Skip to main content
Erschienen in: Thrombosis Journal 1/2016

Open Access 01.10.2016 | Review

Platelets and platelet adhesion molecules: novel mechanisms of thrombosis and anti-thrombotic therapies

verfasst von: Xiaohong Ruby Xu, Naadiya Carrim, Miguel Antonio Dias Neves, Thomas McKeown, Tyler W. Stratton, Rodrigo Matos Pinto Coelho, Xi Lei, Pingguo Chen, Jianhua Xu, Xiangrong Dai, Benjamin Xiaoyi Li, Heyu Ni

Erschienen in: Thrombosis Journal | Sonderheft 1/2016

Abstract

Platelets are central mediators of thrombosis and hemostasis. At the site of vascular injury, platelet accumulation (i.e. adhesion and aggregation) constitutes the first wave of hemostasis. Blood coagulation, initiated by the coagulation cascades, is the second wave of thrombin generation and enhance phosphatidylserine exposure, can markedly potentiate cell-based thrombin generation and enhance blood coagulation. Recently, deposition of plasma fibronectin and other proteins onto the injured vessel wall has been identified as a new “protein wave of hemostasis” that occurs prior to platelet accumulation (i.e. the classical first wave of hemostasis). These three waves of hemostasis, in the event of atherosclerotic plaque rupture, may turn pathogenic, and cause uncontrolled vessel occlusion and thrombotic disorders (e.g. heart attack and stroke). Current anti-platelet therapies have significantly reduced cardiovascular mortality, however, on-treatment thrombotic events, thrombocytopenia, and bleeding complications are still major concerns that continue to motivate innovation and drive therapeutic advances. Emerging evidence has brought platelet adhesion molecules back into the spotlight as targets for the development of novel anti-thrombotic agents. These potential antiplatelet targets mainly include the platelet receptors glycoprotein (GP) Ib-IX-V complex, β3 integrins (αIIb subunit and PSI domain of β3 subunit) and GPVI. Numerous efforts have been made aiming to balance the efficacy of inhibiting thrombosis without compromising hemostasis. This mini-review will update the mechanisms of thrombosis and the current state of antiplatelet therapies, and will focus on platelet adhesion molecules and the novel anti-thrombotic therapies that target them.
Abkürzungen
ADP
Adenosine diphosphate
GLP-1R
Glucagon-like peptide 1 receptor
GP
Glycoprotein
ITAM
Immunoreceptor tyrosine-based activation motif
ITP
Idiopathic thrombocytopenic purpura
LLR
Leucine-rich repeat
PAR
Protease-activated receptor
PCI
Percutaneous coronary intervention
PDI
Protein disulphide isomerase
PSI
Plexin-semaphorin-integrin
TIA
Transient ischemic attack
TTP
Thrombotic thrombocytopenic purpura
VWF
von Willebrand factor

Background

Platelet adhesion, activation and aggregation are critical events in hemostasis and thrombosis [13]. Platelet adhesion molecules, αIIbβ3 integrin and the glycoprotein (GP) Ib-IX-V, are essential for these processes [46]. Other adhesion molecules, such as P-selectin, GPVI and cadherins, are also involved [710]. The important roles of adhesion molecules in normal hemostasis have been well demonstrated in bleeding disorders, for example, Glanzmann thrombasthenia (β3 integrin deficiency) [11] and Bernard-Soulier syndrome (GPIb-IX-V complex deficiency) [12]. However, under pathological conditions, excessive platelet function may lead to thrombotic diseases, such as myocardial infarction and ischemic stroke, which cause far more deaths each year than cancer or respiratory diseases [1, 2, 1315]. Therefore, antiplatelet agents are vital for the treatment of thrombosis [16]. For over a decade, dual antiplatelet therapy with clopidogrel and aspirin has been considered a key treatment of patients with acute coronary syndrome [17, 18]. Nonetheless, some patients undergoing this combination therapy continue to suffer from recurrent thrombotic events, likely a result of platelet activation and aggregation occurring independently of ADP or thromboxane A2 receptor-mediated signalling pathways [17]. Thus, attenuating platelet adhesion appears to be a desirable strategy in effectively controlling pathological thrombosis [18]. Further understanding of the interactions between platelet adhesion molecules and their binding partners is therefore crucial in developing novel anti-thrombotic therapies. This review briefly summarizes the current knowledge of thrombosis and antiplatelet therapies, introduces a number of major platelet adhesion molecules, and highlights some recent advances in the new mechanisms of thrombosis, and anti-thrombotic therapies that are in clinical trials (unless otherwise indicated). There are several excellent available reviews regarding antiplatelet therapies, such as ADP antagonists (e.g. P2Y12 inhibitors), thromboxane antagonists and PAR-1/4 inhibitors [17, 18]. This mini-review will mainly focus on the therapeutic developments targeting platelet adhesion molecules.

Review

Arterial thrombosis and current state of antiplatelet therapies

Arterial thrombosis at the site of atherosclerotic plaque rupture may lead to uncontrolled vessel occlusion, resulting in life-threatening consequences (e.g. unstable angina, myocardial infarction and ischemic stroke) [1, 2, 13]. During plaque rupture, subendothelial matrix proteins, like collagen, von Willebrand factor (VWF), fibrinogen, fibronectin and laminin are exposed to circulation, leading to the rapid response of platelets [6]. Inappropriate platelet adhesion, activation and aggregation promote excessive platelet plug formation. Activated platelets can also provide negatively-charged surfaces that harbor coagulation factors and markedly potentiate cell-based thrombin generation and blood coagulation [1, 2, 19, 20]. The evolving concept of the “protein wave of hemostasis” indicates a potential role of platelet-released plasma fibronectin in thrombosis and hemostasis [21, 22]. Thus, platelets are key mediators of atherothrombosis, which are actively involved in all three waves of thrombus formation: protein wave, platelet accumulation, and blood coagulation [21, 23].
Current FDA-approved antiplatelet therapies (Fig. 1) mainly aim to (i) inhibit thromboxane A2 synthesis, which inhibits platelet activation (e.g. aspirin and triflusal); (ii) antagonize the function of platelet P2Y12 receptors, (e.g. clopidogrel, prasugrel, and ticagrelor); (iii) inhibit platelet integrin αIIbβ3 activity, which inhibits platelet aggregation, (e.g. abciximab, eptifibatide, and tirofiban); (iv) inhibit phosphodiesterase, which increases platelet cAMP/cGMP levels (e.g. dipyridamole and cilostazol) [24]. These antiplatelet drugs have significantly reduced cardiovascular deaths. However, limitations of current therapies, such as weak/poor inhibition of platelet function, excessive bleeding, thrombocytopenia and unexpected platelet activation are concerns that drive therapeutic advances [18, 25, 26]. In 2014, the FDA approved Vorapaxar, a novel antagonist of the thrombin receptor protease-activated receptor 1 (PAR1), which reduces the risk of heart attack and stroke in patients with atherosclerosis or peripheral arterial disease [27, 28]. However, Vorapaxar must not be used in patients who have histories of stroke, transient ischemic attack (TIA) or intracranial hemorrhage, since it increases the risk of intracranial bleeding [28, 29].

Platelet adhesion molecules in hemostasis and thrombosis: novel mechanisms and therapeutic opportunities

Platelet adhesion molecules are proteins/receptors on the platelet surface that interact with other cells or the extracellular matrix, including the integrin family (e.g. α2β1, α5β1, α6β1, αLβ2, αIIbβ3, and ανβ3) [4, 30, 31], the immunoglobulin superfamily (e.g. GPVI, FcγRIIA, ICAM-2, PECAM-1, JAMs and Cadherin 6), the leucine-rich repeat family (LRR; e.g. GPIb-IX-V complex), and the C-type lectin receptor family (e.g. P-selectin and CLEC-2), etc. [3234]. Recent evidence has shown that platelet adhesion molecules play key roles in a variety of pathophysiological processes [23], such as hemostasis and thrombosis [4, 33], immune responses [35, 36], inflammation [3537], atherosclerosis [3840], lymphatic vessel development [4144], angiogenesis [4547], miscarriage [48, 49], and tumor metastasis [5052]. Platelets are versatile cells and the mechanisms of their diverse functions have emerged as hot research topics [23]. This review mainly focuses on their roles in thrombosis and as novel anti-thrombotic targets (Fig. 1).

