Starting 90 years ago with a clinical description by Glanzmann of a bleeding disorder associated with a defect in platelet function, technologic advances helped investigators identify the defect as a mutation(s) in the integrin family receptor, αIIbβ3, which has the capacity to bind fibrinogen (and other ligands) and support platelet-platelet interactions (aggregation). The receptor's activation state was found to be under exquisite control, with activators, inhibitors, and elaborate inside-out signaling mechanisms controlling its conformation. Structural biology has produced high-resolution images defining the ligand binding site at the atomic level. Research on αIIbβ3 has been bidirectional, with basic insights resulting in improved Glanzmann thrombasthenia carrier detection and prenatal diagnosis, assays to identify single nucleotide polymorphisms responsible for alloimmune neonatal thrombocytopenia, and the development of αIIbβ3 antagonists, the first rationally designed antiplatelet agents, to prevent and treat thrombotic cardiovascular disease. The future looks equally bright, with the potential for improved drugs and the application of gene therapy and stem cell biology to address the genetic abnormalities. The αIIbβ3 saga serves as a paradigm of rigorous science growing out of careful clinical observations of a rare disorder yielding both important new scientific information and improved diagnosis, therapy, and prevention of other disorders.

“Thus blood, for all its raw physicality, its heat, color and smell, remains first and foremost a powerfully symbolic substance—capable of representing the most primeval forces of life, and of death.”1 

“… for the blood is life …” Deuteronomy 12:23

To celebrate the 50th anniversary of Blood, we offer an historical account of research on our favorite receptor on the platelet surface, GPIIb/IIIa, or integrin αIIbβ3. This receptor plays an important role in hemostasis and thrombosis, and in accord with the quotations above, both processes have profound effects on life and health. The origin of the English word blood is uncertain. It may derive from a postulated Indo-European root bhel, “bloom” or “sprout,” and it has been speculated that “ancient people looked upon the effusion from incised skin as a sort of blooming,”2  an image well known to practitioners of the bleeding time.

The dominant theme in this review is how improved understanding of the structure and function of αIIbβ3 has led to opportunities to translate that knowledge into biomedical advances, including the development of αIIbβ3 antagonists, the first class of rationally designed antiplatelet agents. The subtheme is how advances in scientific technology deriving from discoveries in other fields have been crucial to improving our understanding of αIIbβ3. Figure 1 is a timeline depicting, by category, approximately when different technologies were introduced into the investigation of blood platelets and/or αIIbβ3. Thus, as with a musical fugue, we will try to tell 2 stories simultaneously, namely the scientific progress in understanding the importance of αIIbβ3 in biology and medicine and the technologic advances that enabled this progress. Because of space constraints, many key observations cannot be cited. Thus, we have chosen to cite a mixture of early work and recent review articles relevant to specific aspects of the αIIbβ3 story. The interested reader may wish to consult several books that contain more detailed information.3-8 

Figure 1

Timeline of application of new technologies to the study of platelets and/or αIIbβ3.

Figure 1

Timeline of application of new technologies to the study of platelets and/or αIIbβ3.

Close modal

Early platelet discoveries, clinical observations, and laboratory studies

Advances in microscopy and intravital technology paved the way for Bizzozero's landmark description in 1881 of blood platelets and their roles in thrombosis and hemostasis (reviewed in Robb-Smith9 ). Hayem made many important contributions, including confirming the relationship between hemorrhage and a low platelet count (1890) and describing the importance of platelets to the retraction of blood clots (1878), the latter providing the first in vitro assay of platelet function. Duke, in his landmark paper in 1910, described the ear lobe bleeding time as an in vivo assay that was prolonged in individuals with thrombocytopenia, and corrected when platelet counts increased after transfusion or disease remission.10  These early studies set the stage for Swiss pediatrician Eduard Glanzmann to describe in 1918 a series of patients with an inherited bleeding disorder characterized by mucocutaneous hemorrhage in which the platelet count was normal, but platelet function, as measured by clot retraction, was impaired. He termed the disorder hereditary hemorrhagic thrombasthenia (“weak platelet”), introducing the concept of a qualitative platelet disorder.11  Subsequently, Forio reported that patients with thrombasthenia had prolonged bleeding times, while others observed that thrombasthenic platelets failed to clump or to spread when visualized on blood smears (reviewed in Caen et al12 ).

Discoveries during the 1950s and early 1960s laid the groundwork for further characterization of the platelet abnormality in Glanzmann thrombasthenia, including observations that normal platelets adhere to connective tissue collagen and aggregate in response to adenosine diphosphate (ADP; reviewed in Marcus and Zucker13 ). In addition, platelets were found by electron microscopy to be surrounded by an electron-dense “glycocalyx” that is rich in fibrinogen,14  a plasma protein that was later discovered to enjoy an intimate relationship with αIIbβ3 (reviewed in Marcus and Zucker,13  Peerschke,15  and Bennett16 ).

A major technical advance was the invention of the platelet aggregometer in 1962, which provided a quantitative optical turbidometric method to measure platelet-platelet interactions in a plasma environment.17,18  Using this methodology, several groups reported that thrombasthenic platelets failed to aggregate in response to all known physiologic agonists, including ADP, collagen, epinephrine, serotonin, and thrombin.12,19,20  In parallel with this discovery was the finding that thrombasthenic platelets were deficient in fibrinogen.20-22 

Application of electrophoretic techniques

The development of polyacrylamide gel electrophoresis, which permitted high-resolution separation of platelet proteins, provided new opportunities to define the defect in thrombasthenic platelets. By the mid-1970s it was reported that patients' platelets were deficient in 2 glycoproteins, one in the second carbohydrate-staining region and one in the third region. With improved gel resolution these broad carbohydrate-staining regions were subdivided and the proteins deficient or abnormal in Glanzmann thrombasthenia were named glycoproteins IIb (GPIIb) and IIIa (GPIIIa)23,24  (reviewed in Nurden25 ). A number of modifications of the technique, including carbohydrate-specific staining, labeling of platelet surface proteins and carbohydrates, and separation of proteins in 2 dimensions, provided more detailed information (reviewed in George et al3,26 ). These studies established that GPIIb and GPIIIa contain carbohydrate residues and that GPIIb (Mr 140 kDa) is composed of a heavy chain and a light chain (Mr 120 and 20 kDa, respectively) held together by disulfide bonds. GPIIIa was found to undergo a paradoxical decrease in electrophoretic mobility upon reduction, suggesting that disulfide bonds in the native protein give it a compact structure. These techniques demonstrated that most, but not all, Glanzmann thrombasthenia patients had dramatic decreases in both GPIIb and GPIIIa. Immunoelectrophoretic techniques were also applied to characterize the abnormalities; they had the advantage of not requiring protein denaturation and were thus able to provide strong evidence that GPIIb and GPIIIa exist as a calcium-dependent heterodimer.27-29 

Application of fibrinogen-binding technology

The significance of the relationship between GPIIb/IIIa and fibrinogen, and of the prolonged bleeding time in patients with afibrinogenemia, became clearer in the late 1970s as investigators showed that radiolabeled fibrinogen binding to platelets required platelet activation, and that platelet aggregation required fibrinogen binding30,31  (reviewed in Peerschke15  and Bennett16 ). The key observations that thrombasthenic platelets fail to bind fibrinogen and that this is not due to a defect in “exposing” the receptor31,32  defined the pathophysiology of Glanzmann thrombasthenia as an inherited deficiency and/or abnormality of the platelet membrane fibrinogen receptor (Figure 2). Confirmation came from later studies showing that fibrinogen could bind to purified GPIIb/IIIa in several different systems.33-35 

Figure 2

Platelet fibrinogen binding studies demonstrate that platelets from patients with Glanzmann thrombasthenia cannot bind fibrinogen in response to ADP stimulation. The upper curve is of platelets from a healthy subject and the 3 lower ones are from 3 different patients. Reprinted with permission from Bennett JS, Vilaire G. J Clin Invest. 1979;64:1393-1401.31 

Figure 2

Platelet fibrinogen binding studies demonstrate that platelets from patients with Glanzmann thrombasthenia cannot bind fibrinogen in response to ADP stimulation. The upper curve is of platelets from a healthy subject and the 3 lower ones are from 3 different patients. Reprinted with permission from Bennett JS, Vilaire G. J Clin Invest. 1979;64:1393-1401.31 

Close modal

Application of pulse-chase labeling

Until the recent advent of growth-factor cocktails to expand various hematopoietic cells in culture, megakaryocytes were difficult to isolate and study because they represent less than 1% of bone marrow cells. As a result, initial studies in the late 1980s to assess the biosynthesis of GPIIb/IIIa relied on model cell lines with megakaryocyte-like features, including the human erythroleukemia (HEL) cell line.36-38  Nonetheless, cell-free and pulse-chase analysis demonstrated that GPIIb and GPIIIa are made separately in the endoplasmic reticulum, where they form a complex that is then transported to the Golgi for further processing, including carbohydrate modifications and cleavage of the GPIIb precursor molecule into the heavy and light chains.39,40  The need for GPIIb and GPIIIa to complex in order to be expressed on the cell surface41  offered an explanation for the enigma that Glanzmann thrombasthenia patients had marked deficiencies in 2 different proteins, since loss of either one would presumably prevent the other from reaching the surface.

Application of monoclonal antibody and flow cytometry technology

The development of the technique for preparing monoclonal antibodies (mAbs) and its application to platelets provided a valuable new tool to study Glanzmann thrombasthenia. In fact, the first reported mAb to platelets was directed at GPIIb,42  and mAbs to GPIIb and GPIIIa were commonly produced when mice were immunized with human platelets. Some of the mAbs reacted with GPIIb, others with GPIIIa, and still others reacted only with the GPIIb/IIIa complex. A subset of mAbs could block the binding of fibrinogen to GPIIb/IIIa and incubating these mAbs with normal platelets could recapitulate the platelet aggregation defect found in patients with Glanzmann thrombasthenia.43,44  Quantitative studies using mAbs eventually established that each platelet expresses approximately 80 000 GPIIb/IIIa receptors on its surface,45  with an additional internal pool of smaller size that can be recruited to the surface with activation, particularly by so-called strong agonists such as thrombin.46,47  When normalized for the platelet's surface area, the surface density of GPIIb/IIIa was estimated to be truly extraordinary, with receptors less than 200 Å apart on average, making it one of the densest adhesion/aggregation receptors in all of biology. The mAbs also aided in the purification of the receptor, allowing more detailed biochemical characterization and the identification of other proteins that interact with the receptor. Conformation-specific mAbs were also extremely valuable in beginning to unravel the mystery of how GPIIb/IIIa activation results in ligand binding. Thus, using appropriate screening assays, investigators were able to make antibodies that preferentially bound to activated receptors48,49  or ligand-bound receptors.50,51  Some mAbs had the reverse property, losing their binding ability with receptor activation. As the epitopes for these antibodies were identified, it became clear that receptor activation produces conformational changes in multiple regions of both glycoproteins.

The mAbs that blocked fibrinogen binding to GPIIb/IIIa were also valuable in establishing that the GPIIb/IIIa receptor was promiscuous. Ligand-binding studies had established that fibronectin, von Willebrand factor, vitronectin, and thrombospondin could all bind to platelets after appropriate stimulation, but a number of other receptors had been implicated in mediating their binding. The ability of GPIIb/IIIa-specific mAbs to inhibit, at least in part, the binding of all these ligands established that GPIIb/IIIa could also serve as a receptor for these adhesive glycoproteins.52 

The mAbs facilitated clinical diagnosis of Glanzmann thrombasthenia since the number of mAb molecules that bound to platelets could readily differentiate patients who lacked the receptor from healthy subjects.53  They also established that the platelets of carriers of Glanzmann thrombasthenia, who do not have a hemorrhagic diathesis, have approximately 50% to 60% of the normal number of surface receptors.54  There was, however, overlap in mAb-binding values between healthy subjects and carriers, and this limited the usefulness of mAb binding to diagnose carriers. The mAbs proved to be extremely useful in prenatal diagnosis because there was a sharp distinction between normal and carrier fetuses on the one hand, and affected fetuses with very low levels of GPIIb/IIIa expression on the other.55  Combining mAbs with electrophorectic techniques and immunoblotting provided even more detailed characterization of the molecular defects in different patients.56,57  These data demonstrated that small amounts of residual GPIIb and/or GPIIIa could be detected in the platelets of nearly all Glanzmann thrombasthenia patients and that the patterns were consistent within kinship groups. They also provided evidence that there was a disproportionate amount of the single-chain precursor form of GPIIb in the platelets of some patients, suggesting a defect in protein maturation.57,58 

Having mAbs specific for the activated and/or ligand-bound form of the receptor made it theoretically possible to detect activated platelets in the circulation of healthy individuals and patients and to identify individuals with a bleeding diathesis due to defective agonist-induced GPIIb/IIIa activation. The introduction of flow cytometry and the development of methods to study small volumes of whole blood (5 μL) made such studies much easier because they eliminated the need for radioactive materials and they provided both whole population data and data on individual platelets59,60  (Figure 3).

Figure 3

Application of flow cytometry and the activation-dependent monoclonal antibody PAC-1 to the study of αIIbβ3 conformational changes and ligand binding. (A) Platelets were identified and differentiated from red and white blood cells by their characteristic forward and side-angle light scatter profiles. (B) Platelets were stimulated with ADP and epinephrine or incubated with PGI2 to block activation. The fluorescence histogram depicts biotin-PAC-1 binding to the platelets detected by phycoerythrin-streptavidin. Reprinted from Shattil et al. Blood. 1987;70:307.59 

Figure 3

Application of flow cytometry and the activation-dependent monoclonal antibody PAC-1 to the study of αIIbβ3 conformational changes and ligand binding. (A) Platelets were identified and differentiated from red and white blood cells by their characteristic forward and side-angle light scatter profiles. (B) Platelets were stimulated with ADP and epinephrine or incubated with PGI2 to block activation. The fluorescence histogram depicts biotin-PAC-1 binding to the platelets detected by phycoerythrin-streptavidin. Reprinted from Shattil et al. Blood. 1987;70:307.59 

Close modal

Application of molecular biology

The development of molecular biologic techniques opened yet another exciting era of inquiry, culminating in the cloning and sequencing of the cDNAs for GPIIb and GPIIIa in 1987.61,62  Analysis of genomic DNA established that the proteins are derived from separate genes and chromosomal localization studies found that the genes are relatively near each other on chromosome 17, but not closely linked.63,64  The primary sequences provided major new insights, establishing both subunits as transmembrane proteins. GPIIb was found to have 4 calcium binding motifs, and as had been anticipated from the nonreduced/reduced sodium dodecyl sulfate–polyacrylamide gel data, GPIIIa contained a multitude of disulfide-linked extracellular cysteine residues (56!). Most importantly, these studies established that GPIIb/IIIa is a member of an extended family of heterodimeric adhesion receptors called integrins, each made up of an α subunit (GPIIb) and a β subunit (GPIIIa; reviewed in Pytela et al65  and Hynes66 ). Moreover, GPIIb/IIIa had a hemi-identical twin, αVβ3,67  which shared the same β subunit (GPIIIa)62  and had an α subunit (αV) that shared 40% homology with αIIb.61,68  Since αVβ3 is widely expressed in different cells, including osteoclasts, endothelial cells, smooth muscle cells and platelets (reviewed in Byzova et al69 ), some investigators speculated that Glanzmann thrombasthenia would be due exclusively to defects in GPIIb, presuming that loss of αVβ3 might be incompatible with life. That speculation ended when defects in either GPIIb or GPIIIa were identified in Glanzmann thrombasthenia patients70  (reviewed in French and Seligsohn71 ).

