-terminal side of Y741, T747, F754, and Y759 (Du et al., 1995). Phosphorylation at both Y747 and Y759 is critical to outside-in signaling of the integrin (Law et al., 1999). The roles of calpain cleavage of the ?3 cytoplasmic domain in bidirectional signaling of the integrin, however, are not clear.

    The requirement for specific sequences in the ?3 cytoplasmic domain in outside-in integrin signaling has been indicated by mutagenesis studies in transfected CHO cell models (Chen et al., 1994; Hughes et al., 1995; Ylanne et al., 1995; Patil et al., 1999) and transgenic mouse models (Law et al., 1999). Deletions or mutations that disrupted the two NXXY motifs in the cytoplasmic domain of the integrin abolished outside-in signal-dependent integrin functions such as stable cell adhesion and cell spreading (Ylanne et al., 1993, 1995; Chen et al., 1994; Patil et al., 1999). In a transgenic mouse model, mutations replacing both Y747 and Y759 in the NXXY motifs with phenylalanine to disrupt tyrosine phosphorylation inhibited outside-in signaling but had no significant effect on inside-out signaling (Law et al., 1999). The requirement for the cytoplasmic domain of ?3 in inside-out signaling has been indicated by defective integrin activation in variant Glanzmann's thrombasthenia patients whose ?3 cytoplasmic domain either has a point mutation replacing S752 with proline (Chen et al., 1992, 1994) or has a truncation mutation at R724 (Wang et al., 1997). However, mapping of functional sites important to inside-out signaling has been hampered by the lack of a suitable inside-out signaling model in cultured cells deficient in endogenous wild-type ?3, as recombinant integrin IIb?3 expressed in CHO cells is not activated by platelet agonists such as thrombin and ADP (O'Toole et al., 1990). Although constitutively active integrin mutants have been useful in characterizing the affinity regulation by integrin cytoplasmic domains (O'Toole et al., 1991, 1994; Hughes et al., 1995), these mutants obviously bypass the normal on–off switch mechanism of inside-out signaling. We and others have recently shown that integrin can be activated in a reconstituted CHO cell model via the GPIb-IX pathway (Gu et al., 1999; Zaffran et al., 2000; Li et al., 2001). In this model, and as shown here, binding of vWF to GPIb-IX induces inside-out signaling and results in activation of the fibrinogen-binding function of IIb?3, which replicates the GPIb-IX–induced inside-out signaling mechanism in platelets. To understand the structural requirement of the ?3 cytoplasmic domain in integrin inside-out signaling and the role of calpain cleavage in regulating integrin signaling, we have expressed different truncation mutants of ?3 in this reconstituted integrin activation model. We show that removal of the NITY sequence abolished inside-out signaling, whereas truncation of RGT sequence COOH terminal to the NITY motif reduced outside-in signaling without affecting inside-out signaling. Furthermore, a point mutation changing Y759 to alanine abolished inside-out signaling and reduced outside-in signaling. Thus, the NITY sequence is essential for both inside-out and outside-in signaling of IIb?3, and the RGT sequence is important for outside-in signaling. Furthermore, localized calpain cleavage of ?3 during platelet activation mainly occurs at a site COOH terminal to Y759, suggesting that calpain cleavage may selectively regulate outside-in signaling.

    Results

    CHO cell lines coexpressing mutants of the human integrin IIb?3 and GPIb-IX

    To study the roles of the COOH-terminal region of ?3 in integrin signaling, we have generated four truncation mutants of the ?3 subunit with truncations at sites COOH terminal to T741, Y747, F754, and Y759. Truncations at these sites also coincide with previously identified calpain cleavage sites in ?3 (Du et al., 1995; Fig. 1 A). These mutants were cotransfected with the integrin IIb subunit into a CHO cell line that expresses recombinant human GPIb-IX complex (1b9). Complex formation between these ?3 mutants and IIb was verified by flow cytometry using an IIb?3 complex–specific monoclonal antibody, D57. Stable cell lines expressing both GPIb-IX and integrin mutants (1b9/741, 1b9/747, 1b9/754, and 1b9/759) were obtained by cell sorting with antibodies specific for integrin IIb?3 and GPIb-IX. Expression of correct mutants at each of the four sites was verified by immunoblotting with antibodies that specifically recognize each of these sites only when the site is truncated (Du et al., 1995; Fig. 1 B). In addition, a cell line coexpressing GPIb-IX and a mutant IIb?3 bearing alanine substitution of Y759 in ?3 was also established (1b9/Y759A). Expression levels of GPIb-IX and integrin IIb?3 on the above mutant cell lines were comparable with a cell line expressing GPIb-IX and wild-type integrin IIb?3 (123 cells) at the time of experiments (Fig. 1 C).

    Figure 1. Truncation mutants of IIb?3 coexpressed with GPIb-IX in CHO cells. (A) Schematics showing truncation mutants of ?3 that mimic calpain cleavage at four previously identified sites. A Y759A point mutation is also depicted. (B) The mutants described in A were cotransfected with IIb in a CHO cell line already expressing GPIb-IX, and stable cell lines were established. Expression of correct truncation mutants in each of the cell lines was verified by immunoblotting cell lysates with antibodies specifically recognizing calpain-cleaved forms of ?3 (Ab 741, Ab 747, Ab 754, and Ab 759) and with an antibody recognizing the COOH terminus of ?3 (Ab 762). A cell line expressing wild-type IIb?3 and GPIb-IX (123) was also immunoblotted with these antibodies as a control. (C) Levels of surface expression of wild type or mutants of IIb?3 were examined by flow cytometry with an antibody recognizing IIb?3 complex (D57) (dotted lines), and levels of GPIb-IX expression were examined with an antibody against GPIb, SZ2 (shaded lines). Nonspecific mouse IgG was used as a negative control (solid lines). Please note that levels of expression of each of these mutants were comparable with 123 cells. Expression of GPIb-IX and integrin was also comparable with a cell line expressing GPIb-IX alone (1b9) and a cell line expressing IIb?3 alone (2b3a), respectively.

    Reconstituted CHO cell model of integrin IIb?3 activation

    We recently reported the reconstitution of GPIb-IX–mediated integrin activation in CHO cells expressing recombinant human GPIb-IX and integrin IIb?3 (123 cells) (Gu et al., 1999). In this reconstituted integrin activation model, vWF binding to GPIb-IX in the presence of ristocetin activates fibrinogen binding to the 123 cells in an RGDS-dependent manner, thus allowing the study of integrin inside-out signaling using a specific recombinant DNA approach. Although ?3-transfected CHO cells also express an v?3 integrin composed of endogenous v and recombinant ?3, Fig. 2 shows that vWF-induced fibrinogen binding to 123 cells is specifically inhibited by a monoclonal antibody that recognizes human IIb?3, but not by a blocking monoclonal antibody that recognizes v?3. Also, a cell line expressing GPIb-IX and ?3 without IIb failed to show RGDS-dependent fibrinogen binding under identical conditions (unpublished data). These results indicate that vWF-induced fibrinogen binding is mediated by IIb?3 but not v?3. Thus, vWF-induced fibrinogen binding to 123 cells specifically reflects the activation of the platelet integrin IIb?3.