The GPIb-IX-V complex: emerging targets of antiplatelet therapy

New insights into the GPIb-IX-V complex
Platelet GPIb-IX-V complex (LRR family protein) has approximately 50,000 copies/platelet. It is composed of one GPIbα subunit disulfide-linked to two molecules of GPIbβ, and non-covalently linked with GPIX and GPV in a 2:4:2:1 ratio [53]. GPIb-IX-V is a key platelet receptor in initiating platelet translocation and adhesion to the vessel wall during vascular injury, especially under high shear stress (e.g. in small or stenosed arteries) [54, 55]. Platelet translocation onto the subendothelium is mediated by the binding of GPIbα to the immobilized VWF, a multimeric adhesive protein secreted from activated endothelial cells and platelets. The crystal structure of the GPIbα N-terminal ligand-binding domain and the VWF A1 domain gives useful information regarding their interaction [56]. This interaction induces intracellular signalling events that can activate integrins, leading to platelet stable adhesion and subsequent platelet aggregation. Interestingly, platelet-derived VWF was recently shown not essential for hemostasis and thrombosis, but instead fosters thrombo-inflammatory diseases such as ischemic stroke in mice via a GPIb-dependent mechanism [57]. This suggests that targeting GPIbα-VWF may be a promising anti-thrombotic strategy, particularly in thrombo-inflammatory conditions.
Furthermore, GPIb-IX-V complex has a high affinity for thrombin [58, 59]. Two thrombin binding sites on GPIbα LRR C-terminal flank region have been revealed [58]. Consequently, thrombin can activate platelets via GPIbα in two ways [60]: accelerating the cleavage of PAR-1 and platelet activation [61], or direct signaling via GPIbα, particularly after cleaving GPV, which is generally considered a “brake” in GPIb-IX-V activation [62, 63]. It is currently unknown but it is reasonable to consider that targeting both VWF and thrombin binding sites of GPIbα might provide additional benefits in effectively controlling thrombosis.
GPIbα can also interact with multiple other ligands, leading to platelet activation (e.g. thrombospondin [64] and P-selectin), pro-coagulant activity (e.g. factors XI [65], XII [66], VIIa [67] and kininogen [68]), inflammatory responses (e.g. P-selectin [69, 70], αMβ2 [71]), arterial remodeling [72] and others. Recently, the antibody-GPIbα interaction in immune thrombocytopenia has been highlighted. Some anti-GPIbα antibodies cause platelet activation and desialylation (removal of sugars), followed by the clearance of desialylated platelets via Ashwell-Morell receptors on hepatocytes [73, 74].
Developing novel antiplatelet agents against GPIbα
Given the critical roles of GPIbα or GPIbα-VWF interactions in platelet adhesion, particularly under stenosis high-shear conditions, they are attractive targets in attenuating thrombosis [54, 75, 76]. Currently, two such agents are in active clinical trials. ALX-0081 (Caplacizumab), an anti-VWF humanized single-variable-domain immunoglobulin (Nanobody), binds to the A1 domains of VWF with high affinity [77]. The phase I and II clinical trials of ALX-0081 in patients with stable angina undergoing percutaneous coronary intervention (PCI) or high risk PCI patients have shown a promising antiplatelet effects, and a relatively safe profile [77, 78]. The phase III clinical trials will investigate its effects on acquired thrombotic thrombocytopenic purpura (TTP) [7981]. ARC1779, an anti-VWF aptamer, was previously reported as an encouraging agent; however, the clinical trial of ARC1779 was prematurely terminated [82]. These VWF inhibitors may be useful candidates for TTP treatment.
A direct anti-GPIbα drug, Anfibatide, is purified from the snake venom of Agkistrodon acutus [83, 84]. Notably, Anfibatide inhibits both VWF and α-thrombin binding to GPIbα, representing a more potent anti-thrombotic effect [85]. In experimental models, Anfibatide inhibited platelet adhesion, aggregation and thrombus formation, without increasing bleeding time [83]. The phase II human clinical trials have also shown the promise of Anfibatide being utilized as a novel antiplatelet agent in cardiovascular diseases without significantly affecting hemostasis in patients with non-ST segment elevation myocardial infarction (unpublished data) [85]. Additionally, anti-GPIbα antibody displayed a strong protective effect in the mouse stroke models without inducing significant intracranial bleeding [8688]. Anfibatide has also been shown as a candidate to treat ischemic stroke in experimental models [89] (the same may hold true for anti-VWF therapy) and deserves further investigation. There are some other preclinical agents targeting GPIbα that are under investigation, such as h6B4-Fab [90], GPG-290 [91], and anti-GPIbα NIT family monoclonal antibodies [92]. The generation of these novel antagonists is reaching the forefront of treatment against heart attack and stroke, although the efficacy and safety of these drugs remain to be further established or evaluated in human clinical trials. Notably, there are currently no clinically available direct GPIbα antagonists.

GPVI: a potential anti-thrombotic target

GPVI (immunoglobulin superfamily protein) is exclusively expressed on platelets and megakaryocytes. It is associated with the Fc receptor γ-chain, which contains an immunoreceptor tyrosine-based activation motif (ITAM). Cross-linking by ligands, such as collagen, leads to ITAM-dependent signalling, and platelet activation. A possible anti-thrombotic benefit of targeting PI3-kinase/Akt pathway on ITAM receptors was suggested [93]. Fibrin has also been identified as a new GPVI ligand [94]. The GPVI ectodomain interacts with immobilized fibrin, which amplifies thrombin generation, and promotes thrombus stabilization [94, 95].
The role of platelet GPVI in the pathogenesis of ischemic stroke has been gradually acknowledged [9698]. Notably, platelet adhesion/activation can enhance infarct growth by promoting an inflammatory response [88, 99, 100]. GPVI-mediated platelet activation can lead to the release of interleukin-1α that drives cerebrovascular inflammation [100]. GPVI may be thus a potential antiplatelet target [97, 101, 102]. In animal models, anti-GPVI protected against thrombosis, ischemia-reperfusion injury [103] and stroke [104]. In phase I clinical trials, Revacept (the humanized Fc fusion protein of the GPVI ectodomain), inhibited collagen-induced human platelet aggregation [105]. Phase II trials of Revacept in patients with carotid artery stenosis, TIA, or stroke are ongoing [106]. The efficacy and safety of Revacept in these patients will be further determined. Some other GPVI targeted agents that are under preclinical development, such as Losartan [107] and scFv9012 [108], have been shown to inhibit the binding of GPVI to collagen.

Platelet integrin receptors

Integrins are heterodimeric transmembrane receptors, which are involved in cell-cell and cell-matrix interactions [30]. There are six different integrins on platelet surfaces: α2β1, α5β1, α6β1, αLβ2, αIIbβ3, and ανβ3. Platelet integrin αIIbβ3 is the dominant integrin expressed on platelets. Given the critical roles of αIIbβ3 integrin in mediating platelet aggregation, αIIbβ3 antagonists have been widely used for nearly two decades.
Integrin αIIbβ3 as anti-thrombotic targets: lessons and opportunities
Approximately 17 % of total platelet surface proteins are αIIbβ3 integrin, which contains both αIIb and β3 subunits [4]. Platelet “outside-in” signals are induced following platelet adhesion and platelet activation (e.g. GPIbα-VWF, GPVI/α2β1-collagen, P2Y12-ADP, PARs-thrombin), resulting in an increased Ca2+ influx and ultimately “inside-out” signaling. These “inside-out” signals further drive the conformational changes of αIIbβ3, from a low to high affinity state for binding to its ligands (e.g. fibrinogen/fibrin, VWF, fibronectin, thrombospondin, vitronectin and unidentified “X” ligands) [109112].
Fibrinogen, a major prothrombotic ligand of αIIbβ3, has been documented to be required for platelet aggregation for over 50 years. However, platelet aggregation still occurs in the absence of fibrinogen and VWF, although in the absence of αIIbβ3, aggregation is abolished [5, 8, 21, 113116]. The discovery of “fibrinogen-independent platelet aggregation” demonstrates that unidentified αIIbβ3 ligands also mediate platelet aggregation [5, 8, 21, 113, 116], and have the potential to be novel anti-thrombotic targets. Interestingly, some ligands (e.g. plasma fibronectin, vitronectin) may block prothrombotic ligand (e.g. fibrinogen)-αIIbβ3 interactions and attenuate thrombosis [21, 117].
Three FDA-approved αIIbβ3 antagonists are available: Abciximab (ReoPro), Eptifibatide (Integrilin) and Tirofiban (Aggrastat) [118120]. Abciximab is a fragmented antibody that binds close to the ligand binding-pocket on αIIbβ3. Eptifibatide, isolated from snake venom, binds via a KGD sequence and is a competitive inhibitor for fibrinogen-αIIbβ3, whilst tirofiban is a small molecule RGD inhibitor. Currently, αIIbβ3 antagonists are used in patients undergoing PCI and significantly decrease the incidence of myocardial infarction and death [121]. However, these antagonists can induce further conformational changes in the β3 subunit that may have negative consequences, such as exposing previously hidden epitopes, and causing platelet activation [122]. αIIbβ3 antagonists are also associated with intracranial hemorrhage in patients with acute ischemic stroke [123]. Therefore, a safer and more specific on-target drug is required to provide better patient care. Recently, a novel αIIbβ3 antagonist, RUC-4 (a more potent and more soluble congener of RUC-2 that disrupts Mg2+ binding to the metal ion-dependent adhesion site of αIIbβ3), is suggested for prehospital therapy of myocardial infarction in animal models, without significantly priming the receptor to bind fibrinogen [124]. However, the possibility of increased bleeding with therapeutic doses of RUC-4 remains to be evaluated [124].
The plexin-semaphorin-integrin (PSI) domain, located near the N-terminus of the β3 subunit, is highly conserved across the integrin family in different species, and contains seven cysteine residues which have been implicated in regulating β2 integrin activation [125, 126]. Previous studies described a role for cysteine-derived thiol/disulfide groups in the conformational switches of the β3 integrin [127130]. Disulfide bond remodeling is mediated primarily by thiol isomerase enzymatic activity, which is derived from active CXXC thioredoxin motifs and plays a role in the activation of αIIbβ3 [131]. Our group has recently identified that integrin PSI domain has endogenous thiol isomerase function and could be a novel target for anti-thrombotic therapy (unpublished data) [132]. We found that both CXXC motifs of β3 integrin PSI domain are required to maintain the optimal enzyme function, since mutations to one or both of the CXXC motifs decrease or abolish their protein disulphide isomerase (PDI)-like activity. We developed anti-PSI monoclonal antibodies and found that these antibodies cross-reacted with β3 PSI domains of human and other species and specifically inhibited the PDI-like activity, integrin activation and reduced PAC-1 binding to β3 integrin. Importantly, anti-PSI abrogated murine and human platelet aggregation in vitro and thrombus growth ex vivo and in vivo in both small and large vessels without significantly affecting bleeding time or platelet count. Thus, integrin PSI domain contains endogenous PDI activity and is a key regulator of integrin activation that can be a new target for therapy.
Interestingly, targeting activated platelets αIIbβ3 has been considered into the development of novel fibrinolytic drugs, which may allow effective thrombolysis and thromboprophylaxis [14, 133]. For example, scFvSCE5 (a single-chain urokinase plasminogen activator fused to a small recombinant antibody that binds activated αIIbβ3) directly targets thrombi and exerts an effective thrombolysis [133]. A chimeric platelet-targeted urokinase prodrug (composed of a single-chain version of the variable region of an anti-αIIbβ3 mAb and a thrombin-activatable, low-molecular-weight pro-uPA) selectively targets new thrombus formation [134].
Other platelet integrins: α2β1, α6β1 and α5β1
Other integrin receptors may also be considered as novel anti-thrombotic targets [16, 135]. Platelet α2β1 promotes stable platelet adhesion to collagen and may be a viable option, since overexpression of α2β1 in humans increases atherothrombotic risk, but lower level of α2β1 does not enhance bleeding risk [16]. Experimental evidence shows that α2β1 inhibitors (e.g. snake venom EMS-16) reduced pathological thrombus formation in vivo [136138]. Platelet α6β1, the main receptor for laminin, plays a role in platelet adhesion/activation and arterial thrombosis, and may also be a new target [135]. Platelet α5β1, the major receptor for fibronectin, plays a supplementary role in platelet adhesion [139], but evidence is lacking regarding the anti-thrombotic benefits of antagonizing α5β1.