In recognition of GPIIb/IIIa's newly discovered parentage and familial relationships, it adopted a new name, αIIbβ3, that merged the old platelet glycoprotein nomenclature with the agreed-upon conventions for integrin receptors. We will use this designation for the remainder of this review. It is difficult to convey the excitement those in the field felt during the late 1980s and early 1990s, as each day brought profound new insights into the relationships among the integrins. One memorable moment epitomizing the underlying similarities of biologic phenomena occurred at an early conference on integrin receptors when an investigator studying developmental biology showed a polyacrylamide gel of a Drosophila melanogaster integrin receptor mutation (“lethal myospheroid”72 ) that looked similar to the gels obtained with the platelets of patients with Glanzmann thrombasthenia. It was soon discovered that platelets contain 4 other integrins. In contrast to αIIbβ3, however, these receptors were expressed at low levels, with approximately 1000 copies per platelet of α2β1, α5β1, and α6β1, and only 50 to 100 copies of αVβ3.73-76  The tiny amount of αVβ3, however, was very precious because its presence or absence provided a hint as to whether a Glanzmann thrombasthenic patient's molecular defect was in αIIb or β3, respectively.77 

Insights from studies of other integrin receptors began to provide important information about the process of ligand binding to αIIbβ3. Thus, the discovery that the Arg-Gly-Asp (RGD) sequence in fibronectin mediates its interaction to α5β1 (reviewed in Ruoslahti78 ) rapidly led to the recognition that small peptides and snake venoms containing the RGD sequence could inhibit fibrinogen binding to αIIbβ3 (reviewed in Gould et al79  and Ojima et al80 ). Moreover, these studies provided the missing link to understanding how von Willebrand factor, fibronectin, vitronectin, and thrombospondin could all bind to αIIbβ3, since as each of these was cloned and their amino acid sequences deduced, they all were found to contain RGD sequences in the regions mediating binding to αIIbβ3. Paradoxically, although fibrinogen contains 2 pairs of RGD sequences, the primary binding sites for αIIbβ3 necessary for platelet aggregation are at the C-termini of the 2 fibrinogen γ-chains, where a KQAGDV sequence provides a motif that can also bind to αIIbβ3.81,82 

Application of the polymerase chain reaction

Platelets contain only small amounts of mRNA, and this was a serious limitation in obtaining enough cDNA to study platelet-specific proteins. Thus, the fastidious application of the technique of reverse transcriptase polymerase chain reaction (PCR), which greatly amplifies mRNA signals, to platelets in the late 1980s added an extraordinarily powerful method to identify αIIbβ3 polymorphisms and mutations.83  The most important platelet polymorphism, termed P1A1 or HPA-1, was found to be due to a β3 Leu33Pro polymorphism84  and forms the antigenic epitope responsible for a sizable fraction of patients with neonatal alloimmune thrombocytopenia due to maternal alloimmunization and for most adults with posttransfusion purpura (Figure 4). Additional polymorphisms on αIIb or β3 implicated in causing neonatal alloimmune thrombocytopenia were also identified (reviewed in Valentin and Newman85 ). These discoveries provided vital information for families at risk of having an affected child. They also permitted embryo selection based on preimplantation diagnosis in cases where the mother is heterozygous for the polymorphism. Functional differences have been ascribed to some of these polymorphisms, but the true extent to which they impart hemorrhagic or thrombotic risk remains to be determined, and is in the purview of the burgeoning field of association studies attempting to link variations in platelet genes, including single nucleotide polymorphisms (SNPs) to variations in platelet function (reviewed in Bray86 ).

Figure 4

Application of reverse transcription and the polymerase chain reaction to identify the PlA1 polymorphism as due to a nucleotide mutation leading to a Leu33Pro substitution in the integrin β3 subunit. Bases 56-408 of integrin β3 were enzymatically amplified from individuals who were homozygous PlA2 or heterozygous PlA1/PlA2 and analyzed on agarose tells. The enzyme is sensitive to the T→C change in the sequence at base 196 associated with the PlA2 polymorphism. Reprinted with permission from Newman et al. J Clin Invest. 1989;83:1778-1781.84 

Figure 4

Application of reverse transcription and the polymerase chain reaction to identify the PlA1 polymorphism as due to a nucleotide mutation leading to a Leu33Pro substitution in the integrin β3 subunit. Bases 56-408 of integrin β3 were enzymatically amplified from individuals who were homozygous PlA2 or heterozygous PlA1/PlA2 and analyzed on agarose tells. The enzyme is sensitive to the T→C change in the sequence at base 196 associated with the PlA2 polymorphism. Reprinted with permission from Newman et al. J Clin Invest. 1989;83:1778-1781.84 

Close modal

The application of PCR to establishing the molecular basis of Glanzmann thrombasthenia in different kindreds has provided a wealth of information about the relationship between αIIbβ3 structure and function. PCR has also permitted rapid and unequivocal carrier determination and DNA-based prenatal diagnosis (by direct gene analysis and by linkage) using amniotic fluid or chorionic villus samples87  (reviewed in Wautier and Gruel88 ). Since the latter can be obtained at approximately 11 weeks of gestation, the information can be provided to families much earlier than it previously could using mAb-based and functional studies of blood obtained by percutaneous umbilical cord blood sampling at approximately 20 weeks of gestation.

Studies of patients whose platelets express at least 50% of the normal amount of αIIbβ3 but fail to bind fibrinogen, termed variant Glanzmann thrombasthenia, have been particularly instructive in identifying residues involved in ligand binding. For example, the β3 Asp119Tyr and Arg214Gln mutations established the importance of both of these regions of β3 for ligand binding.89,90  Similarly, αIIb mutations of Leu183 and Pro145 affected ligand binding disproportionately to their effects on receptor expression.91,92  Still other mutations in the cytoplasmic domains of patients with variant Glanzmann thrombasthenia provided valuable clues to the mechanisms of inside-out activation of αIIbβ3 by platelet agonists and outside-in signaling initiated by ligand engagement.93,94  Some Glanzmann thrombasthenia mutations paradoxically produced constitutively active receptors, including ones affecting cysteine residues in the thiol-rich regions of β3.95 

Application of technology to produce genetically modified mice

Gene targeting has produced a mouse null for β3, resulting in a hemorrhagic diathesis and platelet function abnormalities similar to those observed in patients with Glanzmann thrombasthenia.96  These mice are protected from developing acute thrombosis using a variety of models,97  but are not protected from developing intimal hyperplasia after vascular injury.98  They are also providing valuable new information about the role of αVβ3 in endothelial cells, osteoclasts,99  and wound healing.100  Mice lacking αIIb have also been produced and they, too, have a hemorrhagic diathesis101,102 ; they have also provided insights into the expression of αIIb during early hematopoiesis. In recent years, the use of genetic technology has been expanded to study the function of mouse platelets that contain mutations of the β3 subunit, thereby shedding light on the role of specific amino acid residues in the β3 cytoplasmic domain.103-105 

Application of electron microscopy, X-ray crystallography, nuclear magnetic resonance, and computational chemistry

One of the primary goals of biologic science is to visualize the 3-dimensional structures of molecules under different conditions and observe how they interact with other molecules. Coarse insights about αIIbβ3 structure were inferred from the primary sequences of αIIb and β3 and the impact of natural and site-directed mutations.106,107  Electron microscopy added additional structural information, suggesting that both subunits have “head” and “tail” regions; that each “tail” inserts into the plasma membrane; that the subunits make contact in their “head” regions; that fibrinogen binds to the region comprised of the 2 “heads”; and that fibrinogen binding results in long-range conformational changes in the “tail” region of the receptor.108-110  Landmark X-ray crystallographic studies in 2001 and 2002 on the extracellular domains of the αVβ3 receptor, alone and in the presence of the RGD peptide cilengitide, provided the first high resolution structures of an integrin and identified a number of different structural domains that are also common to αIIbβ3 (Figure 5).111,112  The ligand binding pocket was found to span the 2 subunits in the head region, and, most surprising, the receptor was found to adopt a bent conformation.

Figure 5

Model of αIIbβ3 based on αVβ3 crystal structure and depiction of switchblade model of αIIbβ3 conformational changes associated with activation and ligand binding. Inside-out signaling ultimately results in the binding of the talin head (H) domain binding to the cytoplasmic domain of the β3 subunit, resulting in subunit separation. This is transmitted through the transmembrane domains to the ectodomain where it results in extension of the α and β subunits and perhaps additional changes in the ligand binding region of β3. Ligand then binds, resulting in a swing-out motion of the β3 hybrid and PSI domains that may initiate outside-in signaling. Additional post-ligand binding events may occur, including homo-oligomerization of integrin transmembrane domains, leading to receptor clustering. The “deadbolt” hypothesis posits that modest changes in the β3 βA (I-like) domain brought about by movement of a nearby β3 β-terminal domain loop results in ligand binding, which is then followed by receptor extension and the swing-out motion. Adapted from Qin et al.143  The molecular models of αIIbβ3 were constructed using MODELLER 8v2 and the PDBs ITY6, IU8C, and IYUK as previously described.241  I-EGF, integrin epidermal growth factor domain; β-TD, β-terminal domain.

Figure 5

Model of αIIbβ3 based on αVβ3 crystal structure and depiction of switchblade model of αIIbβ3 conformational changes associated with activation and ligand binding. Inside-out signaling ultimately results in the binding of the talin head (H) domain binding to the cytoplasmic domain of the β3 subunit, resulting in subunit separation. This is transmitted through the transmembrane domains to the ectodomain where it results in extension of the α and β subunits and perhaps additional changes in the ligand binding region of β3. Ligand then binds, resulting in a swing-out motion of the β3 hybrid and PSI domains that may initiate outside-in signaling. Additional post-ligand binding events may occur, including homo-oligomerization of integrin transmembrane domains, leading to receptor clustering. The “deadbolt” hypothesis posits that modest changes in the β3 βA (I-like) domain brought about by movement of a nearby β3 β-terminal domain loop results in ligand binding, which is then followed by receptor extension and the swing-out motion. Adapted from Qin et al.143  The molecular models of αIIbβ3 were constructed using MODELLER 8v2 and the PDBs ITY6, IU8C, and IYUK as previously described.241  I-EGF, integrin epidermal growth factor domain; β-TD, β-terminal domain.

Close modal

In 2004, the first crystal structures of the isolated αIIbβ3 headpiece were reported, including the structures of the headpiece complexed with the drugs eptifibatide or tirofiban.113  These studies provided insights into the αIIbβ3 specificity of certain ligands and αIIbβ3 antagonists based on differences in the αIIb binding pocket compared with the αV binding pocket. The αIIbβ3 crystal structure also differed from the αVβ3 structure in the angle between the β3 domain involved in ligand binding (βA [I-like]) and the adjacent domain (β3 hybrid), suggesting that ligand binding is associated with a dramatic swing-out motion of the receptor in this region (Figure 5).

Taken together, the structural data indicate that the αIIbβ3 receptor can undergo several different conformational changes, but it is still uncertain which ones are necessary and/or sufficient for attaining the high-affinity ligand binding state(s). Two different models of αIIbβ3 activation have been proposed. The deadbolt hypothesis suggests that platelet activation leads to movement of the external domain of the β3 subunit adjacent to the membrane (β-terminal domain [β-TD]), which in turn releases a constraint on a portion of the βA (I-like) domain involved in ligand binding and allows it to undergo a subtle conformational change that results in the receptor adopting a high-affinity ligand binding state.114,115  Extension of the receptor at both the genu of αIIb and the interface between the integrin epidermal growth factor-1 (IEGF-1) and IEGF-2 domains of β3 is proposed to occur after ligand binding in this model. The switchblade hypothesis posits that the αIIbβ3 receptor undergoes extension before ligand binding and that swing-out of the β3 hybrid domain occurs concurrent with or after ligand binding.113,116,117  The swing-out motion has been proposed to participate in the initiation of outside-in signaling induced by ligand binding. Thiol-disulfide exchange has also been implicated in αIIbβ3 activation based on biochemical studies and studies of Glanzmann thrombasthenia patients with Cys mutations that result in constitutively active receptors.95,118-123  Post-ligand binding events have also been implicated in controlling the avidity of αIIbβ3, including receptor clustering124,125  and irreversible ligand binding.126 

Molecular dynamic simulations of select groups of atoms in the ligand binding regions of αIIbβ3 and αVβ3 have been performed in an attempt to understand the allosteric pathways leading to activation and the energetics of ligand binding.127-129  These computation-intensive studies have been made possible by advances in the application of biophysical and thermodynamic principles to biologic systems and the availability of more powerful computers. The results have provided models of variations in regional flexibility and interactions with water molecules over time, the force needed to remove ligands from the binding pocket under different conditions, and the allosteric pathways leading from one conformation to another. Other computational programs allow one to dock small molecules into the αIIbβ3 ligand binding pocket.130  Finally, NMR studies of the transmembrane and cytoplasmic domains of αIIbβ3 have defined interactions between the subunits and between the subunits and cytoplasmic proteins important in regulating integrin activation in addition to their heterotypic interactions131-133  (reviewed in Ma et al134 ). These have been proposed to promote receptor clustering into oligomers after activation releases the heterotypic interactions, although other workers posit that αIIbβ3 oligomerization may be driven largely by the binding of fibrinogen or other multivalent ligands. Defining the conformational changes associated with the transition to high affinity ligand binding remains a high priority, with a variety of advanced biophysical and imaging approaches being brought to bear, but a clear consensus has not emerged.135,136 

Biochemical, molecular, and genetic analyses of αIIbβ3 signaling

The presence of some regulated stimulus-response pathway leading to αIIbβ3 activation was implicit in early studies that demonstrated that platelet aggregation can be triggered by platelet agonists.17  As additional information emerged, the process that links platelet agonists to αIIbβ3 activation has come to be known as inside-out signaling. This is in contrast to direct activation of αIIbβ3 by the binding of activating monoclonal antibodies51,137,138  or low-molecular-weight ligand mimetics,139  and to outside-in signals that are sent into the platelet as the result of ligand binding and receptor clustering. Indeed, integrins have been aptly described as “bidirectional, allosteric signaling machines,”140  and studies with αIIbβ3 have played a prominent role in our understanding of the machinery (reviewed in Shattil and Newman,141  Watson et al,142  and Qin et al143 ). In reality, the 2 phases of integrin signaling are likely to be quite interdependent as evidenced by the identification of some signaling molecules, such as certain phospholipases and protein and lipid kinases that participate in both inside-out and outside-in signaling. Furthermore, some outside-in responses (for example, increases in cytoplasmic free calcium or activation of protein kinase C and cytosolic phospholipase A2) may feed back to enhance inside-out activation of additional αIIbβ3 complexes. To characterize integrin signaling, platelet researchers in the 1980s and 1990s began to take advantage of several emerging technologies, including flow cytometry (reviewed in Michelson and Shattil144 ), activation-dependent antibodies and antibodies specific for phosphorylated amino acids and proteins,49,145-148  expression of αIIbβ3 and recombinant signaling proteins in heterologous cells,149  and genetically modified mice.96,150,151  More recently, knockdown of proteins in embryonic stem cell–derived megakaryocytes by RNA interference has provided additional valuable information.152 

Inside-out signaling

Inside-out αIIbβ3 signaling can be considered in terms of (1) stimulators and inhibitors of integrin activation and the platelet receptors with which they interact; (2) intracellular protein-protein interactions and biochemical reactions that couple agonist/antagonist receptor occupancy to the final events that directly regulate αIIbβ3 affinity; and (3) the final regulatory events.