    Figure 2. vWF-induced activation of fibrinogen binding to cell lines coexpressing GPIb-IX and IIb?3 reflects specific activation of integrin IIb?3. (A) CHO cells (CHO), CHO cells coexpressing GPIb-IX and integrin IIb?3 (123), and CHO cells coexpressing GPIb-IX and ?3 in the absence of IIb (1b9/?3) were incubated with control mouse IgG, IIb?3-specific antibodies, 2G12 and D57, and an antibody recognizing integrin v?3, anti-VNR1. The reactivity of these antibodies was detected by flow cytometry. (B) 123 cells were preincubated with 100 μg/ml of control mouse IgG, 2G12, or anti-VNR1 (previously shown to inhibit ligand-binding functions of IIb?3 or v?3, respectively) for 10 min at 22°C, and then Oregon green–labeled fibrinogen (30 μg/ml) was added and activated with vWF in the presence of ristocetin. Nonspecific binding was estimated by adding the integrin inhibitor RGDS. Quantitative results from three experiments (means ± SD) are expressed as fibrinogen binding index (total bound fibrinogen/nonspecifically bound fibrinogen in the presence of RGDS). Statistical differences between samples were analyzed using the RRISM software and the Repeated Measures ANOVA method.

    Effects of ?3 mutations on integrin activation (inside-out signaling)

    To understand the roles of the COOH-terminal region of ?3 in inside-out signaling of IIb?3, we examined vWF-induced fibrinogen binding to cell lines expressing GPIb-IX and truncation mutants of IIb?3 (1b9/741, 1b9/747, 1b9/754, and 1b9/759). Fig. 3 shows that truncation of the ?3 cytoplasmic domain at the site COOH terminal to Y759 did not affect vWF-induced activation of fibrinogen binding to integrin IIb?3. In contrast, truncations at F754, Y747, or T741 abolished vWF-induced integrin activation. Thus, it appears that the RGT sequence at the COOH terminus of the ?3 cytoplasmic domain is not required for integrin activation, whereas the sequence between F754 and Y759, which contains an NXXY motif, is required for integrin activation. To further determine if the NXXY motif is important in integrin activation, we examined vWF-induced fibrinogen binding to CHO cells coexpressing GPIb-IX and the Y759A mutant of integrin IIb?3. Indeed, vWF-induced fibrinogen binding was abolished in the Y759A mutant, indicating that the NXXY motif, particularly Y759, is required for vWF-induced activation of IIb?3.

    Figure 3. vWF-induced fibrinogen binding to wild type or various mutants of IIb?3. (A) Cells (123, 1b9/741, 1b9/747, 1b9/754, 1b9/759, and 1b9/Y759A) suspended in modified Tyrode's buffer were incubated at 22°C for 30 min with Oregon green–labeled fibrinogen (30 μg/ml) and 1 mg/ml of ristocetin with (right) or without (left) adding 25 μg/ml of vWF, and then were analyzed for fluorescence intensity by flow cytometry (solid lines). Nonspecific binding of fibrinogen was estimated by adding 1 mM RGDS (shaded lines). (B) Quantitative results from three experiments (means ± SD) are expressed as fibrinogen binding index (total bound fluorescence intensity/nonspecifically bound fluorescence intensity).

    Fibrinogen-binding function of the integrin IIb?3 mutants

    A possible alternative interpretation to the above results is that the activation-defective mutants of IIb?3 (754, 747, and 741) may have a loss of function in the extracellular ligand-binding site. To exclude this possibility, we examined the fibrinogen-binding function of these mutants. It has been demonstrated previously (Du et al., 1991) that binding of the ligand mimetic peptide, RGDS, to the extracellular ligand-binding domain of IIb?3 transforms the integrin into an active conformation. After fixation and removal of RGDS, the activated integrin is able to bind fibrinogen. Thus, we examined RGDS-induced fibrinogen binding to cells expressing mutants of IIb?3 (Fig. 4). All of the above mutant cell lines bound fibrinogen in a manner similar to wild-type integrin IIb?3 (123 cells). In contrast, 1b9 cells, in which no integrin IIb?3 was expressed, did not bind fibrinogen after RGDS treatment. These data suggest that the activation-defective mutants used in this study retained fibrinogen-binding function. Therefore, the inability of these mutants to bind fibrinogen after vWF stimulation results from a deficiency in inside-out signaling.

    Figure 4. Fibrinogen binding to mutants of IIb?3 pretreated with RGDS peptide. Cells coexpressing GPIb-IX and wild-type IIb?3 (123) or integrin mutants were preincubated with or without 1 mM of RGDS peptide for 10 min and then fixed with paraformaldehyde (1% in PBS) for 30 min at 22°C. As a negative control, cells expressing GPIb-IX alone (1b9) were identically treated. The cells were washed, resuspended in modified Tyrode's buffer at a concentration of 5 x 105/ml, and then incubated with Oregon green–labeled fibrinogen for 30 min. Fibrinogen binding was detected by flow cytometry. Nonspecific binding was estimated by adding RGDS peptide to the assay. Specific fibrinogen binding was determined by subtracting the geometric means of fluorescence intensity of the nonspecific binding from that of total binding. Shown in the figure are the data (mean ± SD) from three experiments.