Other novel anti-thrombotic candidates: Glucagon-like peptide 1 receptor, P-selectin, CD40/CD40L, and Toll-like receptors

Strategies to target other platelet receptors beyond adhesive proteins have also been developed, such as P2Y12, PAR1, TP, 5HT2A antagonists [17, 140]. Interestingly, some chronic diseases, such as diabetes mellitus and atherosclerosis, are associated with arterial thrombosis [23, 141]. Recently, our group identified that a functional Glucagon-like peptide 1 receptor (GLP-1R) is expressed on human megakaryocytes and platelets [142]. Importantly, GLP-1R agonists (e.g. Exenatide), likely through increasing the intracellular cAMP levels, inhibit platelet function and thrombus formation [142]. This study provides important insights into why diabetic patients who are receiving GLP-1-targeted therapies have a reduced number of cardiovascular events [142, 143]. In addition, given the cross-talks between platelets and immune systems, thrombosis also intensively communicates with the inflammatory pathway [23]. Some anti-inflammatory/anti-atherosclerotic agents may therefore also indirectly inhibit thrombosis, especially in deep vein thrombosis [144]. For example, antagonists of P-selectin/PSGL-1, such as rPSGL-Ig [145], PSI-697 [146], PSI-421 [147], inhibit platelet-mediated leukocyte attachment and recruitment of procoagulant microparticles, and may represent a safe therapeutic intervention in accelerating thrombolysis [148]. Antagonists of CD40/CD40L [149], such as CD40 antibody, reduce atherosclerotic burden in a murine model [150]. In addition, as the important roles of Toll-like receptors in atherosclerosis are gradually recognized [151, 152], they may also be potential targets for the treatment of atherothrombosis.

Conclusions

Arterial thrombotic events, such as myocardial infarction and ischemic stroke, and venous thromboembolism, are three leading causes of morbidity and mortality worldwide [153]. Platelets play a central role in the pathogenesis of atherothrombosis, and contribute profoundly to the pathology of venous thrombosis [23]. Platelet adhesion molecules, act as the contacts between platelets and other cells or extracellular matrix proteins and, to a great extent, may determine the reactivity of platelets and thus are attractive anti-thrombotic targets (Fig. 1) [23]. Although evidence-based antiplatelet therapy has markedly improved patient care, on-treatment events and bleeding are still major concerns [17, 148].
Optimization of the use of currently available therapies, and improvements to the understanding of individual differences in response to anti-platelet treatments are still the most cost-effective treatment strategies [17, 148]. Additionally, improved understanding of the mechanisms of platelet accumulation has been critical for further developing novel antiplatelet therapies, such as the PAR1 antagonist Vorapaxar (recently approved by the FDA), GPIbα/VWF antagonists (e.g. ALX-0081 and Anfibatide; undergoing clinical trials), and GPVI antagonist (e.g. Revacept; undergoing clinical trials) (See section II. A-C). Another cost-effective strategy may be to repurpose already-established drugs by discovering novel mechanisms of action in anti-thrombotic diseases, such as the recently-identified GLP-1R agonist, Exenatide, an anti-diabetic drug that has potential anti-thrombotic effects [142, 154]. Future studies in the areas of atherothrombosis, inflammation, metabolic syndrome, diabetes, lipid metabolism and cancer-related thrombotic diseases in the next few years should advance our knowledge and the application of these and other new anti-platelet agents. Of note, clinical trials provide important evidence regarding the safety and efficacy of the treatments. However, difficulties such as narrow eligibility criteria, low enrollment of patients and the necessity to test the new drugs on top of the current dual antiplatelet therapy (e.g. aspirin and clopidogrel), may add complexity to the development of new drugs and also deserve our attention.

Acknowledgements

The authors would like to thank Dr. Richard O. Hynes, Dr. Zaverio M. Ruggeri, Dr. Denisa D. Wagner, and Dr. John Freedman for their long-term support for these research projects.

Declaration

Publication fees for this article have been funded by APSTH 2016.
This article has been published as part of Thrombosis Journal Volume 14 Supplement 1, 2016. The full contents of the supplement are available at https://​thrombosisjourna​l.​biomedcentral.​com/​articles/​supplements/​volume-14-supplement-1.

Funding

This work was supported by the Canadian Institutes of Health Research (MOP 119540, MOP 97918, and MOP 119551), Heart and Stroke Foundation of Canada (Ontario), Equipment Funds from Canada Foundation for Innovation, St. Michael’s Hospital, and Canadian Blood Services; and research Funds from CCOA Therapeutics Inc and Lee’s Pharmaceutical Holdings limited. X. R. X is a recipient of China National Scholarship award, Meredith & Malcolm Silver Scholarship in Cardiovascular Studies of Department of Laboratory Medicine and Pathobiology, and the Heart and Stroke/Richard Lewar Centre of Excellence Studentship award, University of Toronto. N.C. is a recipient of the Canadian Blood Services Postdoctoral Fellowship.

Availability of data and material

Not applicable.

Authors’ contributions

XRX and NC drafted the manuscript. MADN drew the figure. TM, TWS and RMPC contributed to preparation of the manuscript. XL, XD and BXL contributed to the original findings on the phase II human clinical trials of Anfibatide. PC and JX contributed to the original findings and further development of anti-PSI monoclonal antibodies. HN is the principal investigator who defined the topic and revised the manuscript. All of authors read, commented and approved the final manuscript.