As might be expected for a reaction so critical to hemostasis, there is redundancy in the process of αIIbβ3 activation in the form of regulation by multiple agonists that are either immobilized at the vascular wound site (eg, von Willebrand factor, collagen), generated within the wound (eg, thrombin), or either stored or generated and then released by platelets (eg, ADP, thromboxane A2).153  Numerous signaling receptors for agonists and antagonists have been identified in platelets. Initial application of pharmacologic approaches in the 1960s and 1970s using dose-response relationships and selective agonists and antagonists provided important insights into the structural and functional classes of these receptors (reviewed in Mustard and Packham154 ). The introduction of molecular biologic techniques led to the cloning and characterization of many different receptors, with studies of platelets in vitro and in vivo often facilitating the elucidation of receptor families. For example, agonist receptors include the G protein–coupled protease-activated thrombin receptors PAR1 and PAR4 in human platelets and PAR3 and PAR4 in murine platelets155-157 ; the purinergic receptors for ADP (P2Y1, P2Y12 and P2X1)158,159 ; and the thromboxane A2 receptor.160  Agonist receptors not directly coupled to G proteins include primary adhesion receptors for collagen (GPVI/FcRγ and integrin α2β1) and von Willebrand factor (GP Ib-V-IX; reviewed in Watson et al142  and Varga-Szabo et al161 ). αIIbβ3 is also considered a stimulatory receptor in the sense that it undergoes conformational changes and clustering to trigger outside-in signals in response to fibrinogen binding147  (reviewed in Shattil and Newman141  and Watson et al142 ). In circulating platelets, αIIbβ3 activation is ordinarily prevented by several activities derived from endothelial cells, including a cell surface ADPase (CD39) that removes stimulatory ADP,162,163  prostacyclin, which binds to Gs-coupled platelet receptors to activate adenylate cyclase (reviewed in Brass153  and Samuelsson164 ), and nitric oxide, which activates guanylate cyclase and can also be generated within platelets.165,166 

Occupancy of some agonist receptors (eg, PAR1, PAR4, GP Ib-V-IX, GP VI/FcR γ) is usually sufficient to activate αIIbβ3. Other receptors appear to function primarily as mediators of primary platelet adhesion (α2β1; reviewed in Varga-Szabo et al161 ) or shape change (P2X1 ATP receptors; reviewed in Kunapuli et al167 ) or promote αIIbβ3 activation in concert with other stimulatory receptors. For example, P2Y1 and P2Y12 function together to induce full fibrinogen binding and platelet aggregation in response to ADP (reviewed in Kunapuli et al167 ), and PAR3 functions in mouse platelets as a cofactor for the authentic signaling thrombin receptor PAR4.156  The physiologic importance of specific agonist receptors has been established by basic scientists studying mutant mice and by the careful observations by clinicians of patients with bleeding disorders who manifest defective αIIbβ3 activation or platelet aggregation. The introduction of gene targeting technologies to knock out genes has been particularly important in defining the roles of platelet receptors (reviewed in Lee and Blajchman168 ); however, extrapolation of mouse data to human platelets must be done with caution because, despite many similarities, there can be importance species differences, as exemplified by the PAR receptors (reviewed in Sambrano et al,157  Tsakiris et al,169  and Jirouskova et al170 ).

The intracellular reactions that couple receptor occupancy to αIIbβ3 activation are complex. Significant progress has been made in establishing which G protein-coupled receptors couple to which G proteins, and how each G protein can initiate downstream signaling (reviewed in Brass153 ). Some initial biochemical responses to agonist occupancy of non-G protein–linked receptors have also been defined (reviewed in Watson et al142  and Varga-Szabo et al161 ). Specific second messengers and signaling proteins involved in inside-out signaling have been identified by biochemical studies of normal human platelets and megakaryocytes (reviewed in Shattil and Newman141 ), genetic approaches in mice (reviewed in Lee and Blajchman168 ), and studies of humans with rare inherited forms of platelet dysfunction (reviewed in Rao et al171 ). Messengers include products of phospholipase C (IP3, which increases cytoplasmic free Ca2+; diacylglycerol, which activates several protein kinase C isoforms in platelets as well as CalDAG-GEFI, a Rap1 GTPase exchange factor) and products of phosphatidylinositol 3-kinase (phosphatidylinositol 3,4 bisphosphate and phosphatidylinositol 3,4,5 trisphosphate, which recruit proteins with pleckstrin homology domains to membranes; reviewed in Brass153 ). An ongoing challenge is to understand how these and other mediators are integrated to regulate αIIbβ3 activation.

Talin is an actin-binding protein of approximately 250 kDa with a globular head and a rod-like tail that is highly expressed in platelets and can promote integrin activation, primarily by interactions of the talin head's FERM domain with integrin β cytoplasmic domains.172  Strong evidence in support of a necessary role for the protein talin in αIIbβ3 activation has been obtained by means of platelet-specific knockout of the protein in mice.173,174  During platelet activation, talin is recruited from the cytoplasm to αIIbβ3 in a process that is regulated, in part, by the Rap1 GTPase175  and RIAM, a molecular adapter and Rap1 effector176  (Figure 6). A pathway from agonist receptors to Rap1 is suggested by recent findings that platelet activation through G protein–coupled receptors or GP VI leads to Rap1 activation.177,178  Activation of Rap1 may be mediated by CalDAG-GEF1 since platelets from CalDAG-GEF1–deficient mice and humans exhibit defective αIIbβ3 activation and a bleeding diathesis.179,180  Because Rap1 depletion does not result in complete loss of platelet aggregation, pathways to αIIbβ3 activation that are independent of Rap1 must also exist.

Figure 6

Protein interactions with the cytoplasmic domains of αIIb or β3 regulate integrin signaling. Shown are some, but not all, of the proteins reported to associate with the αIIbβ3 cytoplasmic domains, many in a dynamic fashion. Some associate with αIIbβ3 in resting platelets, while others are recruited to, or dissociate from, the integrin during inside-out or outside-in signaling, leading to F-actin assembly. In addition, several proteins with enzymatic function become activated (asterisk) after fibrinogen binding to αIIbβ3. It is difficult to imagine that all of these proteins can interact with a single αIIbβ3 heterodimer in platelets; however, they might interact with and further regulate oligomers of αIIbβ3 that form in response to fibrinogen binding. Not shown are the many additional adapter molecules, enzymes and substrates that may become recruited through more indirect interactions during the various phases of integrin signaling. Abbreviations: PP1c, protein phosphatase 1c; RACK1, receptor for activated C kinase 1; Csk, c-Src tyrosine kinase; PKCβ, protein kinase Cβ; ILK, integrin-linked kinase; ITAM, a yet-to-be-identified protein with one or more immunoreceptor tyrosine activation motifs; CIB, calcium and integrin-binding 1; Syk, spleen tyrosine kinase.

Figure 6

Protein interactions with the cytoplasmic domains of αIIb or β3 regulate integrin signaling. Shown are some, but not all, of the proteins reported to associate with the αIIbβ3 cytoplasmic domains, many in a dynamic fashion. Some associate with αIIbβ3 in resting platelets, while others are recruited to, or dissociate from, the integrin during inside-out or outside-in signaling, leading to F-actin assembly. In addition, several proteins with enzymatic function become activated (asterisk) after fibrinogen binding to αIIbβ3. It is difficult to imagine that all of these proteins can interact with a single αIIbβ3 heterodimer in platelets; however, they might interact with and further regulate oligomers of αIIbβ3 that form in response to fibrinogen binding. Not shown are the many additional adapter molecules, enzymes and substrates that may become recruited through more indirect interactions during the various phases of integrin signaling. Abbreviations: PP1c, protein phosphatase 1c; RACK1, receptor for activated C kinase 1; Csk, c-Src tyrosine kinase; PKCβ, protein kinase Cβ; ILK, integrin-linked kinase; ITAM, a yet-to-be-identified protein with one or more immunoreceptor tyrosine activation motifs; CIB, calcium and integrin-binding 1; Syk, spleen tyrosine kinase.

Close modal

The mechanism by which talin activates αIIbβ3 has been illuminated by recent nuclear magnetic resonance (NMR) and crystallographic studies of purified components and mutational studies of talin and αIIbβ3 in a heterologous expression system.172  Thus, the F2-3 subdomains of talin's FERM domain may first gain a foothold within a membrane-distal region of the β3 cytoplasmic domain centered at Asn-Pro-Leu-Tyr (NPXY). Then a second interaction with a membrane-proximal region of the β3 cytoplasmic domain may flip the activation switch by disrupting a salt bridge between αIIb and β3, resulting in separation of the αIIb and β3 subunits. Other αIIbβ3-binding proteins, for example members of the FERM-domain–containing kindlin family,181,182  may also regulate αIIbβ3 activation. Whether they do so in concert with talin or independent of it remains an area of active investigation. Similarly, the mechanism(s) linking intracytoplasmic αIIb and β3 subunit separation to the conformational changes in the ectodomains responsible for high-affinity ligand binding still need to be established.

Outside-in signaling

Outside-in signaling by integrins provides a means by which these receptors mediate anchorage-dependent cellular responses. In nucleated cells, outside-in signals regulate cell adhesion, motility, and gene expression programs (reviewed in Hynes140 ). The anucleate platelet is bereft of DNA synthetic machinery, but it does possess translational machinery and can splice RNA in response to signals both from thrombin receptors and ligand binding to αIIbβ3 (reviewed in Zimmerman and Weyrich183 ). This provides a potential mechanism for activated platelets to participate, via both release of stored substances and new synthesis of substances, in processes beyond hemostasis and thrombosis, including immunity, inflammation, promotion of tumor metastasis, and angiogenesis184-186  (reviewed in Zarbock et al187 ). It is intriguing to speculate that outside-in αIIbβ3 signaling might also affect nuclear programs in both megakaryocytes and early definitive hematopoietic stem cells.188,189 

In platelets, outside-in αIIbβ3 signaling promotes actin polymerization and cytoskeletal reorganization. Accordingly, it is important for platelet spreading on extracellular matrices under conditions of shear flow, for platelet aggregate stability, and for other post-ligand binding responses, such as clot retraction. Our current knowledge of outside-in signaling in platelets is derived from careful morphologic and biochemical analyses of the cytoskeletons in resting and thrombin-activated platelets190-192 ; identification of integrin-binding proteins, including various molecular adapters as well as protein and lipid kinases and phosphatases193 ; studies of platelet responses after interaction with fibrinogen and other αIIbβ3 ligands147 ; studies of mutant mice; and investigations of platelets from patients with variant Glanzmann thrombasthenia.194  The network of signaling reactions triggered primarily and secondarily by fibrinogen binding to αIIbβ3 is truly impressive, the equal of integrin signaling patterns observed in the much larger blood leukocytes. Many unforeseen parallels are apparent between outside-in αIIbβ3 signaling and signaling triggered by immunoreceptors in platelets and leukocytes, including dependence on the Src family and Syk protein tyrosine kinases (reviewed in Shattil and Newman141  and Watson et al142 ) and apparent involvement of proteins containing ITAM motifs (reviewed in Abtahian et al195  and Jakus et al196 ). An emerging picture of αIIbβ3 signaling posits a dynamic interplay among integrin-binding proteins, with the cytoplasmic domains of αIIb and β3 serving a scaffolding function.