    Effect of mutations on cell adhesion and spreading under static conditions

    An important function for IIb?3 is to mediate stable platelet adhesion and spreading on immobilized integrin ligands such as vWF and fibrinogen. Integrin-dependent stable cell adhesion and spreading on vWF are significantly enhanced by inside-out signaling induced by GPIb-IX–vWF interaction (Savage et al., 1992) and also require integrin outside-in signaling. Integrin IIb?3–mediated stable cell adhesion and spreading to immobilized fibrinogen, however, do not require inside-out signaling (Coller, 1980; Savage et al., 1992) but are dependent upon ligand-induced integrin activation and outside-in signaling (Du et al., 1991; Phillips et al., 1991; Ginsberg et al., 1995; Shattil et al., 1998). Thus, to determine the roles of the COOH-terminal region of ?3 in outside-in and inside-out signaling, we examined stable adhesion and spreading of the mutant cell lines to fibrinogen and vWF. As previously reported (Gu et al., 1999), CHO cells (without transfected GPIb-IX and integrin IIb?3) poorly adhere to vWF or fibrinogen. CHO cells expressing GPIb-IX alone also poorly adhere to both vWF and fibrinogen (Fig. 5). Even in the presence of botrocetin, which enhances the GPIb-IX binding affinity of vWF and allows GPIb-IX–dependent stable cell adhesion to vWF, these cells poorly spread on vWF (unpublished data), indicating that ?3 integrin is required for cell spreading on vWF (Gu et al., 1999). As expected, CHO cells expressing integrin IIb?3 (2b3a) showed high levels of adhesion and spreading on fibrinogen but poorly adhered to vWF within the first hour of incubation (Fig. 5). However, with prolonged incubation, adhesion of 2b3a cells to vWF was slowly increased (unpublished data), suggesting a low affinity interaction between ?3 integrins and vWF. Coexpression of GPIb-IX with IIb?3 (123 cells) significantly enhanced and accelerated integrin-dependent cell adhesion and spreading on vWF, indicating that GPIb-IX activates integrin, leading to integrin-dependent cell spreading and stable adhesion. Cleavage of ?3 at or NH2 terminal to F754 inhibited integrin-dependent stable cell adhesion and spreading on vWF and fibrinogen (Fig. 5), which is consistent with previous findings that this region of the integrin cytoplasmic domain is important for adhesion and spreading (Ylanne et al., 1993, 1995). Interestingly, truncation of ?3 at Y759 also showed a reduced stable adhesion to both vWF and fibrinogen, and most 1b9/759 cells that adhered to fibrinogen or vWF spread poorly (Fig. 5). Reduced stable adhesion in 1b9/759 cells was not caused by a defect in inside-out signaling, as IIb?3-dependent cell adhesion to fibrinogen under these conditions does not require inside-out signaling, and 1b9/759 cells showed normal inside-out signaling, as indicated by soluble fibrinogen binding (Fig. 3). Reduced stable adhesion of 1b9/759 cells is likely resultant from a decreased resistance to the washing procedure in the adhesion assay, as gentler washing can significantly increase 1b9/759 cell adhesion (unpublished data; Fig. 6). Thus, reduced cell adhesion and spreading in 1b9/759 cells suggest that integrin outside-in signaling in this truncation mutant is impaired.

    Figure 5. Effects of mutations on integrin-dependent stable cell adhesion and spreading on vWF or fibrinogen (Fg) under static conditions. The indicated cell lines in modified Tyrode's buffer (106/ml) were allowed to incubate in fibrinogen- or vWF-coated microtiter wells at 37°C for 1 h. The wells were washed three times, and the cells remaining adherent were quantitated using an acid phosphatase assay, as described under the Materials and methods (A), and photographed to document cell spreading (B). The means and standard deviations of triplicate samples are shown in A.

    Figure 6. Effects of mutations on GPIb-IX–induced, integrin-dependent cell adhesion to vWF under flow conditions. Cells coexpressing GPIb-IX with wild type and different mutants of IIb?3 as well as control cells expressing GPIb-IX alone or IIb?3 alone (5 x 106/ml in modified Tyrode's buffer containing 5% BSA) were perfused though the capillary tubes precoated with vWF at indicated shear rates for 2 min, followed by a further 10-min perfusion with Tyrode's buffer at the same shear rate to wash out transient adherent cells. Cell interaction with immobilized vWF under flow conditions was observed under an inverted microscope and recorded on videotapes. The number of stable adherent cells on vWF was counted on images obtained at 10 randomly selected positions in each vWF-coated capillary tube. The difference between the control 123 cells with each of the other cell lines was analyzed by t test and shown to be highly significant (P < 0.01), unless otherwise indicated.

    Effects of integrin ?3 mutations on cell adhesion under flow

    A major physiological role of GPIb-IX–induced integrin activation is to mediate stable platelet adhesion to immobilized vWF. Under flow conditions, initial transient platelet adhesion and rolling on vWF are mediated by GPIb-IX. Interaction of vWF with GPIb-IX induces integrin activation and integrin-dependent stable platelet adhesion. Thus, if the mutations of IIb?3 abolished integrin inside-out signaling, these mutants should also show defective stable adhesion to vWF under flow conditions. To determine this, the above-described cell lines were perfused through a capillary tube precoated with human vWF. At shear rates below 50 s-1, the cells expressing integrin mutants 1b9/754, 1b9/747, and 1b9/741 (all defective in inside-out signaling) were defective in stable cell adhesion to vWF compared with 123 cells expressing wild-type IIb?3 (Fig. 6). The amount of adherent 1b9/759 mutant cells on vWF, in contrast, was not reduced. Thus, the results in cell adhesion to vWF under this low shear rate condition mirrored integrin activation indicated by fibrinogen binding. However, when the flow shear rate was further increased to 100 s-1 or above, not only the integrin activation–deficient mutants, but also 1b9/759 cells showed significantly reduced adhesion to vWF, suggesting that the ability of 1b9/759 cells to resist shear force is impaired. This is consistent with the above results that 1b9/759 cells showed decreased adhesion in the static adhesion assay, which involves multiple washes with considerable shear force. As the vWF-induced ligand-binding function of the 759 mutant is normal, we conclude that the impaired outside-in signaling function of this mutant reduced the ability of 1b9/759 cells to resist shear stress. Together, these results indicate that the RGT sequence at the COOH terminus of ?3 is important for outside-in signaling of IIb?3, but the NITY sequence is important for both inside-out and outside-in signaling.

    Differential cleavage at different sites of the ?3 cytoplasmic domain by calpain in platelets

    We have shown previously that calpain may cleave the cytoplasmic domain of ?3 at sites COOH terminal to T741, Y747, F754, and Y759, which generates ?3 fragments identical to the above-described truncation mutants of ?3 (741, 747, 754, and 759). As we showed above that truncations at these different sites result in different functional effects on integrin signaling, our results also indicate that cleavage of the ?3 cytoplasmic domain at these different sites has the potential to differentially regulate outside-in and inside-out signaling of integrin IIb?3. To determine if calpain differentially cleaves the ?3 integrin at different sites during platelet activation, washed platelets were treated with or without thrombin (0.1 U/ml) and then immunoblotted for calpain cleavage by the cleavage-specific antibodies (Du et al., 1995). We found that anti-759 antibody, which only recognizes ?3 molecules with cleavage at Y759, reacted with 0.8% of the integrin IIb?3 molecules in washed "resting" platelets (Fig. 7). This reaction was unlikely to result from cross-reaction of this antibody with the intact ?3 subunit, because we showed that the antibody did not react with the intact ?3 subunit but reacted with the 759 mutant expressed in CHO cells (Fig. 2). Thus a very small percentage of the ?3 molecules in resting platelets has been cleaved at the Y759 site. Stimulation of platelets with thrombin caused a time-dependent and significant increase in the cleavage at Y759. In contrast to the 759 site, calpain cleavage at T754 or Y747 (Fig. 7) occurred to a much lesser degree and only after a much longer exposure to thrombin. Calpain cleavage at the 741 site was not detectable in thrombin-stimulated platelets (unpublished data) but was detected in platelets treated with calcium ionophore A23187 (Du et al., 1995). Thus, in thrombin-activated platelets, calpain preferentially cleaves ?3 at Y759. As we showed above that cleavage at Y759 selectively reduced integrin outside-in signaling without affecting inside-out signaling, this result indicates that calpain cleavage has the potential to selectively regulate outside-in signaling during platelet activation.