Competing interests

J. X. is supported by the CCOA Therapeutics Inc. X. D. and B.X.L. are supported by the Lee’s Pharmaceutical Holdings limited. Some of the research fund of the projects is supported by CCOA Therapeutics and Lee’s Pharmaceutical Holdings limited. Canadian Blood Services have held the patents on the anti-GPIb NIT family monoclonal antibodies and anti-PSI monoclonal antibodies.
Not applicable.
Not applicable.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
3.
Zurück zum Zitat Xu XR, Gallant RC, Ni H. Platelets, immune-mediated thrombocytopenias, and fetal hemorrhage. Thromb Res. 2016;141 Suppl 2:S76–9.PubMedCrossRef Xu XR, Gallant RC, Ni H. Platelets, immune-mediated thrombocytopenias, and fetal hemorrhage. Thromb Res. 2016;141 Suppl 2:S76–9.PubMedCrossRef
4.
Zurück zum Zitat Ni H, Freedman J. Platelets in hemostasis and thrombosis: role of integrins and their ligands. Transfus Apher Sci. 2003;28:257–64.PubMedCrossRef Ni H, Freedman J. Platelets in hemostasis and thrombosis: role of integrins and their ligands. Transfus Apher Sci. 2003;28:257–64.PubMedCrossRef
5.
Zurück zum Zitat Yang H, Reheman A, Chen P, Zhu G, Hynes RO, Freedman J, et al. Fibrinogen and von Willebrand factor-independent platelet aggregation in vitro and in vivo. J Thromb Haemost. 2006;4:2230–7.PubMedCrossRef Yang H, Reheman A, Chen P, Zhu G, Hynes RO, Freedman J, et al. Fibrinogen and von Willebrand factor-independent platelet aggregation in vitro and in vivo. J Thromb Haemost. 2006;4:2230–7.PubMedCrossRef
6.
Zurück zum Zitat Wang Y, Gallant RC, Ni H. Extracellular matrix proteins in the regulation of thrombus formation. Curr Opin Hematol. 2016;23:280–7.PubMedCrossRef Wang Y, Gallant RC, Ni H. Extracellular matrix proteins in the regulation of thrombus formation. Curr Opin Hematol. 2016;23:280–7.PubMedCrossRef
7.
Zurück zum Zitat Moroi M, Jung SM, Okuma M, Shinmyozu K. A patient with platelets deficient in glycoprotein VI that lack both collagen-induced aggregation and adhesion. J Clin Invest. 1989;84:1440–5.PubMedPubMedCentralCrossRef Moroi M, Jung SM, Okuma M, Shinmyozu K. A patient with platelets deficient in glycoprotein VI that lack both collagen-induced aggregation and adhesion. J Clin Invest. 1989;84:1440–5.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Dunne E, Spring CM, Reheman A, Jin W, Berndt MC, Newman DK, et al. Cadherin 6 has a functional role in platelet aggregation and thrombus formation. Arterioscler Thromb Vasc Biol. 2012;32:1724–31.PubMedPubMedCentralCrossRef Dunne E, Spring CM, Reheman A, Jin W, Berndt MC, Newman DK, et al. Cadherin 6 has a functional role in platelet aggregation and thrombus formation. Arterioscler Thromb Vasc Biol. 2012;32:1724–31.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Palabrica T, Lobb R, Furie BC, Aronovitz M, Benjamin C, Hsu YM, et al. Leukocyte accumulation promoting fibrin deposition is mediated in vivo by P-selectin on adherent platelets. Nature. 1992;359:848–51.PubMedCrossRef Palabrica T, Lobb R, Furie BC, Aronovitz M, Benjamin C, Hsu YM, et al. Leukocyte accumulation promoting fibrin deposition is mediated in vivo by P-selectin on adherent platelets. Nature. 1992;359:848–51.PubMedCrossRef
10.
Zurück zum Zitat Yang H, Lang S, Zhai Z, Li L, Kahr WH, Chen P, et al. Fibrinogen is required for maintenance of platelet intracellular and cell-surface P-selectin expression. Blood. 2009;114:425–36.PubMedCrossRef Yang H, Lang S, Zhai Z, Li L, Kahr WH, Chen P, et al. Fibrinogen is required for maintenance of platelet intracellular and cell-surface P-selectin expression. Blood. 2009;114:425–36.PubMedCrossRef
11.
Zurück zum Zitat Nurden AT. Platelet membrane glycoproteins: a historical review. Semin Thromb Hemost. 2014;40:577–84.PubMedCrossRef Nurden AT. Platelet membrane glycoproteins: a historical review. Semin Thromb Hemost. 2014;40:577–84.PubMedCrossRef
12.
Zurück zum Zitat Lopez JA, Andrews RK, Afshar-Kharghan V, Berndt MC. Bernard-Soulier syndrome. Blood. 1998;91:4397–418.PubMed Lopez JA, Andrews RK, Afshar-Kharghan V, Berndt MC. Bernard-Soulier syndrome. Blood. 1998;91:4397–418.PubMed
13.
Zurück zum Zitat Jackson SP. Arterial thrombosis--insidious, unpredictable and deadly. Nat Med. 2011;17:1423–36.PubMedCrossRef Jackson SP. Arterial thrombosis--insidious, unpredictable and deadly. Nat Med. 2011;17:1423–36.PubMedCrossRef
14.
Zurück zum Zitat Reheman A, Xu X, Reddy EC, Ni H. Targeting activated platelets and fibrinolysis: hitting two birds with one stone. Circ Res. 2014;114:1070–3.PubMedCrossRef Reheman A, Xu X, Reddy EC, Ni H. Targeting activated platelets and fibrinolysis: hitting two birds with one stone. Circ Res. 2014;114:1070–3.PubMedCrossRef
15.
Zurück zum Zitat Writing Group Members, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, et al. Heart disease and stroke statistics-2016 update: a report from the American Heart Association. Circulation. 2016;133:e38–60.CrossRef Writing Group Members, Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, et al. Heart disease and stroke statistics-2016 update: a report from the American Heart Association. Circulation. 2016;133:e38–60.CrossRef
16.
Zurück zum Zitat Michelson AD. Antiplatelet therapies for the treatment of cardiovascular disease. Nat Rev Drug Discov. 2010;9:154–69.PubMedCrossRef Michelson AD. Antiplatelet therapies for the treatment of cardiovascular disease. Nat Rev Drug Discov. 2010;9:154–69.PubMedCrossRef
17.
Zurück zum Zitat Franchi F, Angiolillo DJ. Novel antiplatelet agents in acute coronary syndrome. Nat Rev Cardiol. 2015;12:30–47.PubMedCrossRef Franchi F, Angiolillo DJ. Novel antiplatelet agents in acute coronary syndrome. Nat Rev Cardiol. 2015;12:30–47.PubMedCrossRef
18.
Zurück zum Zitat Gachet C. Antiplatelet drugs: which targets for which treatments? J Thromb Haemost. 2015;13 Suppl 1:S313–22.PubMedCrossRef Gachet C. Antiplatelet drugs: which targets for which treatments? J Thromb Haemost. 2015;13 Suppl 1:S313–22.PubMedCrossRef
19.
Zurück zum Zitat Hou Y, Carrim N, Wang Y, Gallant RC, Marshall A, Ni H. Platelets in hemostasis and thrombosis: novel mechanisms of fibrinogen-independent platelet aggregation and fibronectin-mediated protein wave of hemostasis. J Biomed Res. 2015;29:437–44.PubMedCentral Hou Y, Carrim N, Wang Y, Gallant RC, Marshall A, Ni H. Platelets in hemostasis and thrombosis: novel mechanisms of fibrinogen-independent platelet aggregation and fibronectin-mediated protein wave of hemostasis. J Biomed Res. 2015;29:437–44.PubMedCentral
20.
Zurück zum Zitat Wang H, Bang KW, Blanchette VS, Nurden AT, Rand ML. Phosphatidylserine exposure, microparticle formation and mitochondrial depolarisation in Glanzmann thrombasthenia platelets. Thromb Haemost. 2014;111:1184–6.PubMedCrossRef Wang H, Bang KW, Blanchette VS, Nurden AT, Rand ML. Phosphatidylserine exposure, microparticle formation and mitochondrial depolarisation in Glanzmann thrombasthenia platelets. Thromb Haemost. 2014;111:1184–6.PubMedCrossRef
21.
Zurück zum Zitat Wang Y, Reheman A, Spring CM, Kalantari J, Marshall AH, Wolberg AS, et al. Plasma fibronectin supports hemostasis and regulates thrombosis. J Clin Invest. 2014;124:4281–93.PubMedPubMedCentralCrossRef Wang Y, Reheman A, Spring CM, Kalantari J, Marshall AH, Wolberg AS, et al. Plasma fibronectin supports hemostasis and regulates thrombosis. J Clin Invest. 2014;124:4281–93.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Xu XR, Zhang D, Oswald BE, Carrim N, Wang X, Hou Y, et al. Platelets are versatile cells: New discoveries in hemostasis, thrombosis, immune responses, tumor metastasis and beyond. Crit Rev Clin Lab Sci. 2016. Published online:1–69. doi: 10.1080/10408363.2016.1200008. Xu XR, Zhang D, Oswald BE, Carrim N, Wang X, Hou Y, et al. Platelets are versatile cells: New discoveries in hemostasis, thrombosis, immune responses, tumor metastasis and beyond. Crit Rev Clin Lab Sci. 2016. Published online:1–69. doi: 10.​1080/​10408363.​2016.​1200008.
24.
Zurück zum Zitat Metharom P, Berndt MC, Baker RI, Andrews RK. Current state and novel approaches of antiplatelet therapy. Arterioscler Thromb Vasc Biol. 2015;35:1327–38.PubMedCrossRef Metharom P, Berndt MC, Baker RI, Andrews RK. Current state and novel approaches of antiplatelet therapy. Arterioscler Thromb Vasc Biol. 2015;35:1327–38.PubMedCrossRef
25.
Zurück zum Zitat Michelson AD. Advances in antiplatelet therapy. Hematology Am Soc Hematol Educ Program. 2011;2011:62–9.PubMed Michelson AD. Advances in antiplatelet therapy. Hematology Am Soc Hematol Educ Program. 2011;2011:62–9.PubMed
26.
Zurück zum Zitat Jackson SP, Schoenwaelder SM. Antiplatelet therapy: in search of the ‘magic bullet’. Nat Rev Drug Discov. 2003;2:775–89.PubMedCrossRef Jackson SP, Schoenwaelder SM. Antiplatelet therapy: in search of the ‘magic bullet’. Nat Rev Drug Discov. 2003;2:775–89.PubMedCrossRef
27.
Zurück zum Zitat Morrow DA, Braunwald E, Bonaca MP, Ameriso SF, Dalby AJ, Fish MP, et al. Vorapaxar in the secondary prevention of atherothrombotic events. N Engl J Med. 2012;366:1404–13.PubMedCrossRef Morrow DA, Braunwald E, Bonaca MP, Ameriso SF, Dalby AJ, Fish MP, et al. Vorapaxar in the secondary prevention of atherothrombotic events. N Engl J Med. 2012;366:1404–13.PubMedCrossRef
30.
31.