αIIbβ3 as a therapeutic target

A potential role of platelets in ischemic vascular disease was inferred from data derived from human pathologic specimens, animal models of thrombus formation, and biochemical studies of patients with acute ischemic events (reviewed in Coller197 ). There was, however, considerable uncertainty about whether platelets played a sufficiently important role to make antiplatelet therapy an effective intervention. Although an association between aspirin ingestion and bleeding, particularly gastrointestinal bleeding, was recognized by the 1940s, the antiplatelet effects of aspirin were first described in the mid-1960s, greatly facilitated by bleeding time measurements and the introduction of platelet aggregometry (reviewed in Weiss,198  Mustard,199  and Quick200 ). Subsequent biochemical studies established the dramatic effect of aspirin in suppressing thromboxane A2 production via irreversible acetylation of the enzyme cyclooxygenase-1 (reviewed in Marcus201  and Vane 202 ). However, early clinical studies of the effects of aspirin on ischemic vascular disease were equivocal, reflecting in part difficulties in designing large clinical trials.203  It was only with the publication of the landmark ISIS-2 study in 1988,204  in which aspirin alone decreased the mortality of acute myocardial infarction by almost 25% and adding aspirin to streptokinase further reduced mortality by almost 25%, that the potential of antiplatelet therapy was clearly demonstrated. The thienopyridine compound ticlopidine, which demonstrated greater inhibition of platelet aggregation than aspirin, entered the U.S. market in the early 1990s based on its greater efficacy than aspirin in the secondary prevention of stroke (reviewed in Savi and Herbert205 ). Its mechanism of action was later identified as irreversible blockade of the P2Y12 ADP receptor.205 

With 2 effective antiplatelet agents available, one of which was very inexpensive and had a long history of acceptable toxicity, it wasn't clear that additional agents would be worth developing. The rationale for trying to develop αIIbβ3 antagonists as therapeutic agents rested on a number of considerations (reviewed in Coller197,206 ): (1) neither aspirin nor ticlopidine was completely effective in preventing ischemic events; (2) αIIbβ3 antagonists could more completely inhibit platelet aggregation in vitro than either aspirin or ticlopidine; (3) experimental data demonstrated that vaso-occlusion leading to ischemia resulted from platelet-platelet interactions, the primary target of αIIbβ3 antagonists; and (4) the lack of effect of αIIbβ3 antagonists on other platelet adhesion receptors might theoretically allow a single layer of platelets to contribute to hemostasis, whereas the anti-αIIbβ3 effect would greatly diminish platelet thrombus formation. Equally important was the clinical observation that despite suffering variably severe mucocutaneous hemorrhage, patients with Glanzmann thrombasthenia rarely have spontaneous central nervous system bleeding—the most feared hemorrhagic complication of fibrinolytic therapy.207 

The mAb 7E3 was selected for in vivo antithrombotic studies based on its ability to bind to dog, primate, and human platelets (reviewed in Coller197,206 ). Fragments of 7E3 lacking the Fc region were used, rather than the intact antibody, to avoid platelet clearance by Fc receptor-bearing cells of the monocyte-macrophage system that recognize antibody-coated platelets. Administering the F(ab′)2 fragment of 7E3 to dogs was able to produce greater inhibition of ADP-induced platelet aggregation than could be achieved with aspirin and it offered greater pro-tection than aspirin from platelet-mediated thrombosis in dog and primate models of unstable angina or myocardial infarction.208,209  Similar results in these and other animal models were obtained with low molecular weight αIIbβ3 antagonists, including eptifibatide, a cyclic heptapeptide patterned on a KGD motif that conferred selectivity for αIIbβ3,210  and the nonpeptide RGD-mimetic tirofiban.211 

For in vivo studies in humans, a chimeric 7E3 Fab molecule designated abciximab was developed containing murine variable regions and human IgG1 constant regions.212  Based on the 2099-patient EPIC study, abciximab was approved by the Food and Drug Administration in 1994 as adjunctive therapy to prevent ischemic complications of coronary artery angioplasty in high-risk patients (reviewed in Topol et al213 ). Based on additional studies, it was approved for use in percutaneous coronary interventions (PCI) involving stents and in patients with unstable angina who are expected to undergo PCI. Eptifibatide and tirofiban were subsequently approved for human use in patients with unstable angina and those undergoing PCI.214,215 

In the EPIC study, abciximab increased the absolute risk of major bleeding by approximately 7%, but by reducing the dose of heparin used in combination with abciximab in subsequent studies, the absolute increase in risk was reduced to approximately 2% or less.216  Eptifibatide and tirofiban confer similar or lesser increase in risk of bleeding. Thrombocytopenia has been reported with all of the αIIbβ3 antagonists, although it is more common with abciximab (reviewed in Aster et al217  and Aster and Bougie218 ). A number of mechanisms have been proposed to explain the thrombocytopenia, including the presence of antibodies to the murine component of abciximab and antibodies to neoepitopes on αIIbβ3 exposed and/or created by the binding of the drugs to the receptor.

In a large number of randomized, placebo-controlled studies of patients undergoing PCI and/or treatment for unstable angina conducted in the 1990s, these agents demonstrated benefits in reducing the risk of myocardial infarction and the need for urgent reinterventions to treat threatened occlusions (reviewed in Topol et al,213  Curran and Keating,214  Manozzi et al,215  and De et al219 ). Long-term mortality advantages have also been demonstrated, with most of the benefit paradoxically occurring long after the antiplatelet effects of the drug wore off (reviewed in Topol et al213 ). Recently, a number of factors have narrowed the indications for these agents in treating cardiovascular disease, including the availability of clopidogrel, a thienopyridine related to ticlopidine that has a better toxicity profile and can achieve rapid inhibition of the P2Y12 receptor with a loading dose (reviewed in in Savi and Herbert205  and Plosker and Lyseng-Williamson220 ), and bivalirudin, a direct thrombin inhibitor that acts as both an anticoagulant and antiplatelet agent.221  Remaining questions include whether the αIIbβ3 antagonists will prove uniquely beneficial when given very early after the onset of symptoms,222,223  when given by the intracoronary route224,225  (reviewed in Gibson et al226 ), or when used in combination with a thrombus aspiration device227  or reduced-dose thrombolytic agents when PCI is not immediately available.223,228  The use of αIIbβ3 antagonists to treat acute stroke appeared promising in case reports and early phase studies, but a phase 3 study with abciximab failed to show a benefit; whether modifications in patient selection or dosing would improve the results remains an area of investigation.229 

In contrast to the intravenous αIIbβ3 antagonists, oral small molecule αIIbβ3 antagonists have not demonstrated clinical efficacy when administered for secondary prophylaxis and, in fact, have been associated with increased mortality, increased bleeding, and occasionally severe thrombocytopenia230  (reviewed in Quinn et al231 ). Although it is not understood why these agents failed to have a beneficial effect, it has been speculated that their binding to platelets may induce conformational changes in αIIbβ3 that can both activate the receptor, resulting in paradoxical thrombosis, and expose neoepitopes recognized by some patients' preformed antibodies, resulting in thrombocytopenia.139,230-232 

The future

The scientific advances in understanding the structure and function of αIIbβ3 are impressive, but the therapy of Glanzmann thrombasthenia remains unsatisfactory. Thus, while there have been advances in platelet transfusion therapy (including better storage methods, HL-A matching, and leukoreduction), and the introduction of recombinant factor VIIa therapy has provided a blood-free alternative that is frequently effective, though costly (reviewed in Poon233 ), patients commonly lead lives compromised by variably severe, and sometimes continuous, mucocutaneous hemorrhage, seriously increased risks of surgery and childbirth, and occasional life-threatening gastrointestinal bleeding or trauma-related cerebral hemorrhage.

Bone marrow transplantation and stem cell reconstitution can cure the disorder,234,235  but the risks of these procedures remain substantial, even with newer conditioning regimens, and thus they are still not desirable options for most patients (reviewed in Flood et al236 ). Proof of concept for gene therapy of Glanzmann thrombasthenia has been provided in animal models237  (reviewed in Wilcox and White238 ), but much more remains to be done, including how to deal with the immune response to the newly expressed αIIbβ3. Select patients with appropriate nonsense mutations may be candidates for drug therapy to promote DNA “read-through,” but this technology is still in its early phase of development.239  Stem cell biology also offers promise if autologous hematopoietic precursors can be genetically corrected and used to populate the patient's bone marrow or produce platelets in vitro, but the immunologic barriers will likely remain.

Because αIIbβ3 is a proven therapeutic target for antithrombotic therapy, there is reason to speculate on potential ways to improve on the currently available drugs. Drugs that inhibit the receptor without altering its conformation may have several advantages, including a reduction in both thrombocytopenia and receptor activation. Because engagement of the β3 MIDAS motif may be important in initiating the swing-out motion of the hybrid domain, agents that selectively bind to αIIb without effects on β3 may be advantageous.130  Similarly, it may be useful to identify agents that do not bind to the ligand binding site at all but can selectively inhibit inside-out or outside-in signaling.105  In this regard, the currently available high-resolution structures of the β3 integrins, enlightening as they are, represent but snapshots of partial fragments of the whole transmembrane heterodimer. A more complete understanding of the biologically relevant conformational changes in the intact receptor remains an exciting goal for future investigators. In addition, the application of high-throughput screening,130  molecular docking,130  and rational drug design offers hope for identifying novel compounds. Finally, it is possible that monitoring the antiplatelet effects of existing αIIbβ3 antagonists may improve their safety and/or efficacy (reviewed in Harrison et al240 ). Controlled studies will be needed, however, to test whether dose adjustment based on such monitoring improves clinical outcome.

It is remarkable that studies of αIIbβ3 during the 9 decades since Glanzmann reported patients with the disease that bears his name, and the 5 decades of the American Society of Hematology's existence have gone from intact individual humans to individual atoms at a resolution of 2.8 Å, representing in effect, a span of 27 logs in mass. It is gratifying that the molecular advances have provided patients with Glanzmann thrombasthenia and their families improved diagnosis, carrier detection, and prenatal diagnosis, but it is frustrating that therapeutic advances have lagged. It is also gratifying that understanding the role of αIIbβ3 in platelet thrombus formation has led to the first rationally designed antiplatelet therapeutics and the first anti-integrin therapeutics. The current agents, however, have significant limitations, and the failure of oral αIIbβ3 antagonists was a major disappointment. It is also gratifying that studies of αIIbβ3 have led the way across a wide range of fundamental biologic phenomena related to cell activation, signal transduction, and cytoskeletal rearrangements, as well as genetic and molecular biologic phenomena, including SNPs and the phenomenon of activation-dependent mRNA translation. Given the remarkable trajectory of biomedical science, we anticipate that many more conceptual and practical breakthroughs in this area of hematology await members of the Society and their patients in the next 50 years.

We wish to thank Mark Ginsberg, Deborah French, and Uri Seligsohn for their careful review of the manuscript, Dr Marta Murcia for the molecular modeling and images depicted in Figure 6, Suzanne Rivera for outstanding secretarial assistance, and our many colleagues in the field of αIIbβ3 biology who have made important contributions to this remarkable story. We regret that space and format limitations did not allow for more extensive recognition of all of their achievements.

This work was supported in part by grants HL19278, HL56595, HL57900, and HL78784 from the National Heart, Lung, and Blood Institute, grant MH083257 from the National Institute of Mental Health, a Clinical and Translational Science Award (UL1 RR024143) from the National Center for Research Resources, and funds from Stony Brook University (NY).

National Institutes of Health

Contribution: B.S.C. and S.J.S. wrote the paper.

Conflict-of-interest disclosure: In accord with federal law and the policies of the Research Foundation of the State University of New York, B.S.C. has a royalty interest in abciximab (Centocor), and in accord with federal law and the policies of the Mount Sinai School of Medicine, B.S.C. has a royalty interest in the VerifyNow assay system (Accumetrics). In addition, B.S.C. is a consultant to Accumetrics and Novo-Nordisk and is an inventor of an αIIbβ3 antagonist compound identified by high-throughput screening. S.J.S. declares no competing financial interests.

Correspondence: Dr Barry S. Coller, Laboratory of Blood and Vascular Biology, The Rockefeller University, 1230 York Avenue, Box 309, New York, NY 10065; e-mail: collerb@rockefeller.edu.

1
Gabriele
 
M
Bradburne
 
JM
Blood and magic in classical antiquity.
Blood: Art, Power, Politics and Pathology
2002
Munich
Prestel
(pg. 
33
-
39
)
2
Medical Meanings: A Glossary of Word Origins
1997
Philadelphia
American College of Physicians
3
George
 
JN
Nurden
 
AT
Phillips
 
DR
Platelet membrane glycoproteins
1985
New York
Plenum
4
Michelson
 
AD
Platelets
2007
Burlington, MA
Academic Press
5
Lichtman
 
MA
Beutler
 
E
Kipps
 
TJ
Seligsohn
 
U
Kaushansky
 
K
Prchal
 
JT
Williams Hematology
2006
New York
McGraw-Hill
6
Hoffman
 
R
Benz
 
EJ
Shattil
 
S
Furie
 
B
Cohen
 
HJ
Silberstein
 
LE
McGlave
 
P
Hematology: Basic Principles and Practice
2005
Philadelphia
Elsevier Churchill Livingstone
7
Colman
 
RW
Marder
 
VJ
Clowes
 
AW
George
 
JN
Hemostasis and Thrombosis: Basic Principles and Clinical Practice
2000
Philadelphia
Lippincott Williams & Wilkins
8
Kunick
 
TJ
George
 
JG
Platelet Immunobiology: Molecular and Clinical Aspects
1989
Philadelphia
Lippincott Williams & Wilkins
9
Robb-Smith
 
AH
Why the platelets were discovered [review].
Br J Haematol
1967
, vol. 
13
 (pg. 
618
-
637
)
10
Duke
 
WW
The relation of blood platelets to hemorrhagic disease. Description of a method for determining the bleeding time and the coagulation time and report of three cases of hemorrhagic disease relieved by transfusion.
JAMA
1910
, vol. 
55
 (pg. 
1185
-
1192
)
11
Glanzmann
 
E
Hereditäre hämmorhagische Thrombasthenie. Ein Beitrag zur Pathologie der Blutplättchen.
Jahrbuch fur Kinderheilkunde und physiche Erziehung
1918
, vol. 
88
 (pg. 
113
-
141
)
12
Caen
 
JP
Castaldi
 
PA
Leclerc
 
JC
, et al. 
Congenital bleeding disorders with long bleeding time and normal platelet count. I. Glanzmann's thrombasthenia.
Am J Med
1966
, vol. 
41
 (pg. 
4
-
26
)
13
Marcus
 
AJ
Zucker
 
MB
The Physiology of Blood Platelets
1965
New York
Gurne and Stratton
14
Behnke
 
O
Electron microscopical observations on the surface coating of human blood platelets.
J Ultrastruct Res
1968
, vol. 
24
 (pg. 
51
-
69
)
15
Peerschke
 
EI
The platelet fibrinogen receptor [review].
Semin Hematol
1985
, vol. 
22
 (pg. 
241
-
259
)
16
Bennett
 
JS
George
 
JN
Nurden
 
AT
Phillips
 
DR
The platelet-fibrinogen interaction.
Platelet Membrane Glycoproteins
1985
New York
Plenum
(pg. 
51
-
85
)
17
Born
 
GV
Aggregation of blood platelets by adenosine diphosphate and its reversal.
Nature
1962
, vol. 
194
 (pg. 
927
-
929
)
18
O'Brien
 
J
Platelet aggregation. II. Some results from a new method of study.
J Clin Pathol
1962
, vol. 
15
 (pg. 
452
-
481
)
19
Hardisty
 
RM
Dormandy
 
KM
Hutton
 
RA
Thrombasthenia: studies on three cases.
Br J Haematol
1964
, vol. 
10
 (pg. 
371
-
383
)
20
Zucker
 
MB
Pert
 
JH
Hilgartner
 
MW
Platelet function in a patient with thrombasthenia.
Blood
1966
, vol. 
28
 (pg. 
524
-
534
)
21
Jackson
 
DP
Morse
 
EE
Zieve
 
PD
Conley
 
CL
Thrombocytopathic purpura associated with defective clot retraction and absence of platelet fibrinogen [abstract].
Blood
1963
, vol. 
22
 pg. 
827
 
22
Nachman
 
RL
Marcus
 
AJ
Immunological studies of proteins associated with the subcellular fractions of thrombasthenic and afibrinogenaemic platelets.
Br J Haematol
1968
, vol. 
15
 (pg. 
181
-
189
)
23
Nurden
 
AT
Caen
 
JP
An abnormal platelet glycoprotein pattern in three cases of Glanzmann's thrombasthenia.
Br J Haematol
1974
, vol. 
28
 (pg. 
253
-
260
)
24
Phillips
 