    Figure 7. Progressive cleavage at different sites of the ?3 cytoplasmic domain by calpain. Washed human platelets (109/ml) were directly solubilized in SDS-PAGE sample buffer containing EDTA and E64 (Control), or treated with thrombin (0.1 U/ml) for increasing lengths of time at 37°C before being solubilized. Platelet lysates were then immunoblotted with Ab759, Ab754, Ab747, and Ab741 to detect calpain cleavages at Y759, F754, T747, and Y741 sites, with Ab762 to detect uncleaved ?3 and Mab 15 to detect total ?3 levels. Quantification of antibody reactions was performed by scanning the immunoblots and analyzing using NIH Image as described under the Materials and methods. Percentages of the molecules cleaved (or uncleaved) at a particular site are shown in B.

    Distribution of calpain-cleaved ?3 in spreading platelets

    Calpain only cleaves a percentage of ?3 molecules in platelets stimulated with thrombin, suggesting that cleavage of ?3 is likely to have only a localized effect on integrin function. It is known that during cell spreading on integrin ligands, integrin forms a localized signaling complex with cytoskeletal and signaling molecules that is dynamically regulated and involves calpain activity (Bialkowska et al., 2000). To determine if selective integrin cleavage occurred at specific regions during platelet spreading on integrin ligands, platelets were allowed to spread on fibrinogen for 60 min and then double stained with calpain cleavage–specific antibodies and a monoclonal antibody against an extracellular epitope of ?3 that is not affected by calpain cleavage. Consistent with the above results in thrombin-stimulated platelets, calpain cleavage occurred mainly at Y759 (Fig. 8), but cleavages at F754 (Fig. 8) or the more NH2-terminal sites (unpublished data) were significantly weaker. Whereas the ?3 molecules were distributed throughout the spreading platelets with particularly strong staining at the edges, calpain-cleaved ?3 was not seen at the edges (or in the pseudopods) but was concentrated in the central region of the spreading platelets as clusters. As integrin engagement with immobilized ligands starts from the central region where discoid platelets initially adhere, our data suggest that cleavage of ?3 by calpain occurred only to the population of the integrin that is no longer at the leading edge of spreading platelets. This selective cleavage of the centrally located integrin population (mainly at Y759) may serve to down-regulate local integrin outside-in signaling in these areas without affecting the activation state of the integrin. It is possible that selective cleavage of integrin at Y759 may serve to facilitate the reorganization of the integrin–cytoskeleton signaling complex during platelet spreading.

    Figure 8. Localization of calpain-cleaved ?3 in spreading platelets. Platelets in Tyrode's buffer (1 x 108/ml) were incubated in the chamber slides precoated with 20 μg/ml fibrinogen for 2 h at 37°C. After three washes, the adherent platelets were fixed with 4% paraformaldehyde and permeabilized. The slides were incubated subsequently with Mab15, specific for integrin ?3 extracellular domain (green), and with one of the cleavage site–specific antibodies (Ab759 and Ab754) or an antibody against the COOH terminus of ?3 (Ab762) (red), and then with Alexa Fluor 488–conjugated anti–mouse IgG antibody and Alexa Fluor 546–conjugated anti–rabbit IgG antibodies. The slides were scanned under a confocal microscope (63x lens). Images of the green and red fluorescence overlay are shown in the left column. Green fluorescence (Mab15) from the same images is shown in the middle column. Red fluorescence (cleavage site–specific antibodies) from the same images is shown in the right column.

    Discussion

    By using a reconstituted integrin activation and adhesion model in transfected CHO cells and a panel of truncation mutants of the integrin IIb?3, we show that the N756ITY759 motif of the ?3 cytoplasmic domain plays a critical role in inside-out signaling of the integrin IIb?3, and the COOH-terminal R760GT762 sequence of ?3 is important in outside-in signaling–dependent events. Furthermore, we show that calpain preferentially cleaves a population of ?3 at Y759 in thrombin-activated platelets as well as in platelets spreading on fibrinogen, suggesting that calpain may selectively regulate outside-in signaling of the integrin.

    We conclude that the NITY sequence of the ?3 cytoplasmic domain is critical to the inside-out signaling function of IIb?3. This conclusion is supported by the finding that deletion of the RGT sequence COOH terminal to the NITY motif did not affect vWF-induced integrin activation, but deletion of the TNITY sequence completely abolished integrin activation. The importance of the NITY sequence in inside-out signaling is further supported by the data that the Y759A point mutation inhibited vWF-induced integrin activation. Also, we show that inhibition of vWF-induced fibrinogen binding is not caused by loss of the fibrinogen-binding function per se, as RGDS-induced fibrinogen binding to either 1b9/754 or 1b9/Y759A mutant cells was not different from the wild-type integrin. Furthermore, our data indicate that the NITY sequence and the COOH-terminal RGT sequence are both important for outside-in signaling of IIb?3 and integrin-dependent stable cell adhesion and spreading. This is consistent with previous work showing that this region of the ?3 cytoplasmic domain is important in cell spreading and focal adhesion formation (Ylanne et al., 1993, 1995). Thus, the NITY motif is important for both inside-out and outside-in signaling of the integrin IIb?3.

    The NITY motif contains a tyrosine residue that becomes phosphorylated during platelet aggregation (Law et al., 1996). The phosphorylated NXXY motifs have been shown to interact with several intracellular molecules, including myosin and phosphotyrosine-binding proteins such as GRB2 and SHC (Cowan et al., 2000), which are implicated in integrin outside-in signaling. It has been shown that mutation of both tyrosine residues to phenolalanines selectively abolished outside-in signaling in transgenic mouse platelets (Law et al., 1999), suggesting that phosphorylation of one or both of these tyrosine residues is important for outside-in signaling but not for integrin activation (inside-out signaling). Thus, although we conclude that the NITY sequence is essential for inside-out signaling, phosphorylation at Y759 is unlikely to be involved in this process. In this regard, a functional difference between Y759F and Y759A mutations has been shown previously (Schaffner-Reckinger et al., 1998). Y759A, but not Y759F, inhibited integrin-dependent cell adhesion. Furthermore, tyrosine phosphorylation occurs only after platelet aggregation, suggesting that inside-out signaling does not involve tyrosine phosphorylation in the NXXY motifs (Law et al., 1996, 1999).

    It is interesting to note that although the NITY sequence is important for both inside-out and outside-in signaling, there is a significant difference in the structural requirement between inside-out signaling (integrin activation) and outside-in signaling. Cleavage of the COOH-terminal three residues did not affect inside-out signaling but significantly inhibited outside-in signaling–dependent integrin function, such as cell spreading and stable cell adhesion. On the other hand, disruption of the NITY sequence by Y759A mutation abolished vWF-induced integrin activation (as indicated by soluble fibrinogen binding) and partially (although significantly) inhibited cell spreading and stable cell adhesion on fibrinogen. In addition, previous work suggests that outside-in signaling, but not inside-out signaling, requires phosphorylation at tyrosine residues (Law et al., 1999). The difference in the structural requirements between inside-out and outside-in signals suggests that inside-out and outside-in signals may be mediated by different molecules (or mechanisms) that interact with the COOH-terminal region of ?3 during platelet adhesion and aggregation. This also suggests that inside-out and outside-in signals can be differentially regulated.