Zurück zum Zitat Mou Y, Ni H, Wilkins JA. The selective inhibition of beta 1 and beta 7 integrin-mediated lymphocyte adhesion by bacitracin. J Immunol. 1998;161:6323–9.PubMed Mou Y, Ni H, Wilkins JA. The selective inhibition of beta 1 and beta 7 integrin-mediated lymphocyte adhesion by bacitracin. J Immunol. 1998;161:6323–9.PubMed
32.
Zurück zum Zitat Clemetson KJ, Clemetson JM. Platelet receptors. In: Michelson AD, editor. Platelets. 3rd ed. Amsterdam: Academic Press/Elsevier; 2013. p. 169–94.CrossRef Clemetson KJ, Clemetson JM. Platelet receptors. In: Michelson AD, editor. Platelets. 3rd ed. Amsterdam: Academic Press/Elsevier; 2013. p. 169–94.CrossRef
33.
Zurück zum Zitat Varga-Szabo D, Pleines I, Nieswandt B. Cell adhesion mechanisms in platelets. Arterioscler Thromb Vasc Biol. 2008;28:403–12.PubMedCrossRef Varga-Szabo D, Pleines I, Nieswandt B. Cell adhesion mechanisms in platelets. Arterioscler Thromb Vasc Biol. 2008;28:403–12.PubMedCrossRef
34.
Zurück zum Zitat Tamura S, Suzuki-Inoue K, Tsukiji N, Shirai T, Sasaki T, Osada M, et al. Podoplanin-positive periarteriolar stromal cells promote megakaryocyte growth and proplatelet formation in mice by CLEC-2. Blood. 2016;127:1701–10.PubMedCrossRef Tamura S, Suzuki-Inoue K, Tsukiji N, Shirai T, Sasaki T, Osada M, et al. Podoplanin-positive periarteriolar stromal cells promote megakaryocyte growth and proplatelet formation in mice by CLEC-2. Blood. 2016;127:1701–10.PubMedCrossRef
35.
Zurück zum Zitat Li C, Li J, Li Y, Lang S, Yougbare I, Zhu G, et al. Crosstalk between platelets and the immune system: old systems with new discoveries. Adv Hematol. 2012;2012:384685.PubMedPubMedCentral Li C, Li J, Li Y, Lang S, Yougbare I, Zhu G, et al. Crosstalk between platelets and the immune system: old systems with new discoveries. Adv Hematol. 2012;2012:384685.PubMedPubMedCentral
36.
Zurück zum Zitat Semple JW, Italiano Jr JE, Freedman J. Platelets and the immune continuum. Nat Rev Immunol. 2011;11:264–74.PubMedCrossRef Semple JW, Italiano Jr JE, Freedman J. Platelets and the immune continuum. Nat Rev Immunol. 2011;11:264–74.PubMedCrossRef
37.
Zurück zum Zitat Wagner DD, Burger PC. Platelets in inflammation and thrombosis. Arterioscler Thromb Vasc Biol. 2003;23:2131–7.PubMedCrossRef Wagner DD, Burger PC. Platelets in inflammation and thrombosis. Arterioscler Thromb Vasc Biol. 2003;23:2131–7.PubMedCrossRef
38.
Zurück zum Zitat Siegel-Axel D, Daub K, Seizer P, Lindemann S, Gawaz M. Platelet lipoprotein interplay: trigger of foam cell formation and driver of atherosclerosis. Cardiovasc Res. 2008;78:8–17.PubMedCrossRef Siegel-Axel D, Daub K, Seizer P, Lindemann S, Gawaz M. Platelet lipoprotein interplay: trigger of foam cell formation and driver of atherosclerosis. Cardiovasc Res. 2008;78:8–17.PubMedCrossRef
39.
Zurück zum Zitat Lindemann S, Kramer B, Seizer P, Gawaz M. Platelets, inflammation and atherosclerosis. J Thromb Haemost. 2007;5 Suppl 1:203–11.PubMedCrossRef Lindemann S, Kramer B, Seizer P, Gawaz M. Platelets, inflammation and atherosclerosis. J Thromb Haemost. 2007;5 Suppl 1:203–11.PubMedCrossRef
40.
Zurück zum Zitat Murphy AJ, Bijl N, Yvan-Charvet L, Welch CB, Bhagwat N, Reheman A, et al. Cholesterol efflux in megakaryocyte progenitors suppresses platelet production and thrombocytosis. Nat Med. 2013;19:586–94.PubMedPubMedCentralCrossRef Murphy AJ, Bijl N, Yvan-Charvet L, Welch CB, Bhagwat N, Reheman A, et al. Cholesterol efflux in megakaryocyte progenitors suppresses platelet production and thrombocytosis. Nat Med. 2013;19:586–94.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Hess PR, Rawnsley DR, Jakus Z, Yang Y, Sweet DT, Fu J, et al. Platelets mediate lymphovenous hemostasis to maintain blood-lymphatic separation throughout life. J Clin Invest. 2014;124:273–84.PubMedCrossRef Hess PR, Rawnsley DR, Jakus Z, Yang Y, Sweet DT, Fu J, et al. Platelets mediate lymphovenous hemostasis to maintain blood-lymphatic separation throughout life. J Clin Invest. 2014;124:273–84.PubMedCrossRef
42.
Zurück zum Zitat Navarro-Nunez L, Langan SA, Nash GB, Watson SP. The physiological and pathophysiological roles of platelet CLEC-2. Thromb Haemost. 2013;109:991–8.PubMedPubMedCentralCrossRef Navarro-Nunez L, Langan SA, Nash GB, Watson SP. The physiological and pathophysiological roles of platelet CLEC-2. Thromb Haemost. 2013;109:991–8.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Osada M, Inoue O, Ding G, Shirai T, Ichise H, Hirayama K, et al. Platelet activation receptor CLEC-2 regulates blood/lymphatic vessel separation by inhibiting proliferation, migration, and tube formation of lymphatic endothelial cells. J Biol Chem. 2012;287:22241–52.PubMedPubMedCentralCrossRef Osada M, Inoue O, Ding G, Shirai T, Ichise H, Hirayama K, et al. Platelet activation receptor CLEC-2 regulates blood/lymphatic vessel separation by inhibiting proliferation, migration, and tube formation of lymphatic endothelial cells. J Biol Chem. 2012;287:22241–52.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Herzog BH, Fu J, Wilson SJ, Hess PR, Sen A, McDaniel JM, et al. Podoplanin maintains high endothelial venule integrity by interacting with platelet CLEC-2. Nature. 2013;502:105–9.PubMedPubMedCentralCrossRef Herzog BH, Fu J, Wilson SJ, Hess PR, Sen A, McDaniel JM, et al. Podoplanin maintains high endothelial venule integrity by interacting with platelet CLEC-2. Nature. 2013;502:105–9.PubMedPubMedCentralCrossRef
45.
Zurück zum Zitat Italiano Jr JE, Richardson JL, Patel-Hett S, Battinelli E, Zaslavsky A, Short 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;111:1227–33.PubMedPubMedCentralCrossRef Italiano Jr JE, Richardson JL, Patel-Hett S, Battinelli E, Zaslavsky A, Short 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;111:1227–33.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Chatterjee M, Huang Z, Zhang W, Jiang L, Hultenby K, Zhu L, et al. Distinct platelet packaging, release, and surface expression of proangiogenic and antiangiogenic factors on different platelet stimuli. Blood. 2011;117:3907–11.PubMedCrossRef Chatterjee M, Huang Z, Zhang W, Jiang L, Hultenby K, Zhu L, et al. Distinct platelet packaging, release, and surface expression of proangiogenic and antiangiogenic factors on different platelet stimuli. Blood. 2011;117:3907–11.PubMedCrossRef
47.
Zurück zum Zitat Yougbare I, Lang S, Yang H, Chen P, Zhao X, Tai WS, et al. Maternal anti-platelet beta3 integrins impair angiogenesis and cause intracranial hemorrhage. J Clin Invest. 2015;125:1545–56.PubMedPubMedCentralCrossRef Yougbare I, Lang S, Yang H, Chen P, Zhao X, Tai WS, et al. Maternal anti-platelet beta3 integrins impair angiogenesis and cause intracranial hemorrhage. J Clin Invest. 2015;125:1545–56.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Li C, Piran S, Chen P, Lang S, Zarpellon A, Jin JW, et al. The maternal immune response to fetal platelet GPIbalpha causes frequent miscarriage in mice that can be prevented by intravenous IgG and anti-FcRn therapies. J Clin Invest. 2011;121:4537–47.PubMedPubMedCentralCrossRef Li C, Piran S, Chen P, Lang S, Zarpellon A, Jin JW, et al. The maternal immune response to fetal platelet GPIbalpha causes frequent miscarriage in mice that can be prevented by intravenous IgG and anti-FcRn therapies. J Clin Invest. 2011;121:4537–47.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Yougbare I, Wei-She T, Zdravic D, Chen P, Zhu G, Leong-Poi H, et al. Natural killer cells contribute to pathophysiology of placenta leading to miscarriage in fetal and neonatal alloimmune thrombocytopenia. Blood. 2015;126:2254.CrossRef Yougbare I, Wei-She T, Zdravic D, Chen P, Zhu G, Leong-Poi H, et al. Natural killer cells contribute to pathophysiology of placenta leading to miscarriage in fetal and neonatal alloimmune thrombocytopenia. Blood. 2015;126:2254.CrossRef
50.
Zurück zum Zitat Labelle M, Hynes RO. The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discov. 2012;2:1091–9.PubMedPubMedCentralCrossRef Labelle M, Hynes RO. The initial hours of metastasis: the importance of cooperative host-tumor cell interactions during hematogenous dissemination. Cancer Discov. 2012;2:1091–9.PubMedPubMedCentralCrossRef
51.
53.
Zurück zum Zitat Luo SZ, Mo X, Afshar-Kharghan V, Srinivasan S, Lopez JA, Li R. Glycoprotein Ibalpha forms disulfide bonds with 2 glycoprotein Ibbeta subunits in the resting platelet. Blood. 2007;109:603–9.PubMedPubMedCentralCrossRef Luo SZ, Mo X, Afshar-Kharghan V, Srinivasan S, Lopez JA, Li R. Glycoprotein Ibalpha forms disulfide bonds with 2 glycoprotein Ibbeta subunits in the resting platelet. Blood. 2007;109:603–9.PubMedPubMedCentralCrossRef
54.
55.
Zurück zum Zitat Ruggeri ZM, Mendolicchio GL. Adhesion mechanisms in platelet function. Circ Res. 2007;100:1673–85.PubMedCrossRef Ruggeri ZM, Mendolicchio GL. Adhesion mechanisms in platelet function. Circ Res. 2007;100:1673–85.PubMedCrossRef
56.
Zurück zum Zitat Huizinga EG, Tsuji S, Romijn RA, Schiphorst ME, de Groot PG, Sixma JJ, et al. Structures of glycoprotein Ibalpha and its complex with von Willebrand factor A1 domain. Science. 2002;297:1176–9.PubMedCrossRef Huizinga EG, Tsuji S, Romijn RA, Schiphorst ME, de Groot PG, Sixma JJ, et al. Structures of glycoprotein Ibalpha and its complex with von Willebrand factor A1 domain. Science. 2002;297:1176–9.PubMedCrossRef
57.