DR
Agin
 
PP
Platelet membrane defects in Glanzmann's thrombasthenia. Evidence for decreased amounts of two major glycoproteins.
J Clin Invest
1977
, vol. 
60
 (pg. 
535
-
545
)
25
Nurden
 
AT
Platelet membrane glycoproteins: a look back into the past and a view to the future [review].
Thromb Haemost
2007
, vol. 
98
 (pg. 
49
-
54
)
26
George
 
JN
Nurden
 
AT
Phillips
 
DR
Molecular defects in interactions of platelets with the vessel wall [review].
N Engl J Med
1984
, vol. 
311
 (pg. 
1084
-
1098
)
27
Hagen
 
I
Nurden
 
AT
Bjerrum
 
OJ
Solum
 
NO
Caen
 
JP
Immunochemical evidence for protein abnormalities from patients with Glanzmann's thrombasthenia and Bernard-Soulier syndrome.
J Clin Invest
1980
, vol. 
65
 (pg. 
722
-
731
)
28
Kunicki
 
TJ
Pidard
 
D
Rosa
 
JP
Nurden
 
AT
The formation of Ca++-dependent complexes of platelet membrane glycoproteins IIb and IIIa in solution as determined by crossed immunoelectrophoresis.
Blood
1981
, vol. 
58
 (pg. 
268
-
278
)
29
Howard
 
L
Shulman
 
S
Sadanandan
 
S
Karpatkin
 
S
Crossed immunoelectrophoresis of human platelet membranes. The major antigen consists of a complex of glycoproteins, GPIIb and GPIIIa, held together by Ca2+ and missing in Glanzmann's thrombasthenia.
J Biol Chem
1982
, vol. 
257
 (pg. 
8331
-
8336
)
30
Mustard
 
JF
Packham
 
MA
Kinlough-Rathbone
 
RL
Perry
 
DW
Regoeczi
 
E
Fibrinogen and ADP-induced platelet aggregation.
Blood
1978
, vol. 
52
 (pg. 
453
-
466
)
31
Bennett
 
JS
Vilaire
 
G
Exposure of platelet fibrinogen receptors by ADP and epinephrine.
J Clin Invest
1979
, vol. 
64
 (pg. 
1393
-
1401
)
32
Marguerie
 
GA
Plow
 
EF
Edgington
 
TS
Human platelets possess an inducible and saturable receptor specific for fibrinogen.
J Biol Chem
1979
, vol. 
254
 (pg. 
5357
-
5363
)
33
Nachman
 
RL
Leung
 
LL
Complex formation of platelet membrane glycoproteins IIb and IIIa with fibrinogen.
J Clin Invest
1982
, vol. 
69
 (pg. 
263
-
269
)
34
Baldassare
 
JJ
Kahn
 
RA
Knipp
 
MA
Newman
 
PJ
Reconstruction of platelet proteins into phospholipid vesicles. Functional proteoliposomes.
J Clin Invest
1988
, vol. 
75
 (pg. 
35
-
39
)
35
Parise
 
LV
Phillips
 
DR
Reconstitution of the purified platelet fibrinogen receptor. Fibrinogen binding properties of the glycoprotein IIb-IIIa complex.
J Biol Chem
1985
, vol. 
260
 (pg. 
10698
-
10707
)
36
Tabilio
 
A
Rosa
 
JP
Testa
 
U
, et al. 
Expression of platelet membrane glycoproteins and alpha-granule proteins by a human erythroleukemia cell line (HEL).
EMBO J
1984
, vol. 
3
 (pg. 
453
-
459
)
37
Bray
 
PF
Rosa
 
JP
Johnston
 
GI
, et al. 
Platelet glycoprotein IIb. Chromosomal localization and tissue expression.
J Clin Invest
1987
, vol. 
80
 (pg. 
1812
-
1817
)
38
Thiagarajan
 
P
Shapiro
 
SS
Sweterlitsch
 
L
McCord
 
S
A human erythroleukemia cell line synthesizes a functionally active glycoprotein IIb-IIIa complex capable of binding fibrinogen.
Biochim Biophys Acta
1987
, vol. 
924
 (pg. 
127
-
134
)
39
Bray
 
PF
Rosa
 
JP
Lingappa
 
VR
, et al. 
Biogenesis of the platelet receptor for fibrinogen: evidence for separate precursors for glycoproteins IIb and IIIa.
Proc Natl Acad Sci U S A
1986
, vol. 
83
 (pg. 
1480
-
1484
)
40
Duperray
 
A
Troesch
 
A
Berthier
 
R
, et al. 
Biosynthesis and assembly of platelet GPIIb-IIIa in human megakaryocytes: evidence that assembly between pro-GPIIb and GPIIIa is a prerequisite for expression of the complex on the cell surface.
Blood
1989
, vol. 
74
 (pg. 
1603
-
1611
)
41
O'Toole
 
TE
Loftus
 
JC
Plow
 
EF
, et al. 
Efficient surface expression of platelet GPIIb-IIIa requires both subunits.
Blood
1989
, vol. 
74
 (pg. 
14
-
18
)
42
McEver
 
RP
Baenziger
 
NL
Majerus
 
PW
Isolation and quantitation of the platelet membrane glycoprotein deficient in thrombasthenia using a monoclonal hybridoma antibody.
J Clin Invest
1980
, vol. 
66
 (pg. 
1311
-
1318
)
43
Bennett
 
JS
Hoxie
 
JA
Leitman
 
SF
Vilaire
 
G
Cines
 
DB
Inhibition of fibrinogen binding to stimulated human platelets by a monoclonal antibody.
Proc Natl Acad Sci U S A
1983
, vol. 
80
 (pg. 
2417
-
2421
)
44
Coller
 
BS
Peerschke
 
EI
Scudder
 
LE
Sullivan
 
CA
A murine monoclonal antibody that completely blocks the binding of fibrinogen to platelets produces a thrombasthenic-like state in normal platelets and binds to glycoproteins IIb and/or IIIa.
J Clin Invest
1983
, vol. 
72
 (pg. 
325
-
338
)
45
Wagner
 
CL
Mascelli
 
MA
Neblock
 
DS
, et al. 
Analysis of GPIIb/IIIa receptor number by quantification of 7E3 binding to human platelets.
Blood
1996
, vol. 
88
 (pg. 
907
-
914
)
46
Woods
 
VL
Wolff
 
LE
Keller
 
DM
Resting platelets contain a substantial centrally located pool of glycoprotein IIb-IIIa complexes which may be accessible to some but not other extracellular proteins.
J Biol Chem
1986
, vol. 
261
 (pg. 
15242
-
15251
)
47
Wencel-Drake
 
JD
Plow
 
EF
Kunicki
 
TJ
, et al. 
Localization of internal pools of membrane glycoproteins involved in platelet adhesive responses.
Am J Pathol
1986
, vol. 
124
 (pg. 
324
-
334
)
48
Coller
 
BS
A new murine monoclonal antibody reports an activation-dependent change in the conformation and/or microenvironment of the platelet GPIIb/IIIa complex.
J Clin Invest
1985
, vol. 
76
 (pg. 
101
-
108
)
49
Shattil
 
SJ
Hoxie
 
JA
Cunningham
 
M
Brass
 
LF
Changes in the platelet membrane glycoprotein IIb.IIIa complex during platelet activation.
J Biol Chem
1985
, vol. 
260
 (pg. 
11107
-
11114
)
50
Frelinger
 
AL
Lam
 
SC
Plow
 
EF
, et al. 
Occupancy of an adhesive glycoprotein receptor modulates expression of an antigenic site involved in cell adhesion.
J Biol Chem
1988
, vol. 
263
 (pg. 
12397
-
12402
)
51
Honda
 
S
Tomiyama
 
Y
Pelletier
 
AJ
, et al. 
Topography of ligand-induced binding sites, including a novel cation-sensitive epitope (AP5) at the amino terminus, of the human integrin beta 3 subunit.
J Biol Chem
1995
, vol. 
270
 (pg. 
11947
-
11954
)
52
Plow
 
EF
McEver
 
RP
Coller
 
BS
, et al. 
Related binding mechanisms for fibrinogen, fibronectin, von Willebrand factor and thrombospondin on thrombin-stimulated human platelets.
Blood
1985
, vol. 
66
 (pg. 
724
-
727
)
53
Montgomery
 
RR
Kunicki
 
TJ
Taves
 
C
, et al. 
Diagnosis of Bernard-Soulier syndrome and Glanzmann's thrombasthenia with a monoclonal assay on whole blood.
J Clin Invest
1983
, vol. 
71
 (pg. 
385
-
389
)
54
Coller
 
BS
Seligsohn
 
U
Zivelin
 
A
, et al. 
Immunologic and biochemical characterization of homozygous and heterozygous Glanzmann's thrombasthenia in Iraqi-Jewish and Arab populations of Israel: comparison of techniques for carrier detection.
Br J Haematol
1986
, vol. 
62
 (pg. 
723
-
735
)
55
Seligsohn
 
U
Mibashan
 
RS
Rodeck
 
CH
, et al. 
Prenatal diagnosis of Glanzmann's thrombasthenia.
Lancet
1985
, vol. 
2
 pg. 
1419
 
56
Coller
 
BS
Seligsohn
 
U
Little
 
PA
Type I Glanzmann thrombasthenia patients from the Iraqi-Jewish and Arab populations in Israel can be differentiated by platelet glycoprotein IIIa immunoblot analysis.
Blood
1987
, vol. 
69
 (pg. 
1696
-
1703
)
57
Nurden
 
AT
Didry
 
D
Kieffer
 
N
McEver
 
RP
Residual amounts of glycoproteins IIb and IIIa may be present in the platelets of most patients with Glanzmann's thrombasthenia.
Blood
1985
, vol. 
65
 (pg. 
1021
-
1024
)
58
Seligsohn
 
U
Coller
 
BS
Zivelin
 
A
Plow
 
EF
Ginsberg
 
MH
Immunoblot analysis of platelet GPIIb in patients with Glanzmann thrombasthenia in Israel.
Br J Haematol
1989
, vol. 
72
 (pg. 
415
-
423
)
59
Shattil
 
SJ
Cunningham
 
M
Hoxie
 
JA
Detection of activated platelets in whole blood using activation-dependent monoclonal antibodies and flow cytometry.
Blood
1987
, vol. 
70
 (pg. 
307
-
315
)
60
Ginsberg
 
MH
Frelinger
 
AL
Lam
 
SC
, et al. 
Analysis of platelet aggregation disorders based on flow cytometric analysis of membrane glycoprotein IIb-IIIa with conformation-specific monoclonal antibodies.
Blood
1990
, vol. 
76
 (pg. 
2017
-
2023
)
61
Poncz
 
M
Eisman
 
R
Heidenreich
 
R
, et al. 
Structure of the platelet membrane glycoprotein IIb: homology to the alpha subunits of the vitronectin and fibronectin membrane receptors.
J Biol Chem
1987
, vol. 
262
 (pg. 
8476
-
8482
)
62
Fitzgerald
 
LA
Steiner
 
B
Rall
 
SC
Lo
 
SS
Phillips
 
DR
Protein sequence of endothelial glycoprotein IIIa derived from a cDNA clone. Identity with platelet glycoprotein IIIa and similarity to “integrin.”
J Biol Chem
1987
, vol. 
262
 (pg. 
3936
-
3939
)
63
Thornton
 
MA
Poncz
 
M
Korostishevsky
 
M
, et al. 
The human platelet alphaIIb gene is not closely linked to its integrin partner beta3.
Blood
1999
, vol. 
94
 (pg. 
2039
-
2047
)
64
Bray
 
PF
Barsh
 
G
Rosa
 
JP
, et al. 
Physical linkage of the genes for platelet membrane glycoproteins IIb and IIIa.
Proc Natl Acad Sci U S A
1988
, vol. 
85
 (pg. 
8683
-
8687
)
65
Pytela
 
R
Pierschbacher
 
MD
Ginsberg
 
MH
Plow
 
EF
Ruoslahti
 
E
Platelet membrane glycoprotein IIb/IIIa: member of a family of Arg-Gly-Asp-specific adhesion receptors [review].
Science
1986
, vol. 
231
 (pg. 
1559
-
1562
)
66
Hynes
 
RO
Integrins: a family of cell surface receptors [review].
Cell
1987
, vol. 
48
 (pg. 
549
-
554
)
67
Ginsberg
 
MH
Loftus
 
J
Ryckwaert
 
JJ
, et al. 
Immunochemical and amino-terminal sequence comparison of two cytoadhesins indicates they contain similar or identical beta subunits and distinct alpha subunits.
J Biol Chem
1987
, vol. 
262
 (pg. 
5437
-
5440
)
68
Fitzgerald
 
LA
Poncz
 
M
Steiner
 
B
, et al. 
Comparison of cDNA-derived protein sequences of the human fibronectin and vitronectin receptor α subunits and platelet glycoprotein IIb.
Biochemistry
1987
, vol. 
26
 (pg. 
8158
-
8165
)
69
Byzova
 
TV
Rabbani
 
R
D'Souza
 
SE
Plow
 
EF
Role of integrin alpha (v) beta3 in vascular biology [review].
Thromb Haemost
1998
, vol. 
80
 (pg. 
726
-
734
)
70
Mt Sinai School of Medicine
Glanzmann Thrombasthenia Database
Accessed September 6, 2008 
71
French
 
DL
Seligsohn
 
U
Platelet glycoprotein IIb/IIIa receptors and Glanzmann's thrombasthenia [review].
Arterioscler Thromb Vasc Biol
2000
, vol. 
20
 (pg. 
607
-
610
)
72
MacKrell
 
AJ
Blumberg
 
B
Haynes
 
SR
Fessler
 
JH
The lethal myospheroid gene of Drosophila encodes a membrane protein homologous to vertebrate integrin beta subunits.
Proc Natl Acad Sci U S A
1988
, vol. 
85
 (pg. 
2633
-
2637
)
73
Kunicki
 
TJ
Nugent
 
DJ
Staats
 
SJ
, et al. 
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
1988
, vol. 
263
 (pg. 
4516
-
4519
)
74
Wayner
 
EA
Carter
 
WG
Piotrowicz
 
RS
Kunicki
 
TJ
The function of multiple extracellular matrix receptors in mediating cell adhesion to extracellular matrix: Preparation of monoclonal antibodies to the fibronectin receptor that specifically inhibit cell adhesion of fibronectin and react with platelet glycoproteins Ic-IIa.
J Cell Biol
1988
, vol. 
107
 (pg. 
1881
-
1891
)
75
Sonnenberg
 
A
Modderman
 
PW
Hogervorst
 
F
Laminin receptor on platelets is the integrin VLA-6.
Nature
1988
, vol. 
336
 (pg. 
487
-
489
)
76
Coller
 