    The family of the calcium-dependent intracellular proteases, calpain, plays important roles in cytoskeletal reorganization, cell migration, platelet aggregation, and clot retraction (Fox et al., 1983; Huttenlocher et al., 1997; Croce et al., 1999; Bialkowska et al., 2000; Azam et al., 2001). We have shown previously that the ?3 cytoplasmic domain is cleaved by either calpain I or calpain II at sites flanking two NXXY motifs in human platelets (Du et al., 1995; Pfaff et al., 1999). Calpain cleavage of the ?3 cytoplasmic domain also occurs during endothelial cell apoptosis (Meredith et al., 1998). The physiological roles of calpain cleavage of ?3 have been unclear. Calpain I knockout in mouse inhibited platelet aggregation but did not affect ?3 cleavage. This suggests that cleavage of the ?3 subunit is not involved in promoting platelet aggregation (Azam et al., 2001). Here we show that one of the functional consequences of calpain cleavage of ?3 is to negatively regulate the signaling functions of integrin IIb?3. Furthermore, we show that cleavage by calpain at different sites of ?3 may result in different regulatory effects. Cleavage at Y759 has no significant effect on inside-out signaling but significantly reduces the integrin functions associated with outside-in signaling. In contrast, cleavage at F754 or further NH2-terminal sites abolishes both inside-out and outside-in signaling. Nevertheless, cleavage at these sites occurs much later during platelet activation (Figs. 7 and 8), suggesting that such cleavages are not important for the early phase of integrin activation. On the other hand, we found that calpain cleavage of integrin ?3 subunit in intact platelets mainly occurs at the most COOH-terminal Y759 site during platelet activation and adhesion (Figs. 7 and 8). Furthermore, cleavage of ?3 does not appear to occur to the integrin molecules at the leading edge of spreading platelets but occurs to more centrally localized integrin molecules. Thus, it is likely that cleavage at Y759 serves to selectively down-regulate outside-in signaling in the integrin–cytoskeletal signaling complexes that have been formed during the earlier stage of platelet spreading, thereby facilitating the dynamic reorganization of the integrin–cytoskeleton signaling complex during platelet spreading.

    Materials and methods

    Reagents and cell lines

    Human vWF was purified as described previously (Booth et al., 1984). Human -thrombin was purchased from Enzyme Research Laboratories. RGDS peptide was from Bachem, and ristocetin was from Sigma-Aldrich. Monoclonal antibodies against integrin v?3 (anti-VNR1) (O'Toole et al., 1990), against integrin ?3 (Mab15) (Frelinger et al., 1990), and against the integrin IIb?3 complex (D57 and 2G12) (Frojmovic et al., 1991) were provided by M. Ginsberg (The Scripps Research Institute, La Jolla, CA) and V. Woods (University of California, San Diego, CA). Monoclonal antibodies against GPIb (SZ2) (Ruan et al., 1987) were a gift from C. Ruan (Jiangsu Institute of Hematology, Suzhou, China). Calpain cleavage site–specific antibodies were generated by immunizing rabbits with pentapeptides (AKWDT for Ab741, NNPLY for Ab747, ATSTF for Ab754, TNITY for Ab759, and TYRGT for Ab762) as described previously (Du et al., 1995). Oregon green 488–conjugated human fibrinogen, Alexa Fluor 488–conjugated goat anti–mouse IgG, and Alexa Fluor 546–conjugated goat anti–rabbit IgG were from Molecular Probes. Integrin IIb and ?3 cDNA clones in pCDM8 vector were provided by M. Ginsberg, and a mutant ?3 cDNA clone bearing the Y759A substitution was a gift from J. Yl?nne (University of Helsinki, Helsinki, Finland). Cell lines expressing GPIb-IX complex (1b9), integrin IIb?3 (2b3a), or both GPIb-IX and IIb?3 (123) were described previously (Gu et al., 1999).

    Construction of truncation mutants of ?3 subunit

    Truncation mutagenesis was performed using PCR to introduce stop codons into integrin ?3 cDNA at sites corresponding to the carboxy side of amino acid residues 741, 747, 754, or 759, respectively. The forward primer has the sequence of AGAGCTTAAGGACAC at an AflII site of ?3 cDNA. The reverse primers contain an XhoI digestion site, a stop codon, and the 18-nucleotide ?3 sequences at the intended COOH terminus of each mutant. The PCR products were digested with restriction enzymes AspI and XhoI and ligated into a ?3 cDNA construct in a modified cDM8 vector containing only the 3'-end XhoI site that was digested with the same restriction enzymes. All mutant constructs were verified by DNA sequencing.

    Expression of mutant ?3 cDNA constructs in CHO cells

    CHO cells expressing GPIb-IX (1b9) were maintained in DEAE medium supplemented with 10% FBS, glutamine, and nonessential amino acid. Transfection was performed using LipofectAMINE 2000 (Life Technologies). Each mutant ?3 cDNA was cotransfected together with wild-type IIb and pcDNA3.1/Hyg plasmid at a ratio of 5:5:1. Stable cell lines expressing mutant ?3 were selected in 0.2 mg/ml of hygromycin (Invitrogen). Expression of integrin IIb?3 was monitored by flow cytometry using D57. Expression of GPIb-IX was detected using SZ2. Cells expressing both GPIb-IX and integrin IIb?3 were selected by cell sorting. All cell lines were sorted using the expression levels of 123 cells as a gate until similar levels of expression were achieved. To verify correct expression of calpain cleavage–mimicking mutants, cells were also solubilized and electrophoresed on 7% polyacrylamide gels and then immunoblotted with various cleavage-specific antibodies followed by detection using the ECL kit from Amersham.

    Fibrinogen binding to transfected CHO cells activated by vWF

    Activation of IIb?3 induced by ristocetin and vWF was examined by flow cytometric analysis of Oregon green 488–conjugated fibrinogen binding to IIb?3, as previously described (Gu et al., 1999; Li et al., 2001). In brief, transfected CHO cells were resuspended to 5 x 105/ml in modified Tyrode's solution (2.5 mM Hepes, 150 mM NaCl, 2.5 mM KCl, 12 mM NaHCO3, 5.5 mM D-glucose, 1 mM CaCl2, 1 mM MgCl2, 0.1% BSA, pH 7.4), incubated with Oregon green 488–conjugated fibrinogen (30 μg/ml) and ristocetin (1 mg/ml) in the presence or absence of purified human vWF (25 μg/ml) at 22°C for 30 min, and analyzed by flow cytometry. Nonspecific binding of fibrinogen was estimated by measuring fibrinogen binding in the presence of an integrin inhibitor, RGDS (1 mM).