Zurück zum Zitat Verhenne S, Denorme F, Libbrecht S, Vandenbulcke A, Pareyn I, Deckmyn H, et al. Platelet-derived VWF is not essential for normal thrombosis and hemostasis but fosters ischemic stroke injury in mice. Blood. 2015;126:1715–22.PubMedCrossRef Verhenne S, Denorme F, Libbrecht S, Vandenbulcke A, Pareyn I, Deckmyn H, et al. Platelet-derived VWF is not essential for normal thrombosis and hemostasis but fosters ischemic stroke injury in mice. Blood. 2015;126:1715–22.PubMedCrossRef
58.
Zurück zum Zitat Dumas JJ, Kumar R, Seehra J, Somers WS, Mosyak L. Crystal structure of the GpIbalpha-thrombin complex essential for platelet aggregation. Science. 2003;301:222–6.PubMedCrossRef Dumas JJ, Kumar R, Seehra J, Somers WS, Mosyak L. Crystal structure of the GpIbalpha-thrombin complex essential for platelet aggregation. Science. 2003;301:222–6.PubMedCrossRef
59.
Zurück zum Zitat Celikel R, McClintock RA, Roberts JR, Mendolicchio GL, Ware J, Varughese KI, et al. Modulation of alpha-thrombin function by distinct interactions with platelet glycoprotein Ibalpha. Science. 2003;301:218–21.PubMedCrossRef Celikel R, McClintock RA, Roberts JR, Mendolicchio GL, Ware J, Varughese KI, et al. Modulation of alpha-thrombin function by distinct interactions with platelet glycoprotein Ibalpha. Science. 2003;301:218–21.PubMedCrossRef
60.
Zurück zum Zitat Andrews RK, Berndt MC. The GPIb-IX-V Complex. In: Michelson AD, editor. Platelets. 3rd ed. Amsterdam: Academic Press/Elsevier; 2013. p. 195–213.CrossRef Andrews RK, Berndt MC. The GPIb-IX-V Complex. In: Michelson AD, editor. Platelets. 3rd ed. Amsterdam: Academic Press/Elsevier; 2013. p. 195–213.CrossRef
61.
Zurück zum Zitat De Candia E, Hall SW, Rutella S, Landolfi R, Andrews RK, De Cristofaro R. Binding of thrombin to glycoprotein Ib accelerates the hydrolysis of Par-1 on intact platelets. J Biol Chem. 2001;276:4692–8.PubMedCrossRef De Candia E, Hall SW, Rutella S, Landolfi R, Andrews RK, De Cristofaro R. Binding of thrombin to glycoprotein Ib accelerates the hydrolysis of Par-1 on intact platelets. J Biol Chem. 2001;276:4692–8.PubMedCrossRef
62.
Zurück zum Zitat Ramakrishnan V, DeGuzman F, Bao M, Hall SW, Leung LL, Phillips DR. A thrombin receptor function for platelet glycoprotein Ib-IX unmasked by cleavage of glycoprotein V. Proc Natl Acad Sci U S A. 2001;98:1823–8.PubMedPubMedCentralCrossRef Ramakrishnan V, DeGuzman F, Bao M, Hall SW, Leung LL, Phillips DR. A thrombin receptor function for platelet glycoprotein Ib-IX unmasked by cleavage of glycoprotein V. Proc Natl Acad Sci U S A. 2001;98:1823–8.PubMedPubMedCentralCrossRef
63.
Zurück zum Zitat Ni H, Ramakrishnan V, Ruggeri ZM, Papalia JM, Phillips DR, Wagner DD. Increased thrombogenesis and embolus formation in mice lacking glycoprotein V. Blood. 2001;98:368–73.PubMedCrossRef Ni H, Ramakrishnan V, Ruggeri ZM, Papalia JM, Phillips DR, Wagner DD. Increased thrombogenesis and embolus formation in mice lacking glycoprotein V. Blood. 2001;98:368–73.PubMedCrossRef
64.
Zurück zum Zitat Jurk K, Clemetson KJ, de Groot PG, Brodde MF, Steiner M, Savion N, et al. Thrombospondin-1 mediates platelet adhesion at high shear via glycoprotein Ib (GPIb): an alternative/backup mechanism to von Willebrand factor. FASEB J. 2003;17:1490–2.PubMed Jurk K, Clemetson KJ, de Groot PG, Brodde MF, Steiner M, Savion N, et al. Thrombospondin-1 mediates platelet adhesion at high shear via glycoprotein Ib (GPIb): an alternative/backup mechanism to von Willebrand factor. FASEB J. 2003;17:1490–2.PubMed
65.
Zurück zum Zitat Baglia FA, Badellino KO, Li CQ, Lopez JA, Walsh PN. Factor XI binding to the platelet glycoprotein Ib-IX-V complex promotes factor XI activation by thrombin. J Biol Chem. 2002;277:1662–8.PubMedCrossRef Baglia FA, Badellino KO, Li CQ, Lopez JA, Walsh PN. Factor XI binding to the platelet glycoprotein Ib-IX-V complex promotes factor XI activation by thrombin. J Biol Chem. 2002;277:1662–8.PubMedCrossRef
66.
Zurück zum Zitat Bradford HN, Pixley RA, Colman RW. Human factor XII binding to the glycoprotein Ib-IX-V complex inhibits thrombin-induced platelet aggregation. J Biol Chem. 2000;275:22756–63. Bradford HN, Pixley RA, Colman RW. Human factor XII binding to the glycoprotein Ib-IX-V complex inhibits thrombin-induced platelet aggregation. J Biol Chem. 2000;275:22756–63.
67.
Zurück zum Zitat Weeterings C, de Groot PG, Adelmeijer J, Lisman T. The glycoprotein Ib-IX-V complex contributes to tissue factor-independent thrombin generation by recombinant factor VIIa on the activated platelet surface. Blood. 2008;112:3227–33.PubMedCrossRef Weeterings C, de Groot PG, Adelmeijer J, Lisman T. The glycoprotein Ib-IX-V complex contributes to tissue factor-independent thrombin generation by recombinant factor VIIa on the activated platelet surface. Blood. 2008;112:3227–33.PubMedCrossRef
68.
Zurück zum Zitat Chavakis T, Santoso S, Clemetson KJ, Sachs UJ, Isordia-Salas I, Pixley RA, et al. High molecular weight kininogen regulates platelet-leukocyte interactions by bridging Mac-1 and glycoprotein Ib. J Biol Chem. 2003;278:45375–81.PubMedCrossRef Chavakis T, Santoso S, Clemetson KJ, Sachs UJ, Isordia-Salas I, Pixley RA, et al. High molecular weight kininogen regulates platelet-leukocyte interactions by bridging Mac-1 and glycoprotein Ib. J Biol Chem. 2003;278:45375–81.PubMedCrossRef
69.
Zurück zum Zitat Romo GM, Dong JF, Schade AJ, Gardiner EE, Kansas GS, Li CQ, et al. The glycoprotein Ib-IX-V complex is a platelet counterreceptor for P-selectin. J Exp Med. 1999;190:803–14.PubMedPubMedCentralCrossRef Romo GM, Dong JF, Schade AJ, Gardiner EE, Kansas GS, Li CQ, et al. The glycoprotein Ib-IX-V complex is a platelet counterreceptor for P-selectin. J Exp Med. 1999;190:803–14.PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat Kaplan ZS, Zarpellon A, Alwis I, Yuan Y, McFadyen J, Ghasemzadeh M, et al. Thrombin-dependent intravascular leukocyte trafficking regulated by fibrin and the platelet receptors GPIb and PAR4. Nat Commun. 2015;6:7835.PubMedCrossRef Kaplan ZS, Zarpellon A, Alwis I, Yuan Y, McFadyen J, Ghasemzadeh M, et al. Thrombin-dependent intravascular leukocyte trafficking regulated by fibrin and the platelet receptors GPIb and PAR4. Nat Commun. 2015;6:7835.PubMedCrossRef
71.
Zurück zum Zitat Simon DI, Chen Z, Xu H, Li CQ, Dong J, McIntire LV, et al. Platelet glycoprotein ibalpha is a counterreceptor for the leukocyte integrin Mac-1 (CD11b/CD18). J Exp Med. 2000;192:193–204.PubMedPubMedCentralCrossRef Simon DI, Chen Z, Xu H, Li CQ, Dong J, McIntire LV, et al. Platelet glycoprotein ibalpha is a counterreceptor for the leukocyte integrin Mac-1 (CD11b/CD18). J Exp Med. 2000;192:193–204.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Chandraratne S, von Bruehl ML, Pagel JI, Stark K, Kleinert E, Konrad I, et al. Critical role of platelet glycoprotein ibalpha in arterial remodeling. Arterioscler Thromb Vasc Biol. 2015;35:589–97.PubMedCrossRef Chandraratne S, von Bruehl ML, Pagel JI, Stark K, Kleinert E, Konrad I, et al. Critical role of platelet glycoprotein ibalpha in arterial remodeling. Arterioscler Thromb Vasc Biol. 2015;35:589–97.PubMedCrossRef
73.
Zurück zum Zitat Li J, van der Wal DE, Zhu G, Xu M, Yougbare I, Ma L, et al. Desialylation is a mechanism of Fc-independent platelet clearance and a therapeutic target in immune thrombocytopenia. Nat Commun. 2015;6:7737.PubMedPubMedCentralCrossRef Li J, van der Wal DE, Zhu G, Xu M, Yougbare I, Ma L, et al. Desialylation is a mechanism of Fc-independent platelet clearance and a therapeutic target in immune thrombocytopenia. Nat Commun. 2015;6:7737.PubMedPubMedCentralCrossRef
74.
Zurück zum Zitat Li J, Callum JL, Lin Y, Zhou Y, Zhu G, Ni H. Severe platelet desialylation in a patient with glycoprotein Ib/IX antibody-mediated immune thrombocytopenia and fatal pulmonary hemorrhage. Haematologica. 2014;99:e61–3.PubMedPubMedCentralCrossRef Li J, Callum JL, Lin Y, Zhou Y, Zhu G, Ni H. Severe platelet desialylation in a patient with glycoprotein Ib/IX antibody-mediated immune thrombocytopenia and fatal pulmonary hemorrhage. Haematologica. 2014;99:e61–3.PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat Gresele P, Momi S. Inhibitors of the interaction between von Willebrand factor and platelet GPIb/IX/V. Handb Exp Pharmacol. 2012;(210):287–309.CrossRef Gresele P, Momi S. Inhibitors of the interaction between von Willebrand factor and platelet GPIb/IX/V. Handb Exp Pharmacol. 2012;(210):287–309.CrossRef
76.
Zurück zum Zitat Ulrichts H, Silence K, Schoolmeester A, de Jaegere P, Rossenu S, Roodt J, et al. Antithrombotic drug candidate ALX-0081 shows superior preclinical efficacy and safety compared with currently marketed antiplatelet drugs. Blood. 2011;118:757–65.PubMedCrossRef Ulrichts H, Silence K, Schoolmeester A, de Jaegere P, Rossenu S, Roodt J, et al. Antithrombotic drug candidate ALX-0081 shows superior preclinical efficacy and safety compared with currently marketed antiplatelet drugs. Blood. 2011;118:757–65.PubMedCrossRef
77.
Zurück zum Zitat Bartunek J, Barbato E, Heyndrickx G, Vanderheyden M, Wijns W, Holz JB. Novel antiplatelet agents: ALX-0081, a Nanobody directed towards von Willebrand factor. J Cardiovasc Transl Res. 2013;6:355–63.PubMedCrossRef Bartunek J, Barbato E, Heyndrickx G, Vanderheyden M, Wijns W, Holz JB. Novel antiplatelet agents: ALX-0081, a Nanobody directed towards von Willebrand factor. J Cardiovasc Transl Res. 2013;6:355–63.PubMedCrossRef
78.