BS
Cheresh
 
DA
Asch
 
E
Seligsohn
 
U
Platelet vitronectin receptor expression differentiates Iraqi-Jewish from Arab patients with Glanzmann thrombasthenia in Israel.
Blood
1991
, vol. 
77
 (pg. 
75
-
83
)
77
Newman
 
PJ
Seligsohn
 
U
Lyman
 
S
Coller
 
BS
The molecular genetic basis of Glanzmann thrombasthenia in the Iraqi-Jewish and Arab populations in Israel.
Proc Natl Acad Sci U S A
1991
, vol. 
88
 (pg. 
3160
-
3164
)
78
Ruoslahti
 
E
Fibronectin and its receptors [review].
Annu Rev Biochem
1988
, vol. 
57
 (pg. 
375
-
413
)
79
Gould
 
RJ
Polokoff
 
MA
Friedman
 
PA
, et al. 
Disintegrins: a family of integrin inhibitory proteins from viper venoms.
Proc Soc Exp Biol Med [review]
1990
, vol. 
195
 (pg. 
168
-
171
)
80
Ojima
 
I
Chakravarty
 
S
Dong
 
Q
Antithrombotic agents: from RGD to peptide mimetics [review].
Bioorg Med Chem
1995
, vol. 
3
 (pg. 
337
-
360
)
81
Kloczewiak
 
M
Timmons
 
S
Bednarek
 
MA
Sakon
 
M
Hawiger
 
J
Platelet receptor recognition domain on the gamma chain of human fibrinogen and its synthetic peptide analogues.
Biochemistry
1989
, vol. 
28
 (pg. 
2915
-
2919
)
82
Farrell
 
DH
Thiagarajan
 
P
Chung
 
DW
Davie
 
EW
Role of fibrinogen α and γ chain sites in platelet aggregation.
Proc Natl Acad Sci U S A
1992
, vol. 
89
 (pg. 
10729
-
10732
)
83
Newman
 
PJ
Gorski
 
J
White
 
GC
, et al. 
Enzymatic amplification of platelet-specific messenger RNA using the polymerase chain reaction.
J Clin Invest
1988
, vol. 
82
 (pg. 
739
-
743
)
84
Newman
 
PJ
Derbes
 
RS
Aster
 
RH
The human platelet alloantigens, PlA1 and PlA2, are associated with a leucine33/proline33 amino acid polymorphism in membrane glycoprotein IIIa, and are distinguishable by DNA typing.
J Clin Invest
1989
, vol. 
83
 (pg. 
1778
-
1781
)
85
Valentin
 
N
Newman
 
PJ
Human platelet alloantigens [review].
Curr Opin Hematol
1994
, vol. 
1
 (pg. 
381
-
387
)
86
Bray
 
PF
Integrin polymorphisms as risk factors for thrombosis [review].
Thromb Haemost
1999
, vol. 
82
 (pg. 
337
-
344
)
87
French
 
DL
Coller
 
BS
Berkowitz
 
R
, et al. 
Prenatal diagnosis of Glanzmann thrombasthenia using the polymorphic markers BRCA 1 and THRA 1 on chromosome 17.
Br J Haematol
1998
, vol. 
102
 (pg. 
582
-
587
)
88
Wautier
 
JL
Gruel
 
Y
Prenatal diagnosis of platelet disorders [review].
Baillieres Clin Haematol
1989
, vol. 
2
 (pg. 
569
-
583
)
89
Loftus
 
JC
O'Toole
 
TE
Plow
 
EF
, et al. 
A β3 integrin mutation abolishes ligand binding and alters divalent cation-dependent conformation.
Science
1990
, vol. 
249
 (pg. 
915
-
918
)
90
Lanza
 
F
Stierle
 
A
Fournier
 
D
, et al. 
A new variant of Glanzmann's thrombasthenia (Strasbourg I).. Platelets with functionally defective glycoprotein IIb-IIIa complexes and a glycoprotein IIIa Arg214Trp mutation.
J Clin Invest
1992
, vol. 
89
 (pg. 
1995
-
2004
)
91
Grimaldi
 
CM
Chen
 
F
Wu
 
C
, et al. 
Glycoprotein IIb Leu214Pro mutation produces Glanzmann thrombasthenia with both quantitative and qualitative abnormalities in GPIIb/IIIa.
Blood
1998
, vol. 
91
 (pg. 
1562
-
1568
)
92
Basani
 
RB
French
 
DL
Vilaire
 
G
, et al. 
A naturally-occurring mutation near the amino terminus of αIIb defines a new region involved in ligand binding to αIIbβ3.
Blood
2000
, vol. 
95
 (pg. 
180
-
188
)
93
Chen
 
Y-P
Djaffar
 
I
Pidard
 
E
Ser752Pro mutation in the cytoplasmic domain of integrin β3 subunit and defective activation of platelet integrin αIIbβ3 (glycoprotein IIb-IIIa) in a variant of Glanzmann thrombasthenia.
Proc Natl Acad Sci U S A
1992
, vol. 
89
 (pg. 
10169
-
10173
)
94
Wang
 
R
Shattil
 
SJ
Ambruso
 
DR
Newman
 
PJ
Truncation of the cytoplasmic domain of β3 in a variant form of Glanzmann thrombasthenia abrogates signaling through the integrin αIIbβ3 complex.
J Clin Invest
1997
, vol. 
100
 (pg. 
2393
-
2403
)
95
Ruiz
 
C
Liu
 
CY
Sun
 
QH
, et al. 
A point mutation in the cysteine-rich domain of glycoprotein (GP) IIIa results in the expression of a GPIIb-IIIa (alphaIIbbeta3) integrin receptor locked in a high-affinity state and a Glanzmann thrombasthenia-like phenotype.
Blood
2001
, vol. 
98
 (pg. 
2432
-
2441
)
96
Hodivala-Dilke
 
KM
McHugh
 
KP
Tsakiris
 
DA
, et al. 
Beta3-integrin-deficient mice are a model for Glanzmann thrombasthenia showing placental defects and reduced survival.
J Clin Invest
1999
, vol. 
103
 (pg. 
229
-
238
)
97
Smyth
 
SS
Reis
 
ED
Vaananen
 
H
Zhang
 
W
Coller
 
BS
Variable protection of β3-integrin-deficient mice from thrombosis initiated by different mechanisms.
Blood
2001
, vol. 
98
 (pg. 
1055
-
1062
)
98
Smyth
 
SS
Reis
 
ED
Zhang
 
W
, et al. 
β3-integrin-deficient mice, but not P-selectin-deficient mice, develop intimal hyperplasia after vascular injury: correlation with leukocyte recruitment to adherent platelets 1 hour after injury.
Circulation
2001
, vol. 
103
 (pg. 
2501
-
2507
)
99
McHugh
 
KP
Hodivala-Dilke
 
K
Zheng
 
MH
, et al. 
Mice lacking beta3 integrins are osteosclerotic because of dysfunctional osteoclasts.
J Clin Invest
2000
, vol. 
105
 (pg. 
433
-
440
)
100
Reynolds
 
LE
Conti
 
FJ
Lucas
 
M
, et al. 
Accelerated re-epithelialization in beta3-integrin-deficient- mice is associated with enhanced TGF-beta1 signaling.
Nat Med
2005
, vol. 
11
 (pg. 
167
-
174
)
101
Tronik-Le
 
RD
Roullot
 
V
Poujol
 
C
, et al. 
Thrombasthenic mice generated by replacement of the integrin alpha(IIb) gene: demonstration that transcriptional activation of this megakaryocytic locus precedes lineage commitment.
Blood
2000
, vol. 
96
 (pg. 
1399
-
1408
)
102
Emamboleus
 
NR
Frampton
 
J
The glycoprotein IIb molecule is expressed on early murine hematopoietic progenitors and regulates their numbers in sites of hematopoiesis.
Immunity
2003
, vol. 
19
 (pg. 
33
-
45
)
103
Law
 
DA
DeGuzmann
 
FR
Heiser
 
P
Ministri-Madrid
 
K
Phillips
 
DR
Integrin cytoplasmic tyrosine motif is required for outside-in alphaIIbbeta3 signalling and platelet function.
Nature
1999
, vol. 
401
 (pg. 
808
-
811
)
104
Zou
 
Z
Chen
 
H
Schmaier
 
AA
Hynes
 
RO
Kahn
 
ML
Structure-function analysis reveals discrete beta3 integrin inside-out and outside-in signaling pathways in platelets.
Blood
2007
, vol. 
109
 (pg. 
3284
-
3290
)
105
Petrich
 
BG
Fogelstrand
 
P
Partridge
 
AW
, et al. 
The antithrombotic potential of selective blockade of talin-dependent integrin alpha IIb beta 3 (platelet GPIIb-IIIa) activation.
J Clin Invest
2007
, vol. 
117
 (pg. 
2250
-
2259
)
106
Collins Tozer
 
EC
Liddington
 
RC
Sutcliffe
 
MJ
Smeeton
 
AH
Loftus
 
JC
Ligand binding to integrin αIIbβ3 is dependent on a MIDAS-like domain in the beta3 subunit.
J Biol Chem
1996
, vol. 
271
 (pg. 
21978
-
21984
)
107
Bajt
 
ML
Loftus
 
JC
Mutation of a ligand binding domain of beta 3 integrin: integral role of oxygenated residues in alpha IIb beta 3 (GPIIb-IIIa) receptor function.
J Biol Chem
1994
, vol. 
269
 (pg. 
20913
-
20919
)
108
Carrell
 
NA
Fitzgerald
 
LA
Steiner
 
B
Erickson
 
HP
Phillips
 
DR
Structure of human platelet membrane glycoproteins IIb and IIIa as determined by electron microscopy.
J Biol Chem
1985
, vol. 
260
 (pg. 
1743
-
1749
)
109
Weisel
 
JW
Nagaswami
 
C
Vilaire
 
G
Bennett
 
JS
Examination of the platelet membrane glycoprotein IIb-IIIa complex and its interaction with fibrinogen and other ligands by electron microscopy.
J Biol Chem
1992
, vol. 
267
 (pg. 
16637
-
16643
)
110
Du
 
X
Gu
 
M
Weisel
 
JW
, et al. 
Long range propagation of conformational changes in integrin alpha IIb beta 3.
J Biol Chem
1993
, vol. 
268
 (pg. 
23087
-
92
)
111
Xiong
 
JP
Stehle
 
T
Diefenbach
 
B
, et al. 
Crystal structure of the extracellular segment of integrin alphaVbeta3.
Science
2001
, vol. 
294
 (pg. 
339
-
345
)
112
Xiong
 
JP
Stehle
 
T
Zhang
 
R
, et al. 
Crystal structure of the extracellular segment of integrin alpha Vbeta3 in complex with an Arg-Gly-Asp ligand.
Science
2002
, vol. 
296
 (pg. 
151
-
155
)
113
Xiao
 
T
Takagi
 
J
Coller
 
BS
Wang
 
J
Springer
 
TA
Structural basis for allostery in integrins and binding to fibrinogen-mimetic therapeutics.
Nature
2004
, vol. 
432
 (pg. 
59
-
67
)
114
Adair
 
BD
Xiong
 
JP
Maddock
 
C
, et al. 
Three-dimensional EM structure of the ectodomain of integrin (alpha)V(beta)3 in a complex with fibronectin.
J Cell Biol
2005
, vol. 
168
 (pg. 
1109
-
1118
)
115
Arnaout
 
MA
Goodman
 
SL
Xiong
 
JP
Structure and mechanics of integrin-based cell adhesion.
Curr Opin Cell Biol
2007
, vol. 
19
 (pg. 
495
-
507
)
116
Zhu
 
J
Boylan
 
B
Luo
 
BH
Newman
 
PJ
Springer
 
TA
Tests of the extension and deadbolt models of integrin activation.
J Biol Chem
2007
, vol. 
282
 (pg. 
11914
-
11920
)
117
Luo
 
BH
Carman
 
CV
Springer
 
TA
Structural basis of integrin regulation and signaling.
Annu Rev Immunol
2007
, vol. 
25
 (pg. 
619
-
647
)
118
Mor-Cohen
 
R
Rosenberg
 
N
Peretz
 
H
, et al. 
Disulfide bond disruption by a beta 3-Cys549Arg mutation in six Jordanian families with Glanzmann thrombasthenia causes diminished production of constitutively active alpha IIb beta 3.
Thromb Haemost
2007
, vol. 
98
 (pg. 
1257
-
1265
)
119
Zucker
 
MB
Masiello
 
NC
Platelet aggregation caused by dithiothreitol.
Thromb Haemost
1984
, vol. 
51
 (pg. 
119
-
124
)
120
Yan
 
B
Smith
 
JW
A redox site involved in integrin activation.
J Biol Chem
2000
, vol. 
275
 (pg. 
39964
-
39972
)
121
Essex
 
DW
Li
 
M
Redox control of platelet aggregation.
Biochemistry
2003
, vol. 
42
 (pg. 
129
-
136
)
122
Lahav
 
J
Gofer-Dadosh
 
N
Luboshitz
 
J
Hess
 
O
Shaklai
 
M
Protein disulfide isomerase mediates integrin-dependent adhesion.
FEBS Lett
2000
, vol. 
475
 (pg. 
89
-
92
)
123
Walsh
 
GM
Sheehan
 
D
Kinsella
 
A
Moran
 
N
O'Neill
 
S
Redox modulation of integrin [correction of integin] alpha IIb beta 3 involves a novel allosteric regulation of its thiol isomerase activity.
Biochemistry
2004
, vol. 
43
 (pg. 
473
-
480
)
124
Li
 
R
Babu
 
CR
Lear
 
JD
, et al. 
Oligomerization of the integrin alphaIIbbeta3: roles of the transmembrane and cytoplasmic domains.
Proc Natl Acad Sci U S A
2001
, vol. 
98
 (pg. 
12462
-
12467
)
125
Hato
 
T
Pampori
 
N
Shattil
 
SJ
Complementary roles for receptor clustering and conformational change in the adhesive and signaling functions of integrin alphaIIb beta3.
J Cell Biol
1998
, vol. 
141
 (pg. 
1685
-
1695
)
126
Peerschke
 
EI
Reversible and irreversible binding of fibrinogen to platelets.
Platelets
1997
, vol. 
8
 (pg. 
311
-
317
)
127
Haas
 
TA
Plow
 
EF
Development of a structural model for the cytoplasmic domain of an integrin.
Protein Eng
1997
, vol. 
10
 (pg. 
1395
-
1405
)
128
Puklin-Faucher
 
E
Gao
 
M
Schulten
 
K
Vogel
 
V
How the headpiece hinge angle is opened: new insights into the dynamics of integrin activation.
J Cell Biol
2006
, vol. 
175
 (pg. 
349
-
360
)
129
Murcia
 