    Fibrinogen binding induced by RGDS peptide

    RGDS pretreatment–induced fibrinogen binding was assayed according to a previously described method (Du et al., 1991). Cells in modified Tyrode's solution were incubated with 1 mM RGDS peptide at 22°C for 10 min. Paraformaldehyde–PBS was then added (to a final concentration of 1%), and the mixture was incubated at 22°C for 1 h. After adding 250 mM NH4Cl, the fixed cells were washed, resuspended in modified Tyrode's solution, and incubated with Oregon green 488–conjugated fibrinogen (30 μg/ml) for 30 min before flow cytometric analyses.

    Cell adhesion under static conditions

    Microtiter wells were coated with 10 μg/ml human vWF or fibrinogen in 0.1 M NaHCO3, pH 8.3, at 4°C overnight and then blocked with 5% BSA–PBS at 22°C for 2 h. Cell suspension (106/ml in modified Tyrode's buffer with 1% BSA) was added to ligand-coated microtiter wells and incubated for 60 min at 37°C in a CO2 incubator. After three washes, cell spreading was examined under an inverted microscope (20x objective lens). In quantitative assays, 50 μl of 0.3% p-nitrophenyl phosphate in 1% Triton X-100, 50 mM sodium acetate, pH 5.0, was added to microtiter wells and incubated at 37°C for 1 h. The reaction was stopped by adding 50 μl of 1 M NaOH. Results were determined by reading the OD at a 405-nm wavelength.

    Cell adhesion under flow

    Purified human vWF (100 μg/ml with 0.1 mM NaHCO3, pH 8.3) was added into a glass capillary tube (0.59 mm ID, 75 mm in length; Harvard Apparatus Inc.) and then incubated overnight in a humid environment at 4°C (Englund et al., 2001). The capillary tubes were rinsed with PBS and then blocked with 5% BSA in PBS. CHO cells expressing human platelet receptors (5 x 106/ml in modified Tyrode's buffer containing 5% BSA) were perfused with a syringe pump (Harvard Apparatus Inc.) through the capillary tube at various shear rates for 2 min followed by perfusion for 10 min with cell-free buffer at the same shear rates. Shear rate was calculated as described by Slack and Turitto (1994). Cell interaction with immobilized vWF was observed in real time under an inverted microscope and recorded on videotapes. The number of stable adherent cells on immobilized vWF was counted on images obtained in 10 randomly selected fields in the vWF-coated tubes. The statistical difference between 123 cells and each one of the mutant cell lines was determined using the t test.

    Calpain cleavage of integrin ?3 subunit in human platelets

    Preparation of washed human platelets has been described previously (Li et al., 2003). Platelet aggregation was induced in a Chronolog lumi-aggregometer by adding 0.1 U/ml thrombin with constant stirring at 1,000 rpm for various lengths of time. The reaction was stopped by adding an equal volume of 2x SDS-PAGE sample buffer containing 1 mM EDTA, 1 mM PMSF, and 0.1 mM E64. Samples were then analyzed by SDS-PAGE on a 7% gel and immunoblotted with cleavage-specific antibodies (Du et al., 1995). The densities of reactive bands were scanned and then quantitated using NIH Image. Lysates prepared from 123, 1b9/747, 1b9/754, and 1b9/579 CHO cell lines were used as internal calibration standards for each of the cleavage-specific antibodies. The ratio of cleaved ?3 molecules was calculated as the ratio between the optical density of the reaction of an antibody with platelet lysates and the reaction of the same antibody with the corresponding standard CHO cell lysates multiplied by the ratio between the ?3 levels in the standard CHO cell lysates and the ?3 level in platelet lysates (as determined by immunoblotting with Mab15 directed against the extracellular domain of ?3).

    Immunofluorescence analysis of calpain-cleaved ?3 in spreading platelets

    Platelets in modified Tyrode's buffer (1 x 108/ml) were allowed to adhere to the Lab-Tek chamber slides (Nunc) precoated with 20 μg/ml of fibrinogen at 37°C for 2 h as previously described (Bodnar et al., 1999). The chamber slides were rinsed three times. Adherent platelets were fixed with 1% paraformaldehyde and permeabilized with 0.1 M Tris, 10 mM EGTA, 0.15 M NaCl, 5 mM MgCl2, 1 mM PMSF, 0.1 mM E64, 0.1% Triton X-100, 1% BSA, pH 7.5. The samples were incubated first with Mab15 and one of the cleavage-specific antibodies and then with Alexa Fluor 488–conjugated goat anti–mouse IgG and Alexa Fluor 546–conjugated goat anti–rabbit IgG. The slides were then scanned under a Carl Zeiss MicroImaging, Inc. LSM510 confocal microscope (63x objective lens).

    Acknowledgments

    This work was supported in part by a Grant-in-Aid (0050306N) from the American Heart Association and by grants from the National Institutes of Health/National Heart, Lung, and Blood Institute (HL62350, HL68819, and HL41793). A part of the work by X. Du was done during the tenure of the Established Investigatorship of the American Heart Association.

    References

    Azam, M., S.S. Andrabi, K.E. Sahr, L. Kamath, A. Kuliopulos, and A.H. Chishti. 2001. Disruption of the mouse μ-calpain gene reveals an essential role in platelet function. Mol. Cell. Biol. 21:2213–2220.

    Bialkowska, K., S. Kulkarni, X. Du, D.E. Goll, T.C. Saido, and J.E. Fox. 2000. Evidence that ?3 integrin-induced Rac activation involves the calpain-dependent formation of integrin clusters that are distinct from the focal complexes and focal adhesions that form as Rac and RhoA become active. J. Cell Biol. 151:685–696.

    Bodnar, R.J., M. Gu, Z. Li, G.D. Englund, and X. Du. 1999. The cytoplasmic domain of the platelet glycoprotein Ib is phosphorylated at serine609. J. Biol. Chem. 274:33474–33479.

    Booth, W.J., F.H. Furby, M.C. Berndt, and P.A. Castaldi. 1984. Factor VIII/von Willebrand factor has potent lectin activity. Biochem. Biophys. Res. Commun. 118:495–501.

    Calderwood, D.A., R. Zent, R. Grant, D.J. Rees, R.O. Hynes, and M.H. Ginsberg. 1999. The Talin head domain binds to integrin ? subunit cytoplasmic tails and regulates integrin activation. J. Biol. Chem. 274:28071–28074.

    Calderwood, D.A., B. Yan, J.M. de Pereda, B.G. Alvarez, Y. Fujioka, R.C. Liddington, and M.H. Ginsberg. 2002. The phosphotyrosine binding-like domain of talin activates integrins. J. Biol. Chem. 277:21749–21758.

    Chen, Y.P., I. Djaffar, D. Pidard, B. Steiner, A.M. Cieutat, J.P. Caen, and J.P. Rosa. 1992. Ser-752Pro mutation in the cytoplasmic domain of integrin ?3 subunit and defective activation of platelet integrin IIb?3 (glycoprotein IIb-IIIa) in a variant of Glanzmann thrombasthenia. Proc. Natl. Acad. Sci. USA. 89:10169–10173.