Zurück zum Zitat Muller O, Bartunek J, Hamilos M, Berza CT, Mangiacapra F, Ntalianis A, et al. von Willebrand factor inhibition improves endothelial function in patients with stable angina. J Cardiovasc Transl Res. 2013;6:364–70.PubMedCrossRef Muller O, Bartunek J, Hamilos M, Berza CT, Mangiacapra F, Ntalianis A, et al. von Willebrand factor inhibition improves endothelial function in patients with stable angina. J Cardiovasc Transl Res. 2013;6:364–70.PubMedCrossRef
79.
Zurück zum Zitat Peyvandi F, Scully M, Kremer Hovinga JA, Cataland S, Knobl P, Wu H, et al. Caplacizumab for acquired thrombotic thrombocytopenic purpura. N Engl J Med. 2016;374:511–22.PubMedCrossRef Peyvandi F, Scully M, Kremer Hovinga JA, Cataland S, Knobl P, Wu H, et al. Caplacizumab for acquired thrombotic thrombocytopenic purpura. N Engl J Med. 2016;374:511–22.PubMedCrossRef
80.
Zurück zum Zitat Lammle B. Thrombotic microangiopathy: caplacizumab accelerates resolution of acute acquired TTP. Nat Rev Nephrol. 2016;12:259–60.PubMedCrossRef Lammle B. Thrombotic microangiopathy: caplacizumab accelerates resolution of acute acquired TTP. Nat Rev Nephrol. 2016;12:259–60.PubMedCrossRef
81.
Zurück zum Zitat Von VA. Willebrand factor--a new target for TTP treatment? N Engl J Med. 2016;374:583–5.CrossRef Von VA. Willebrand factor--a new target for TTP treatment? N Engl J Med. 2016;374:583–5.CrossRef
82.
Zurück zum Zitat Blombery P, Scully M. Management of thrombotic thrombocytopenic purpura: current perspectives. J Blood Med. 2014;5:15–23.PubMedPubMedCentral Blombery P, Scully M. Management of thrombotic thrombocytopenic purpura: current perspectives. J Blood Med. 2014;5:15–23.PubMedPubMedCentral
83.
Zurück zum Zitat Lei X, Reheman A, Hou Y, Zhou H, Wang Y, Marshall AH, et al. Anfibatide, a novel GPIb complex antagonist, inhibits platelet adhesion and thrombus formation in vitro and in vivo in murine models of thrombosis. Thromb Haemost. 2014;111:279–89.PubMedCrossRef Lei X, Reheman A, Hou Y, Zhou H, Wang Y, Marshall AH, et al. Anfibatide, a novel GPIb complex antagonist, inhibits platelet adhesion and thrombus formation in vitro and in vivo in murine models of thrombosis. Thromb Haemost. 2014;111:279–89.PubMedCrossRef
84.
Zurück zum Zitat Hou Y, Li BX, Dai X, Yang Z, Qian F, Zhang G, et al. The first in vitro and in vivo assessment of anfibatide, a novel glycoprotein ib antagonist, in mice and in a phase i human clinical trial. Blood. 2013;122:577. Hou Y, Li BX, Dai X, Yang Z, Qian F, Zhang G, et al. The first in vitro and in vivo assessment of anfibatide, a novel glycoprotein ib antagonist, in mice and in a phase i human clinical trial. Blood. 2013;122:577.
85.
Zurück zum Zitat Li B, Dai X, Yang Z, Qian F, Zhang G, Xu Z, et al. First ex vivo and in vivo assessment of anfibatide, a novel glycoprotein Ib-IV-V complex antagonist, in healthy human volunteers in phase I clinical trial. J Thromb Haemost. 2013;11 Suppl 2:23. Li B, Dai X, Yang Z, Qian F, Zhang G, Xu Z, et al. First ex vivo and in vivo assessment of anfibatide, a novel glycoprotein Ib-IV-V complex antagonist, in healthy human volunteers in phase I clinical trial. J Thromb Haemost. 2013;11 Suppl 2:23.
87.
Zurück zum Zitat Stoll G, Kleinschnitz C, Nieswandt B. Molecular mechanisms of thrombus formation in ischemic stroke: novel insights and targets for treatment. Blood. 2008;112:3555–62.PubMedCrossRef Stoll G, Kleinschnitz C, Nieswandt B. Molecular mechanisms of thrombus formation in ischemic stroke: novel insights and targets for treatment. Blood. 2008;112:3555–62.PubMedCrossRef
88.
Zurück zum Zitat Kleinschnitz C, Pozgajova M, Pham M, Bendszus M, Nieswandt B, Stoll G. Targeting platelets in acute experimental stroke: impact of glycoprotein Ib, VI, and IIb/IIIa blockade on infarct size, functional outcome, and intracranial bleeding. Circulation. 2007;115:2323–30.PubMedCrossRef Kleinschnitz C, Pozgajova M, Pham M, Bendszus M, Nieswandt B, Stoll G. Targeting platelets in acute experimental stroke: impact of glycoprotein Ib, VI, and IIb/IIIa blockade on infarct size, functional outcome, and intracranial bleeding. Circulation. 2007;115:2323–30.PubMedCrossRef
89.
Zurück zum Zitat Li TT, Fan ML, Hou SX, Li XY, Barry DM, Jin H, Luo SY, Kong F, Lau LF, Dai XR, Zhang GH, Zhou LL. A novel snake venomderived GPIb antagonist, anfibatide, protects mice from acute experimental ischaemic stroke and reperfusion injury. Br J Pharmacol. Li TT, Fan ML, Hou SX, Li XY, Barry DM, Jin H, Luo SY, Kong F, Lau LF, Dai XR, Zhang GH, Zhou LL. A novel snake venomderived GPIb antagonist, anfibatide, protects mice from acute experimental ischaemic stroke and reperfusion injury. Br J Pharmacol.
90.
Zurück zum Zitat Fontayne A, Meiring M, Lamprecht S, Roodt J, Demarsin E, Barbeaux P, et al. The humanized anti-glycoprotein Ib monoclonal antibody h6B4-Fab is a potent and safe antithrombotic in a high shear arterial thrombosis model in baboons. Thromb Haemost. 2008;100:670–7.PubMed Fontayne A, Meiring M, Lamprecht S, Roodt J, Demarsin E, Barbeaux P, et al. The humanized anti-glycoprotein Ib monoclonal antibody h6B4-Fab is a potent and safe antithrombotic in a high shear arterial thrombosis model in baboons. Thromb Haemost. 2008;100:670–7.PubMed
91.
Zurück zum Zitat Hennan JK, Swillo RE, Morgan GA, Leik CE, Brooks JM, Shaw GD, et al. Pharmacologic inhibition of platelet vWF-GPIb alpha interaction prevents coronary artery thrombosis. Thromb Haemost. 2006;95:469–75.PubMed Hennan JK, Swillo RE, Morgan GA, Leik CE, Brooks JM, Shaw GD, et al. Pharmacologic inhibition of platelet vWF-GPIb alpha interaction prevents coronary artery thrombosis. Thromb Haemost. 2006;95:469–75.PubMed
92.
Zurück zum Zitat Ni H, Zhu G. Novel monoclonal antibodies against platelet GPIb-alpha: potential anti-thrombotic drugs and research reagents for study of thrombosis and hemostasis. 2012. US8323652. Ni H, Zhu G. Novel monoclonal antibodies against platelet GPIb-alpha: potential anti-thrombotic drugs and research reagents for study of thrombosis and hemostasis. 2012. US8323652.
93.
Zurück zum Zitat Moroi AJ, Watson SP. Impact of the PI3-kinase/Akt pathway on ITAM and hemITAM receptors: haemostasis, platelet activation and antithrombotic therapy. Biochem Pharmacol. 2015;94:186–94.PubMedCrossRef Moroi AJ, Watson SP. Impact of the PI3-kinase/Akt pathway on ITAM and hemITAM receptors: haemostasis, platelet activation and antithrombotic therapy. Biochem Pharmacol. 2015;94:186–94.PubMedCrossRef
94.
Zurück zum Zitat Alshehri OM, Hughes CE, Montague S, Watson SK, Frampton J, Bender M, et al. Fibrin activates GPVI in human and mouse platelets. Blood. 2015;126:1601–8.PubMedPubMedCentralCrossRef Alshehri OM, Hughes CE, Montague S, Watson SK, Frampton J, Bender M, et al. Fibrin activates GPVI in human and mouse platelets. Blood. 2015;126:1601–8.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Mammadova-Bach E, Ollivier V, Loyau S, Schaff M, Dumont B, Favier R, et al. Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood. 2015;126:683–91.PubMedCrossRef Mammadova-Bach E, Ollivier V, Loyau S, Schaff M, Dumont B, Favier R, et al. Platelet glycoprotein VI binds to polymerized fibrin and promotes thrombin generation. Blood. 2015;126:683–91.PubMedCrossRef
96.
Zurück zum Zitat Bigalke B, Stellos K, Geisler T, Kremmer E, Seizer P, May AE, et al. Expression of platelet glycoprotein VI is associated with transient ischemic attack and stroke. Eur J Neurol. 2010;17:111–7.PubMedCrossRef Bigalke B, Stellos K, Geisler T, Kremmer E, Seizer P, May AE, et al. Expression of platelet glycoprotein VI is associated with transient ischemic attack and stroke. Eur J Neurol. 2010;17:111–7.PubMedCrossRef
98.
Zurück zum Zitat Al-Tamimi M, Gardiner EE, Thom JY, Shen Y, Cooper MN, Hankey GJ, et al. Soluble glycoprotein VI is raised in the plasma of patients with acute ischemic stroke. Stroke. 2011;42:498–500.PubMedCrossRef Al-Tamimi M, Gardiner EE, Thom JY, Shen Y, Cooper MN, Hankey GJ, et al. Soluble glycoprotein VI is raised in the plasma of patients with acute ischemic stroke. Stroke. 2011;42:498–500.PubMedCrossRef
99.
Zurück zum Zitat Stoll G, Kleinschnitz C, Nieswandt B. Combating innate inflammation: a new paradigm for acute treatment of stroke? Ann N Y Acad Sci. 2010;1207:149–54.PubMedCrossRef Stoll G, Kleinschnitz C, Nieswandt B. Combating innate inflammation: a new paradigm for acute treatment of stroke? Ann N Y Acad Sci. 2010;1207:149–54.PubMedCrossRef
100.
Zurück zum Zitat Thornton P, McColl BW, Greenhalgh A, Denes A, Allan SM, Rothwell NJ. Platelet interleukin-1alpha drives cerebrovascular inflammation. Blood. 2010;115:3632–9.PubMedCrossRef Thornton P, McColl BW, Greenhalgh A, Denes A, Allan SM, Rothwell NJ. Platelet interleukin-1alpha drives cerebrovascular inflammation. Blood. 2010;115:3632–9.PubMedCrossRef
101.
Zurück zum Zitat Stegner D, Haining EJ, Nieswandt B. Targeting glycoprotein VI and the immunoreceptor tyrosine-based activation motif signaling pathway. Arterioscler Thromb Vasc Biol. 2014;34:1615–20.PubMedCrossRef Stegner D, Haining EJ, Nieswandt B. Targeting glycoprotein VI and the immunoreceptor tyrosine-based activation motif signaling pathway. Arterioscler Thromb Vasc Biol. 2014;34:1615–20.PubMedCrossRef
102.
Zurück zum Zitat Dutting S, Bender M, Nieswandt B. Platelet GPVI: a target for antithrombotic therapy?! Trends Pharmacol Sci. 2012;33:583–90.PubMedCrossRef Dutting S, Bender M, Nieswandt B. Platelet GPVI: a target for antithrombotic therapy?! Trends Pharmacol Sci. 2012;33:583–90.PubMedCrossRef