M
Jirouskova
 
M
Li
 
J
Coller
 
BS
Filizola
 
M
Functional and computational studies of the ligand-associated metal binding site of beta3 integrins.
Proteins
2008
, vol. 
71
 (pg. 
1779
-
1791
)
130
Blue
 
R
Murcia
 
M
Karan
 
C
Jirouskova
 
M
Coller
 
BS
Application of high throughput screening to identify a novel αIIb-specific small molecule inhibitor of αIIbβ3-mediated platelet Interaction with fibrinogen.
Blood
2008
, vol. 
111
 (pg. 
1248
-
1256
)
131
Ulmer
 
TS
Calderwood
 
DA
Ginsberg
 
MH
Campbell
 
ID
Domain-specific interactions of talin with the membrane-proximal region of the integrin beta3 subunit.
Biochemistry
2003
, vol. 
42
 (pg. 
8307
-
8312
)
132
Lau
 
TL
Partridge
 
AW
Ginsberg
 
MH
Ulmer
 
TS
Structure of the integrin beta3 transmembrane segment in phospholipid bicelles and detergent micelles.
Biochemistry
2008
, vol. 
47
 (pg. 
4008
-
4016
)
133
Haas
 
TA
Plow
 
EF
The cytoplasmic domain of alphaIIb beta3. A ternary complex of the integrin alpha and beta subunits and a divalent cation.
J Biol Chem
1996
, vol. 
271
 (pg. 
6017
-
6026
)
134
Ma
 
YQ
Qin
 
J
Plow
 
EF
Platelet integrin alpha(IIb)beta(3): activation mechanisms [review].
J Thromb Haemost
2007
, vol. 
5
 (pg. 
1345
-
1352
)
135
Rocco
 
M
Rosano
 
C
Weisel
 
JW
Horita
 
DA
Hantgan
 
RR
Integrin conformational regulation: uncoupling extension/tail separation from changes in the head region by a multiresolution approach.
Structure
2008
, vol. 
16
 (pg. 
954
-
964
)
136
Ye
 
F
Liu
 
J
Winkler
 
H
Taylor
 
KA
Integrin alpha IIb beta 3 in a membrane environment remains the same height after Mn2+ activation when observed by cryoelectron tomography.
J Mol Biol
2008
, vol. 
378
 (pg. 
976
-
986
)
137
Tokuhira
 
M
Handa
 
M
Kamata
 
T
, et al. 
A novel regulatory epitope defined by a murine monoclonal antibody to the platelet GPIIb-IIIa complex (alpha IIb beta 3 integrin).
Thromb Haemost
1996
, vol. 
76
 (pg. 
1038
-
1046
)
138
Kouns
 
WC
Jennings
 
LK
Activation-independent exposure of the GPIIb-IIIa fibrinogen receptor.
Thromb Res
1991
, vol. 
63
 (pg. 
343
-
354
)
139
Du
 
XP
Plow
 
EF
Frelinger
 
AL
, et al. 
Ligands “activate” integrin alpha IIb beta 3 (platelet GPIIb-IIIa).
Cell
1991
, vol. 
65
 (pg. 
409
-
416
)
140
Hynes
 
R
Integrins: bidirectional, allosteric signaling machines.
Cell
2002
, vol. 
110
 (pg. 
673
-
687
)
141
Shattil
 
SJ
Newman
 
PJ
Integrins: dynamic scaffolds for adhesion and signaling in platelets [review].
Blood
2004
, vol. 
104
 (pg. 
1606
-
1615
)
142
Watson
 
SP
Auger
 
JM
McCarty
 
OJ
Pearce
 
AC
GPVI and integrin alphaIIb beta3 signaling in platelets [review].
J Thromb Haemost
2005
, vol. 
3
 (pg. 
1752
-
1762
)
143
Qin
 
J
Vinogradova
 
O
Plow
 
EF
Integrin bidirectional signaling: a molecular view [review].
PLoS Biol
2004
, vol. 
2
 (pg. 
726
-
729
)
144
Michelson
 
AD
Shattil
 
SJ
Watson
 
SP
Authi
 
KS
The use of flow cytometry to study platelet activation.
Platelets: A Practical Approach
1996
Oxford
Oxford University Press
(pg. 
111
-
129
)
145
McEver
 
RP
Martin
 
MN
A monoclonal antibody to a membrane glycoprotein binds only to activated platelets.
J Biol Chem
1984
, vol. 
259
 (pg. 
9799
-
9804
)
146
Hsu-Lin
 
S
Berman
 
CL
Furie
 
BC
August
 
D
Furie
 
B
A platelet membrane protein expressed during platelet activation and secretion. Studies using a monoclonal antibody specific for thrombin-activated platelets.
J Biol Chem
1984
, vol. 
259
 (pg. 
9121
-
9126
)
147
Ferrell
 
JE
Martin
 
GS
Tyrosine-specific protein phosphorylation is regulated by glycoprotein IIb-IIIa in platelets.
Proc Natl Acad Sci U S A
1989
, vol. 
86
 (pg. 
2234
-
2238
)
148
Smolenski
 
A
Bachmann
 
C
Reinhard
 
K
, et al. 
Analysis and regulation of vasodilator-stimulated phosphoprotein serine 239 phosphorylation in vitro and in intact cells using a phosphospecific monoclonal antibody.
J Biol Chem
1998
, vol. 
273
 (pg. 
20029
-
20035
)
149
O'Toole
 
TE
Mandelman
 
D
Forsyth
 
J
, et al. 
Modulation of the affinity of integrin αIIbβ3 (GPIIb-IIIa) by the cytoplasmic domain of alpha IIb.
Science
1991
, vol. 
254
 (pg. 
845
-
847
)
150
Mayadas
 
TN
Johnson
 
RC
Rayburn
 
H
Hynes
 
RO
Wagner
 
DD
Leukocyte rolling and extravasation are severely compromised in P selectin-deficient mice.
Cell
1993
, vol. 
74
 (pg. 
541
-
554
)
151
Langenbach
 
R
Morham
 
SG
Tiano
 
HF
, et al. 
Prostaglandin synthase 1 gene disruption in mice reduces arachidonic acid-induced inflammation and indomethacin-induced gastric ulceration.
Cell
1995
, vol. 
83
 (pg. 
483
-
492
)
152
Tadokoro
 
S
Shattil
 
SJ
Eto
 
K
, et al. 
Talin binding to integrin beta tails: a final common step in integrin activation.
Science
2003
, vol. 
302
 (pg. 
103
-
106
)
153
Brass
 
LF
Hoffman
 
R
Benz
 
E
Shattil
 
S
Molecular basis for platelet activation.
Hematology Basic Principles and Practice
2005
Philadelphia
Elsevier Churchill-Livingstone
(pg. 
1899
-
1914
)
154
Mustard
 
JF
Packham
 
MA
Factors influencing platelet function: adhesion, release, and aggregation [review].
Pharmacol Rev
1970
, vol. 
22
 (pg. 
97
-
187
)
155
Vu
 
TK
Hung
 
DT
Wheaton
 
VI
Coughlin
 
SR
Molecular cloning of a functional thrombin receptor reveals a novel proteolytic mechanism of receptor activation.
Cell
1991
, vol. 
64
 (pg. 
1057
-
1068
)
156
Kahn
 
ML
Zheng
 
YW
Huang
 
W
, et al. 
A dual thrombin receptor system for platelet activation.
Nature
1998
, vol. 
394
 (pg. 
690
-
694
)
157
Sambrano
 
GR
Weiss
 
EJ
Zheng
 
YW
Huang
 
W
Coughlin
 
SR
Role of thrombin signalling in platelets in haemostasis and thrombosis [review].
Nature
2001
, vol. 
413
 (pg. 
74
-
78
)
158
Fabre
 
JE
Nguyen
 
M
Latour
 
A
, et al. 
Decreased platelet aggregation, increased bleeding time and resistance to thromboembolism in P2Y1-deficient mice.
Nat Med
1999
, vol. 
5
 (pg. 
1199
-
1202
)
159
Hollopeter
 
G
Jantzen
 
HM
Vincent
 
D
, et al. 
Identification of the platelet ADP receptor targeted by antithrombotic drugs.
Nature
2001
, vol. 
409
 (pg. 
202
-
207
)
160
Thomas
 
DW
Mannon
 
RB
Mannon
 
PJ
, et al. 
Coagulation defects and altered hemodynamic responses in mice lacking receptors for thromboxane A2.
J Clin Invest
1998
, vol. 
102
 (pg. 
1994
-
2001
)
161
Varga-Szabo
 
D
Pleines
 
I
Nieswandt
 
B
Cell adhesion mechanisms in platelets [review].
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 (pg. 
403
-
412
)
162
Kaczmarek
 
E
Koziak
 
K
Sevigny
 
J
, et al. 
Identification and characterization of CD39/vascular ATP diphosphohydrolase.
J Biol Chem
1996
, vol. 
271
 (pg. 
33116
-
33122
)
163
Marcus
 
AJ
Broekman
 
MJ
Drosopoulos
 
JH
, et al. 
The endothelial cell ecto-ADPase responsible for inhibition of platelet function is CD39.
J Clin Invest
1997
, vol. 
99
 (pg. 
1351
-
1360
)
164
Samuelsson
 
B
Prostaglandins and thromboxanes.
Recent Prog Horm Res
1978
, vol. 
34
 (pg. 
239
-
258
)
165
Moncada
 
S
Radomski
 
MW
Palmer
 
RM
Endothelium-derived relaxing factor. Identification as nitric oxide and role in the control of vascular tone and platelet function.
Biochem Pharmacol
1988
, vol. 
37
 (pg. 
2495
-
2501
)
166
Naseem
 
KM
Riba
 
R
Unresolved roles of platelet nitric oxide synthase.
J Thromb Haemost
2008
, vol. 
6
 (pg. 
10
-
19
)
167
Kunapuli
 
SP
Dorsam
 
RT
Kim
 
S
Quinton
 
TM
Platelet purinergic receptors [review].
Curr Opin Pharmacol
2003
, vol. 
3
 (pg. 
175
-
180
)
168
Lee
 
DH
Blajchman
 
MA
Michelson
 
AD
Animal models.
Platelets
2007
San Diego
Academic Press
(pg. 
587
-
610
)
169
Tsakiris
 
DA
Scudder
 
L
Hodivala-Dilke
 
KM
Hynes
 
RO
Coller
 
BS
Hemostasis in the mouse (Mus musculus): a review.
Thromb Haemost
1999
, vol. 
81
 (pg. 
177
-
188
)
170
Jirouskova
 
M
Shet
 
AS
Johnson
 
GJ
A guide to murine platelet structure, function, assays, and genetic alterations [review].
J Thromb Haemost
2007
, vol. 
5
 (pg. 
661
-
669
)
171
Rao
 
AK
Jalagadugula
 
G
Sun
 
L
Inherited defects in platelet signaling mechanisms [review].
Semin Thromb Hemost
2004
, vol. 
30
 (pg. 
525
-
535
)
172
Wegener
 
KL
Partridge
 
AW
Han
 
J
, et al. 
Structural basis of integrin activation by talin.
Cell
2007
, vol. 
128
 (pg. 
171
-
182
)
173
Nieswandt
 
B
Moser
 
M
Pleines
 
I
, et al. 
Loss of talin1 in platelets abrogates integrin activation, platelet aggregation, and thrombus formation in vitro and in vivo.
J Exp Med
2007
, vol. 
204
 (pg. 
3113
-
3118
)
174
Petrich
 
BG
Marchese
 
P
Ruggeri
 
ZM
, et al. 
Talin is required for integrin-mediated platelet function in hemostasis and thrombosis.
J Exp Med
2007
, vol. 
204
 (pg. 
3103
-
3111
)
175
Chrzanowska-Wodnicka
 
M
Smyth
 
SS
Schoenwaelder
 
SM
Fischer
 
TH
White
 
GC
Rap1b is required for normal platelet function and hemostasis in mice.
J Clin Invest
2005
, vol. 
115
 (pg. 
680
-
687
)
176
Han
 
J
Lim
 
CJ
Watanabe
 
N
, et al. 
Reconstructing and deconstructing agonist-induced activation of integrin alphaIIbbeta3 [review].
Curr Biol
2006
, vol. 
16
 (pg. 
1796
-
1806
)
177
Woulfe
 
D
Jiang
 
H
Mortensen
 
R
Yang
 
J
Brass
 
LF
Activation of Rap1B by Gi family members in platelets.
J Biol Chem
2002
, vol. 
277
 (pg. 
23382
-
23390
)
178
Larson
 
MK
Chen
 
H
Kahn
 
ML
, et al. 
Identification of P2Y12-dependent and -independent mechanisms of glycoprotein VI-mediated Rap1 activation in platelets.
Blood
2003
, vol. 
101
 (pg. 
1409
-
1415
)
179
Crittenden
 
JR
Bergmeier
 
W
Zhang
 
Y
, et al. 
CalDAG-GEFI integrates signaling for platelet aggregation and thrombus formation.
Nat Med
2004
, vol. 
10
 (pg. 
982
-
986
)
180
Pasvolsky
 
R
Feigelson
 
SW
Kilic
 
SS
, et al. 
A LAD-III syndrome is associated with defective expression of the Rap-1 activator CalDAG-GEFI in lymphocytes, neutrophils, and platelets.
J Exp Med
2007
, vol. 
204
 (pg. 
1571
-
1582
)
181
Moser
 
M
Nieswandt
 
B
Ussar
 
S
Pozgajova
 
M
Fassler
 
R
Kindlin-3 is essential for integrin activation and platelet aggregation.
Nat Med
2008
, vol. 
14
 (pg. 
325
-
330
)
182
Ma
 
YQ
Qin
 
J
Wu
 
C
Plow
 
EF
Kindlin-2 (Mig-2): a co-activator of beta3 integrins.
J Cell Biol
2008
, vol. 
181
 (pg. 
439
-
446
)
183
Zimmerman
 
GA
Weyrich
 
AS
Signal-dependent protein synthesis by activated platelets: new pathways to altered phenotype and function [review].
Arterioscler Thromb Vasc Biol
2008
, vol. 
28
 (pg. 
s17
-
s24
)
184
Gasic
 
GJ
Gasic
 
TB
Stewart
 
CC
Antimetastatic effects associated with platelet reduction.
Proc Natl Acad Sci U S A
1968
, vol. 
61
 (pg. 
46
-
52
)
185
Evangelista
 
V
Manarini
 
S
Sideri
 
R
, et al. 
Platelet/polymorphonuclear leukocyte interaction: P-selectin triggers protein-tyrosine phosphorylation-dependent CD11b/CD18 adhesion: role of PSGL-1 as a signaling molecule.
Blood
1999
, vol. 
93
 (pg. 
876
-
885
)
186
Italiano
 