    Chen, Y.P., T.E. O'Toole, J. Ylanne, J.P. Rosa, and M.H. Ginsberg. 1994. A point mutation in the integrin ?3 cytoplasmic domain (S752P) impairs bidirectional signaling through IIb?3 (platelet glycoprotein IIb-IIIa). Blood. 84:1857–1865.

    Clark, E.A., S.J. Shattil, M.H. Ginsberg, J. Bolen, and J.S. Brugge. 1994. Regulation of the protein tyrosine kinase pp72syk by platelet agonists and the integrin IIb?3. J. Biol. Chem. 269:28859–28864.

    Coller, B.S. 1980. Interaction of normal, thrombasthenic, and Bernard-Soulier platelets with immobilized fibrinogen: defective platelet-fibrinogen interaction in thrombasthenia. Blood. 55:169–178.

    Cowan, K.J., D.A. Law, and D.R. Phillips. 2000. Identification of Shc as the primary protein binding to the tyrosine-phosphorylated ?3 subunit of IIb?3 during outside-in integrin platelet signaling. J. Biol. Chem. 275:36423–36429.

    Croce, K., R. Flaumenhaft, M. Rivers, B. Furie, B.C. Furie, I.M. Herman, and D.A. Potter. 1999. Inhibition of calpain blocks platelet secretion, aggregation, and spreading. J. Biol. Chem. 274:36321–36327.

    Du, X., T.C. Saido, S. Tsubuki, F.E. Indig, M.J. Williams, and M.H. Ginsberg. 1995. Calpain cleavage of the cytoplasmic domain of the integrin ?3 subunit. J. Biol. Chem. 270:26146–26151.

    Du, X.P., E.F. Plow, A.L.I. Frelinger, T.E. O'Toole, J.C. Loftus, and M.H. Ginsberg. 1991. Ligands "activate" integrin IIb?3 (platelet GPIIb-IIIa). Cell. 65:409–416.

    Englund, G.D., R.J. Bodnar, Z. Li, Z.M. Ruggeri, and X. Du. 2001. Regulation of von Willebrand factor binding to the platelet glycoprotein Ib-IX by a membrane skeleton-dependent inside-out signal. J. Biol. Chem. 276:16952–16959.

    Ferrell, J.J., and G.S. Martin. 1989. Tyrosine-specific protein phosphorylation is regulated by glycoprotein IIb-IIIa in platelets. Proc. Natl. Acad. Sci. USA. 86:2234–2238.

    Fox, J.E., C.C. Reynolds, and D.R. Phillips. 1983. Calcium-dependent proteolysis occurs during platelet aggregation. J. Biol. Chem. 258:9973–9981.

    Frelinger, A.L.I., I. Cohen, E.F. Plow, M.A. Smith, J. Roberts, S.C. Lam, and M.H. Ginsberg. 1990. Selective inhibition of integrin function by antibodies specific for ligand-occupied receptor conformers. J. Biol. Chem. 265:6346–6352.

    Frojmovic, M.M., T.E. O'Toole, E.F. Plow, J.C. Loftus, and M.H. Ginsberg. 1991. Platelet glycoprotein IIb-IIIa (IIb?3 integrin) confers fibrinogen- and activation-dependent aggregation on heterologous cells. Blood. 78:369–376.

    Ginsberg, M.H., X. Du, and E.F. Plow. 1992. Inside-out integrin signalling. Curr. Opin. Cell Biol. 4:766–771.

    Ginsberg, M.H., X. Du, T.E. O'Toole, and J.C. Loftus. 1995. Platelet integrins. Thromb. Haemost. 74:352–359.

    Golden, A., J.S. Brugge, and S.J. Shattil. 1990. Role of platelet membrane glycoprotein IIb-IIIa in agonist-induced tyrosine phosphorylation of platelet proteins. J. Cell Biol. 111:3117–3127.

    Gu, M., X. Xi, G.D. Englund, M.C. Berndt, and X. Du. 1999. Analysis of the roles of 14-3-3 in the platelet glycoprotein Ib-IX–mediated activation of integrin IIb?3 using a reconstituted mammalian cell expression model. J. Cell Biol. 147:1085–1096.

    Hannigan, G.E., C. Leung-Hagesteijn, L. Fitz-Gibbon, M.G. Coppolino, G. Radeva, J. Filmus, J.C. Bell, and S. Dedhar. 1996. Regulation of cell adhesion and anchorage-dependent growth by a new ?1-integrin-linked protein kinase. Nature. 379:91–96.

    Hughes, P.E., T.E. O'Toole, J. Ylanne, S.J. Shattil, and M.H. Ginsberg. 1995. The conserved membrane-proximal region of an integrin cytoplasmic domain specifies ligand binding affinity. J. Biol. Chem. 270:12411–12417.

    Hughes, P.E., G.F. Diaz, L. Leong, C. Wu, J.A. McDonald, S.J. Shattil, and M.H. Ginsberg. 1996. Breaking the integrin hinge. A defined structural constraint regulates integrin signaling. J. Biol. Chem. 271:6571–6574.

    Huttenlocher, A., S.P. Palecek, Q. Lu, W. Zhang, R.L. Mellgren, D.A. Lauffenburger, M.H. Ginsberg, and A.F. Horwitz. 1997. Regulation of cell migration by the calcium-dependent protease calpain. J. Biol. Chem. 272:32719–32722.

    Kashiwagi, H., M.A. Schwartz, M. Eigenthaler, K.A. Davis, M.H. Ginsberg, and S.J. Shattil. 1997. Affinity modulation of platelet integrin IIb?3 by ?3-endonexin, a selective binding partner of the ?3 integrin cytoplasmic tail. J. Cell Biol. 137:1433–1443.

    Knezevic, I., T.M. Leisner, and S. Lam. 1996. Direct binding of the platelet integrin IIb?3 (GPIIb-IIIa) to talin. Evidence that interaction is mediated through the cytoplasmic domains of both IIb and ?3. J. Biol. Chem. 271:16416–16421.

    Law, D.A., A.L. Nannizzi, and D.R. Phillips. 1996. Outside-in integrin signal transduction. IIb?3-(GPIIb-IIIa) tyrosine phosphorylation induced by platelet aggregation. J. Biol. Chem. 271:10811–10815.

    Law, D.A., F.R. DeGuzman, P. Heiser, K. Ministri-Madrid, N. Killeen, and D.R. Phillips. 1999. Integrin cytoplasmic tyrosine motif is required for outside-in IIb?3 signalling and platelet function. Nature. 401:808–811.

    Lerea, K.M., K.P. Cordero, K.S. Sakariassen, R.I. Kirk, and V.A. Fried. 1999. Phosphorylation sites in the integrin ?3 cytoplasmic domain in intact platelets. J. Biol. Chem. 274:1914–1919.

    Li, Z., X. Xi, and X. Du. 2001. A mitogen-activated protein kinase-dependent signaling pathway in the activation of platelet integrin IIb?3. J. Biol. Chem. 276:42226–42232.