JE
Richardson
 
JL
Patel-Hett
 
S
, et al. 
Angiogenesis is regulated by a novel mechanism: pro- and antiangiogenic proteins are organized into separate platelet alpha granules and differentially released.
Blood
2008
, vol. 
111
 (pg. 
1227
-
1233
)
187
Zarbock
 
A
Polanowska-Grabowska
 
RK
Ley
 
K
Platelet-neutrophil-interactions: linking hemostasis and inflammation [review].
Blood Rev
2007
, vol. 
21
 (pg. 
99
-
111
)
188
Emambokus
 
NR
Frampton
 
J
The glycoprotein IIb molecule is expressed on early murine hematopoietic progenitors and regulates their numbers in sites of hematopoiesis.
Immunity
2003
, vol. 
19
 (pg. 
33
-
45
)
189
Mikkola
 
HK
Fujiwara
 
Y
Schlaeger
 
TM
Traver
 
D
Orkin
 
SH
Expression of CD41 marks the initiation of definitive hematopoiesis in the mouse embryo.
Blood
2003
, vol. 
101
 (pg. 
508
-
516
)
190
Jennings
 
LK
Fox
 
JE
Edwards
 
HH
Phillips
 
DR
Changes in the cytoskeletal structure of human platelets following thrombin activation.
J Biol Chem
1981
, vol. 
256
 pg. 
6927
 
191
Hartwig
 
JH
DeSisto
 
M
The cytoskeleton of the resting human blood platelet: structure of the membrane skeleton and its attachment to actin filaments.
J Cell Biol
1991
, vol. 
112
 (pg. 
407
-
425
)
192
Hartwig
 
JH
Mechanisms of actin rearrangements mediating platelet activation.
J Cell Biol
1992
, vol. 
118
 (pg. 
1421
-
1442
)
193
Leisner
 
TM
Yuan
 
W
DeNofrio
 
JC
Liu
 
J
Parise
 
LV
Tickling the tails: cytoplasmic domain proteins that regulate integrin alphaIIbbeta3 activation [review].
Curr Opin Hematol
2007
, vol. 
14
 (pg. 
255
-
261
)
194
Chen
 
YP
O'Toole
 
TE
Ylanne
 
J
Rosa
 
JP
Ginsberg
 
MH
A point mutation in the integrin beta 3 cytoplasmic domain (S752–>P) impairs bidirectional signaling through alpha IIb beta 3 (platelet glycoprotein IIb-IIIa).
Blood
1994
, vol. 
84
 (pg. 
1857
-
1865
)
195
Abtahian
 
F
Bezman
 
N
Clemens
 
R
, et al. 
Evidence for the requirement of ITAM domains but not SLP-76/Gads interaction for integrin signaling in hematopoietic cells.
Mol Cell Biol
2006
, vol. 
26
 (pg. 
6936
-
6949
)
196
Jakus
 
Z
Fodor
 
S
Abram
 
CL
Lowell
 
CA
Mocsai
 
A
Immunoreceptor-like signaling by beta 2 and beta 3 integrins [review].
Trends Cell Biol
2007
, vol. 
17
 (pg. 
493
-
501
)
197
Coller
 
BS
Platelet GPIIb/IIIa antagonists: the first anti-integrin receptor therapeutics [review].
J Clin Invest
1997
, vol. 
99
 (pg. 
1467
-
1471
)
198
Weiss
 
HJ
The discovery of the antiplatelet effect of aspirin: a personal reminiscence [review].
J Thromb Haemost
2003
, vol. 
1
 (pg. 
1869
-
1875
)
199
Mustard
 
JF
Aspirin [review].
J Thromb Haemost
2004
, vol. 
2
 (pg. 
336
-
337
)
200
Quick
 
AJ
Bleeding time after aspirin ingestion.
Lancet
1968
, vol. 
1
 pg. 
50
 
201
Marcus
 
AJ
The role of prostaglandins in platelet function [review].
Prog Hematol
1979
, vol. 
11
 (pg. 
147
-
171
)
202
Vane
 
JR
Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs.
Nat New Biol
1971
, vol. 
231
 (pg. 
232
-
235
)
203
Mann
 
CC
Plummer
 
ML
The Asprin Wars: Money, Medicine and 100 Years of Rampant Competition
1993
Boston
Harvard Business School Press
204
Collabroative Group, ISIS-2 (Second International Study of Infarct Survival)
Randomised trial of intravenous streptokinase, oral apsirin, both, or neither among 17 187 cases of suspected acute myocardial infarction: ISIS-2.
Lancet
1988
, vol. 
2
 (pg. 
349
-
360
)
205
Savi
 
P
Herbert
 
JM
Clopidogrel and ticlopidine: P2Y12 adenosine diphosphate-receptor antagonists for the prevention of atherothrombosis [review].
Semin Thromb Hemost
2005
, vol. 
31
 (pg. 
174
-
183
)
206
Coller
 
BS
Blockade of platelet GPIIb/IIIa receptors as an antithrombotic strategy [review].
Circulation
1995
, vol. 
92
 (pg. 
2373
-
2380
)
207
George
 
JN
Caen
 
JP
Nurden
 
AT
Glanzmann's thrombasthenia: the spectrum of clinical disease [review].
Blood
1990
, vol. 
75
 (pg. 
1383
-
1395
)
208
Coller
 
BS
Folts
 
JD
Smith
 
SR
Scudder
 
LE
Jordan
 
R
Abolition of in vivo platelet thrombus formation in primates with monoclonal antibodies to the platelet GPIIb/IIIa receptor: correlation with bleeding time, platelet aggregation and blockade of GPIIb/IIIa receptors.
Circulation
1989
, vol. 
80
 (pg. 
1766
-
1774
)
209
Gold
 
HK
Coller
 
BS
Yasuda
 
T
, et al. 
Rapid and sustained coronary artery recanalization with combined bolus injection of recombinant tissue-type plasminogen activator and monoclonal anti-platelet GPIIb/IIIa antibody in a dog model.
Circulation
1988
, vol. 
77
 (pg. 
670
-
677
)
210
Phillips
 
DR
Scarborough
 
RM
Clinical pharmacology of eptifibatide.
Am J Cardiol
1997
, vol. 
80
 (pg. 
11B
-
20B
)
211
Barrett
 
JS
Murphy
 
G
Peerlinck
 
K
, et al. 
Pharmacokinetics and pharmacodynamics of MK-383, a selective non-peptide platelet glycoprotein-IIb/IIIa receptor antagonist, in healthy men.
Clin Pharmacol Ther
1994
, vol. 
56
 (pg. 
377
-
388
)
212
Coller
 
BS
Scudder
 
LE
Beer
 
J
, et al. 
Monoclonal antibodies to platelet GPIIb/IIIa as antithrombotic agents [review].
Ann NY Acad Sci
1991
, vol. 
614
 (pg. 
193
-
213
)
213
Topol
 
EJ
Lincoff
 
AM
Kereiakes
 
DJ
, et al. 
Multi-year follow-up of abciximab therapy in three randomized, placebo-controlled trials of percutaneous coronary revascularization.
Am J Med
2002
, vol. 
113
 (pg. 
1
-
6
)
214
Curran
 
MP
Keating
 
GM
Eptifibatide: a review of its use in patients with acute coronary syndromes and/or undergoing percutaneous coronary intervention [review].
Drugs
2005
, vol. 
65
 (pg. 
2009
-
2035
)
215
Menozzi
 
A
Merlini
 
PA
Ardissino
 
D
Tirofiban in acute coronary syndromes [review].
Expert Rev Cardiovasc Ther
2005
, vol. 
3
 (pg. 
193
-
206
)
216
The EPILOG Investigators
Platelet glycoprotein IIb/IIIa receptor blockade and low-dose heparin during percutaneous coronary revascularization.
N Engl J Med
1997
, vol. 
336
 (pg. 
1689
-
1696
)
217
Aster
 
RH
Curtis
 
BR
Bougie
 
DW
, et al. 
Thrombocytopenia associated with the use of GPIIb/IIIa inhibitors: position paper of the ISTH working group on thrombocytopenia and GPIIb/IIIa inhibitors.
J Thromb Haemost
2006
, vol. 
4
 (pg. 
678
-
679
)
218
Aster
 
RH
Bougie
 
DW
Drug-induced immune thrombocytopenia [review].
N Engl J Med
2007
, vol. 
357
 (pg. 
580
-
587
)
219
De
 
LG
Suryapranata
 
H
Stone
 
GW
, et al. 
Abciximab as adjunctive therapy to reperfusion in acute ST-segment elevation myocardial infarction: a meta-analysis of randomized trials [review].
JAMA
2005
, vol. 
293
 (pg. 
1759
-
1765
)
220
Plosker
 
GL
Lyseng-Williamson
 
KA
Clopidogrel: a review of its use in the prevention of thrombosis [review].
Drugs
2007
, vol. 
67
 (pg. 
613
-
646
)
221
Ahrens
 
I
Smith
 
BK
Bode
 
C
Peter
 
K
Direct thrombin inhibition with bivalirudin as an antithrombotic strategy in general and interventional cardiology [review].
Expert Opin Drug Metab Toxicol
2007
, vol. 
3
 (pg. 
609
-
620
)
222
Van't Hof
 
AW
Ten Berg
 
J
Heestermans
 
T
, et al. 
Prehospital initiation of tirofiban in patients with ST-elevation myocardial infarction undergoing primary angioplasty (On-TIME2): a multicentre, double-blind, randomized controlled trial.
Lancet
, vol. 
372
 (pg. 
537
-
546
)
223
Ellis
 
SG
Tendera
 
M
de Belder
 
MA
, et al. 
Facilitated PCI in patients with ST elevation myocardial infarction.
N Eng J Med
, vol. 
358
 (pg. 
2205
-
2217
)
224
Thiele
 
H
Schindler
 
K
Friedenberger
 
J
, et al. 
Intracoronary compared with intravenous bolus abciximab application in patients with ST-elevation myocardial infarction undergoing primary percutaneous coronary intervention: the randomized Leipzig immediate percutaneous coronary intervention abciximab IV versus IC in ST-elevation myocardial infarction trial.
Circulation
2008
, vol. 
118
 (pg. 
49
-
57
)
225
Wohrle
 
J
Grebe
 
OC
Nusser
 
T
, et al. 
Reduction of major adverse cardiac events with intracoronary compared with intravenous bolus application of abciximab in patients with acute myocardial infarction or unstable angina undergoing coronary angioplasty.
Circulation
2003
, vol. 
107
 (pg. 
1840
-
1843
)
226
Gibson
 
CM
Zorkun
 
C
Kunadian
 
V
Intracoronary administration of abciximab in ST-elevation myocardial infarction.
Circulation
2008
, vol. 
118
 (pg. 
6
-
8
)
227
Svilaas
 
T
Vlaar
 
PJ
van der Horst
 
IC
, et al. 
Thrombus aspiration during primary percutaneous coronary intervention.
N Engl J Med
2008
, vol. 
358
 (pg. 
557
-
567
)
228
Di
 
MC
Dudek
 
D
Piscione
 
F
, et al. 
Immediate angioplasty versus standard therapy with rescue angioplasty after thrombolysis in the Combined Abciximab REteplase Stent Study in Acute Myocardial Infarction (CARESS-in-AMI): an open, prospective, randomised, multicentre trial.
Lancet
2008
, vol. 
371
 (pg. 
559
-
568
)
229
Mandava
 
P
Thiagarajan
 
P
Kent
 
TA
Glycoprotein IIb/IIIa antagonists in acute ischaemic stroke: current status and future directions [review].
Drugs
2008
, vol. 
68
 (pg. 
1019
-
1028
)
230
Cox
 
D
Oral GPIIb/IIIa antagonists: what went wrong [review]?
Curr Pharm Des
2004
, vol. 
10
 (pg. 
1587
-
1596
)
231
Quinn
 
MJ
Plow
 
EF
Topol
 
EJ
Platelet glycoprotein IIb/IIIa inhibitors: recognition of a two-edged sword [review]?
Circulation
2002
, vol. 
106
 (pg. 
379
-
385
)
232
Hantgan
 
RR
Stahle
 
MC
Connor
 
JH
Connor
 
RF
Mousa
 
SA
AlphaIIbbeta3 priming and clustering by orally active and intravenous integrin antagonists.
J Thromb Haemost
2007
, vol. 
5
 (pg. 
542
-
550
)
233
Poon
 
MC
Clinical use of recombinant human activated factor VII (rFVIIa) in the prevention and treatment of bleeding episodes in patients with Glanzmann's thrombasthenia [review].
Vasc Health Risk Manag
2007
, vol. 
3
 (pg. 
655
-
664
)
234
Bellucci
 
S
Devergie
 
A
Gluckman
 
E
, et al. 
Complete correction of Glanzmann's thrombasthenia by allogeneic bone marrow transplantation.
Br J Haematol
1985
, vol. 
59
 (pg. 
635
-
641
)
235
Connor
 
P
Khair
 
K
Liesner
 
R
, et al. 
Stem cell transplantation for children with Glanzmann thrombasthenia.
Br J Haematol
2008
, vol. 
140
 (pg. 
568
-
571
)
236
Flood
 
VH
Johnson
 
FL
Boshkov
 
LK
, et al. 
Sustained engraftment post bone marrow transplant despite anti-platelet antibodies in Glanzmann thrombasthenia.
Pediatr Blood Cancer
2005
, vol. 
45
 (pg. 
971
-
975
)
237
Fang
 
J
Hodivala-Dilke
 
K
Johnson
 
BD
, et al. 
Therapeutic expression of the platelet-specific integrin, αIIbβ3, in a murine model for Glanzmann thrombasthenia.
Blood
2005
, vol. 
106
 (pg. 
2671
-
2679
)
238
Wilcox
 
DA
White
 
GC
Gene therapy for platelet disorders: studies with Glanzmann's thrombasthenia [review].
J Thromb Haemost
2003
, vol. 
1
 (pg. 
2300
-
2311
)
239
Zingman
 
LV
Park
 
S
Olson
 
TM
Alekseev
 
AE
Terzic
 
A
Aminoglycoside-induced translational read-through in disease: overcoming nonsense mutations by pharmacogenetic therapy [review].
Clin Pharmacol Ther
2007
, vol. 
81
 (pg. 
99
-
103
)
240
Harrison
 
P
Frelinger
 
AL
Furman
 
MI
Michelson
 
AD
Measuring antiplatelet drug effects in the laboratory [review].
Thromb Res
2007
, vol. 
120
 (pg. 
323
-
336
)
241
Mitchell
 
WB
Li
 
J
Murcia
 
M
, et al. 
Mapping early conformational changes in alphaIIb and beta3 during biogenesis reveals a potential mechanism for alphaIIbbeta3 adopting its bent conformation.
Blood
2007
, vol. 
109
 (pg. 
3725
-
3732
)
Sign in via your Institution