    Li, Z., X. Xi, M. Gu, R. Feil, R.D. Ye, M. Eigenthaler, F. Hofmann, and X. Du. 2003. A stimulatory role for cGMP-dependent protein kinase in platelet activation. Cell. 112:77–86.

    Lipfert, L., B. Haimovich, M.D. Schaller, B.S. Cobb, J.T. Parsons, and J.S. Brugge. 1992. Integrin-dependent phosphorylation and activation of the protein tyrosine kinase pp125FAK in platelets. J. Cell Biol. 119:905–912.

    Meredith, J.J., Z. Mu, T. Saido, and X. Du. 1998. Cleavage of the cytoplasmic domain of the integrin ?3 subunit during endothelial cell apoptosis. J. Biol. Chem. 273:19525–19531.

    Naik, U.P., P.M. Patel, and L.V. Parise. 1997. Identification of a novel calcium-binding protein that interacts with the integrin IIb cytoplasmic domain. J. Biol. Chem. 272:4651–4654.

    O'Toole, T.E., J.C. Loftus, X.P. Du, A.A. Glass, Z.M. Ruggeri, S.J. Shattil, E.F. Plow, and M.H. Ginsberg. 1990. Affinity modulation of the IIb?3 integrin (platelet GPIIb-IIIa) is an intrinsic property of the receptor. Cell Regul. 1:883–893.

    O'Toole, T.E., D. Mandelman, J. Forsyth, S.J. Shattil, E.F. Plow, and M.H. Ginsberg. 1991. Modulation of the affinity of integrin IIb?3 (GPIIb-IIIa) by the cytoplasmic domain of IIb. Science. 254:845–847.

    O'Toole, T.E., Y. Katagiri, R.J. Faull, K. Peter, R. Tamura, V. Quaranta, J.C. Loftus, S.J. Shattil, and M.H. Ginsberg. 1994. Integrin cytoplasmic domains mediate inside-out signal transduction. J. Cell Biol. 124:1047–1059.

    Parise, L.V. 1999. Integrin IIb?3 signaling in platelet adhesion and aggregation. Curr. Opin. Cell Biol. 11:597–601.

    Patil, S., A. Jedsadayanmata, J.D. Wencel-Drake, W. Wang, I. Knezevic, and S.C. Lam. 1999. Identification of a talin-binding site in the integrin ?3 subunit distinct from the NPLY regulatory motif of post-ligand binding functions. The talin n-terminal head domain interacts with the membrane-proximal region of the ?3 cytoplasmic tail. J. Biol. Chem. 274:28575–28583.

    Pelletier, A.J., S.C. Bodary, and A.D. Levinson. 1992. Signal transduction by the platelet integrin IIb?3: induction of calcium oscillations required for protein-tyrosine phosphorylation and ligand-induced spreading of stably transfected cells. Mol. Biol. Cell. 3:989–998.

    Pfaff, M., X. Du, and M.H. Ginsberg. 1999. Calpain cleavage of integrin ? cytoplasmic domains. FEBS Lett. 460:17–22.

    Phillips, D.R., L.K. Jennings, and H.H. Edwards. 1980. Identification of membrane proteins mediating the interaction of human platelets. J. Cell Biol. 86:77–86.

    Phillips, D.R., I.F. Charo, and R.M. Scarborough. 1991. GPIIb-IIIa: the responsive integrin. Cell. 65:359–362.

    Phillips, D.R., L. Nannizzi-Alaimo, and K.S. Prasad. 2001. ?3 tyrosine phosphorylation in IIb?3 (platelet membrane GP IIb-IIIa) outside-in integrin signaling. Thromb. Haemost. 86:246–258.

    Ruan, C.G., X.P. Du, X.D. Xi, P.A. Castaldi, and M.C. Berndt. 1987. A murine antiglycoprotein Ib complex monoclonal antibody, SZ 2, inhibits platelet aggregation induced by both ristocetin and collagen. Blood. 69:570–577.

    Savage, B., S.J. Shattil, and Z.M. Ruggeri. 1992. Modulation of platelet function through adhesion receptors. A dual role for glycoprotein IIb-IIIa (integrin IIb?3) mediated by fibrinogen and glycoprotein Ib-von Willebrand factor. J. Biol. Chem. 267:11300–11306.

    Schaffner-Reckinger, E., V. Gouon, C. Melchior, S. Plancon, and N. Kieffer. 1998. Distinct involvement of ?3 integrin cytoplasmic domain tyrosine residues 747 and 759 in integrin-mediated cytoskeletal assembly and phosphotyrosine signaling. J. Biol. Chem. 273:12623–12632.

    Shattil, S.J., T. O'Toole, M. Eigenthaler, V. Thon, M. Williams, B.M. Babior, and M.H. Ginsberg. 1995. ?3-endonexin, a novel polypeptide that interacts specifically with the cytoplasmic tail of the integrin ?3 subunit. J. Cell Biol. 131:807–816.

    Shattil, S.J., H. Kashiwagi, and N. Pampori. 1998. Integrin signaling: the platelet paradigm. Blood. 91:2645–2657.

    Slack, S.M., and V.T. Turitto. 1994. Flow chambers and their standardization for use in studies of thrombosis. On behalf of the Subcommittee on Rheology of the Scientific and Standardization Committee of the ISTH. Thromb. Haemost. 72:777–781.

    Vinogradova, O., A. Velyvis, A. Velyviene, B. Hu, T. Haas, E. Plow, and J. Qin. 2002. A structural mechanism of integrin IIb?3 "inside-out" activation as regulated by its cytoplasmic face. Cell. 110:587–597.

    Wang, R., S.J. Shattil, D.R. Ambruso, and P.J. Newman. 1997. Truncation of the cytoplasmic domain of ?3 in a variant form of Glanzmann thrombasthenia abrogates signaling through the integrin IIb?3 complex. J. Clin. Invest. 100:2393–2403.

    Ylanne, J., Y. Chen, T.E. O'Toole, J.C. Loftus, Y. Takada, and M.H. Ginsberg. 1993. Distinct functions of integrin and ? subunit cytoplasmic domains in cell spreading and formation of focal adhesions. J. Cell Biol. 122:223–233.

    Ylanne, J., J. Huuskonen, T.E. O'Toole, M.H. Ginsberg, I. Virtanen, and C.G. Gahmberg. 1995. Mutation of the cytoplasmic domain of the integrin ?3 subunit. Differential effects on cell spreading, recruitment to adhesion plaques, endocytosis, and phagocytosis. J. Biol. Chem. 270:9550–9557.

    Zaffran, Y., S.C. Meyer, E. Negrescu, K.B. Reddy, and J.E. Fox. 2000. Signaling across the platelet adhesion receptor glycoprotein Ib-IX induces IIb?3 activation both in platelets and a transfected Chinese hamster ovary cell system. J. Biol. Chem. 275:16779–16787.(Xiaodong Xi, Richard J. Bodnar, Zhenyu L)