103.
Zurück zum Zitat Pachel C, Mathes D, Arias-Loza AP, Heitzmann W, Nordbeck P, Deppermann C, et al. Inhibition of platelet GPVI protects against myocardial ischemia-reperfusion injury. Arterioscler Thromb Vasc Biol. 2016;36:629–35.PubMedCrossRef Pachel C, Mathes D, Arias-Loza AP, Heitzmann W, Nordbeck P, Deppermann C, et al. Inhibition of platelet GPVI protects against myocardial ischemia-reperfusion injury. Arterioscler Thromb Vasc Biol. 2016;36:629–35.PubMedCrossRef
104.
Zurück zum Zitat Goebel S, Li Z, Vogelmann J, Holthoff HP, Degen H, Hermann DM, et al. The GPVI-Fc fusion protein Revacept improves cerebral infarct volume and functional outcome in stroke. PLoS One. 2013;8:e66960.PubMedPubMedCentralCrossRef Goebel S, Li Z, Vogelmann J, Holthoff HP, Degen H, Hermann DM, et al. The GPVI-Fc fusion protein Revacept improves cerebral infarct volume and functional outcome in stroke. PLoS One. 2013;8:e66960.PubMedPubMedCentralCrossRef
105.
Zurück zum Zitat Ungerer M, Rosport K, Bultmann A, Piechatzek R, Uhland K, Schlieper P, et al. Novel antiplatelet drug revacept (Dimeric Glycoprotein VI-Fc) specifically and efficiently inhibited collagen-induced platelet aggregation without affecting general hemostasis in humans. Circulation. 2011;123:1891–9.PubMedCrossRef Ungerer M, Rosport K, Bultmann A, Piechatzek R, Uhland K, Schlieper P, et al. Novel antiplatelet drug revacept (Dimeric Glycoprotein VI-Fc) specifically and efficiently inhibited collagen-induced platelet aggregation without affecting general hemostasis in humans. Circulation. 2011;123:1891–9.PubMedCrossRef
107.
Zurück zum Zitat Ono K, Ueda H, Yoshizawa Y, Akazawa D, Tanimura R, Shimada I, et al. Structural basis for platelet antiaggregation by angiotensin II type 1 receptor antagonist losartan (DuP-753) via glycoprotein VI. J Med Chem. 2010;53:2087–93.PubMedCrossRef Ono K, Ueda H, Yoshizawa Y, Akazawa D, Tanimura R, Shimada I, et al. Structural basis for platelet antiaggregation by angiotensin II type 1 receptor antagonist losartan (DuP-753) via glycoprotein VI. J Med Chem. 2010;53:2087–93.PubMedCrossRef
108.
Zurück zum Zitat Muzard J, Bouabdelli M, Zahid M, Ollivier V, Lacapere JJ, Jandrot-Perrus M, et al. Design and humanization of a murine scFv that blocks human platelet glycoprotein VI in vitro. FEBS J. 2009;276:4207–22.PubMedCrossRef Muzard J, Bouabdelli M, Zahid M, Ollivier V, Lacapere JJ, Jandrot-Perrus M, et al. Design and humanization of a murine scFv that blocks human platelet glycoprotein VI in vitro. FEBS J. 2009;276:4207–22.PubMedCrossRef
109.
Zurück zum Zitat Takagi J, Petre BM, Walz T, Springer TA. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell. 2002;110:599–611.PubMedCrossRef Takagi J, Petre BM, Walz T, Springer TA. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell. 2002;110:599–611.PubMedCrossRef
110.
Zurück zum Zitat Li R, Mitra N, Gratkowski H, Vilaire G, Litvinov R, Nagasami C, et al. Activation of integrin alphaIIbbeta3 by modulation of transmembrane helix associations. Science. 2003;300:795–8.PubMedCrossRef Li R, Mitra N, Gratkowski H, Vilaire G, Litvinov R, Nagasami C, et al. Activation of integrin alphaIIbbeta3 by modulation of transmembrane helix associations. Science. 2003;300:795–8.PubMedCrossRef
111.
Zurück zum Zitat Vinogradova O, Vaynberg J, Kong X, Haas TA, Plow EF, Qin J. Membrane-mediated structural transitions at the cytoplasmic face during integrin activation. Proc Natl Acad Sci U S A. 2004;101:4094–9.PubMedPubMedCentralCrossRef Vinogradova O, Vaynberg J, Kong X, Haas TA, Plow EF, Qin J. Membrane-mediated structural transitions at the cytoplasmic face during integrin activation. Proc Natl Acad Sci U S A. 2004;101:4094–9.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Vinogradova O, Velyvis A, Velyviene A, Hu B, Haas T, Plow E, et al. A structural mechanism of integrin alpha(IIb)beta(3) “inside-out” activation as regulated by its cytoplasmic face. Cell. 2002;110:587–97.PubMedCrossRef Vinogradova O, Velyvis A, Velyviene A, Hu B, Haas T, Plow E, et al. A structural mechanism of integrin alpha(IIb)beta(3) “inside-out” activation as regulated by its cytoplasmic face. Cell. 2002;110:587–97.PubMedCrossRef
113.
Zurück zum Zitat Ni H, Denis CV, Subbarao S, Degen JL, Sato TN, Hynes RO, et al. Persistence of platelet thrombus formation in arterioles of mice lacking both von Willebrand factor and fibrinogen. J Clin Invest. 2000;106:385–92.PubMedPubMedCentralCrossRef Ni H, Denis CV, Subbarao S, Degen JL, Sato TN, Hynes RO, et al. Persistence of platelet thrombus formation in arterioles of mice lacking both von Willebrand factor and fibrinogen. J Clin Invest. 2000;106:385–92.PubMedPubMedCentralCrossRef
114.
Zurück zum Zitat Law DA, DeGuzman FR, Heiser P, Ministri-Madrid K, Killeen N, Phillips DR. Integrin cytoplasmic tyrosine motif is required for outside-in alphaIIbbeta3 signalling and platelet function. Nature. 1999;401:808–11.PubMedCrossRef Law DA, DeGuzman FR, Heiser P, Ministri-Madrid K, Killeen N, Phillips DR. Integrin cytoplasmic tyrosine motif is required for outside-in alphaIIbbeta3 signalling and platelet function. Nature. 1999;401:808–11.PubMedCrossRef
115.
Zurück zum Zitat Hodivala-Dilke KM, McHugh KP, Tsakiris DA, Rayburn H, Crowley D, Ullman-Cullere M, et al. Beta3-integrin-deficient mice are a model for Glanzmann thrombasthenia showing placental defects and reduced survival. J Clin Invest. 1999;103:229–38.PubMedPubMedCentralCrossRef Hodivala-Dilke KM, McHugh KP, Tsakiris DA, Rayburn H, Crowley D, Ullman-Cullere M, et al. Beta3-integrin-deficient mice are a model for Glanzmann thrombasthenia showing placental defects and reduced survival. J Clin Invest. 1999;103:229–38.PubMedPubMedCentralCrossRef
116.
Zurück zum Zitat Reheman A, Yang H, Zhu G, Jin W, He F, Spring CM, et al. Plasma fibronectin depletion enhances platelet aggregation and thrombus formation in mice lacking fibrinogen and von Willebrand factor. Blood. 2009;113:1809–17.PubMedCrossRef Reheman A, Yang H, Zhu G, Jin W, He F, Spring CM, et al. Plasma fibronectin depletion enhances platelet aggregation and thrombus formation in mice lacking fibrinogen and von Willebrand factor. Blood. 2009;113:1809–17.PubMedCrossRef
117.
Zurück zum Zitat Reheman A, Gross P, Yang H, Chen P, Allen D, Leytin V, et al. Vitronectin stabilizes thrombi and vessel occlusion but plays a dual role in platelet aggregation. J Thromb Haemost. 2005;3:875–83.PubMedCrossRef Reheman A, Gross P, Yang H, Chen P, Allen D, Leytin V, et al. Vitronectin stabilizes thrombi and vessel occlusion but plays a dual role in platelet aggregation. J Thromb Haemost. 2005;3:875–83.PubMedCrossRef
118.
119.
Zurück zum Zitat Phillips DR, Scarborough RM. Clinical pharmacology of eptifibatide. Am J Cardiol. 1997;80:11B–20B.PubMedCrossRef Phillips DR, Scarborough RM. Clinical pharmacology of eptifibatide. Am J Cardiol. 1997;80:11B–20B.PubMedCrossRef
120.
Zurück zum Zitat Egbertson MS, Chang CT, Duggan ME, Gould RJ, Halczenko W, Hartman GD, et al. Non-peptide fibrinogen receptor antagonists. 2. Optimization of a tyrosine template as a mimic for Arg-Gly-Asp. J Med Chem. 1994;37:2537–51.PubMedCrossRef Egbertson MS, Chang CT, Duggan ME, Gould RJ, Halczenko W, Hartman GD, et al. Non-peptide fibrinogen receptor antagonists. 2. Optimization of a tyrosine template as a mimic for Arg-Gly-Asp. J Med Chem. 1994;37:2537–51.PubMedCrossRef
123.
Zurück zum Zitat Ciccone A, Motto C, Abraha I, Cozzolino F, Santilli I. Glycoprotein IIb-IIIa inhibitors for acute ischaemic stroke. Cochrane Database Syst Rev. 2014;3:CD005208.PubMed Ciccone A, Motto C, Abraha I, Cozzolino F, Santilli I. Glycoprotein IIb-IIIa inhibitors for acute ischaemic stroke. Cochrane Database Syst Rev. 2014;3:CD005208.PubMed
124.
Zurück zum Zitat Li J, Vootukuri S, Shang Y, Negri A, Jiang JK, Nedelman M, et al. RUC-4: a novel alphaIIbbeta3 antagonist for prehospital therapy of myocardial infarction. Arterioscler Thromb Vasc Biol. 2014;34:2321–9.PubMedPubMedCentralCrossRef Li J, Vootukuri S, Shang Y, Negri A, Jiang JK, Nedelman M, et al. RUC-4: a novel alphaIIbbeta3 antagonist for prehospital therapy of myocardial infarction. Arterioscler Thromb Vasc Biol. 2014;34:2321–9.PubMedPubMedCentralCrossRef
125.
Zurück zum Zitat Xiong JP, Stehle T, Goodman SL, Arnaout MA. A novel adaptation of the integrin PSI domain revealed from its crystal structure. J Biol Chem. 2004;279:40252–4.PubMedCrossRef Xiong JP, Stehle T, Goodman SL, Arnaout MA. A novel adaptation of the integrin PSI domain revealed from its crystal structure. J Biol Chem. 2004;279:40252–4.PubMedCrossRef
126.
Zurück zum Zitat Zang Q, Springer TA. Amino acid residues in the PSI domain and cysteine-rich repeats of the integrin beta2 subunit that restrain activation of the integrin alpha(X)beta(2). J Biol Chem. 2001;276:6922–9.PubMedCrossRef Zang Q, Springer TA. Amino acid residues in the PSI domain and cysteine-rich repeats of the integrin beta2 subunit that restrain activation of the integrin alpha(X)beta(2). J Biol Chem. 2001;276:6922–9.PubMedCrossRef
127.
Zurück zum Zitat Ni H, Li A, Simonsen N, Wilkins JA. Integrin activation by dithiothreitol or Mn2+ induces a ligand-occupied conformation and exposure of a novel NH2-terminal regulatory site on the beta1 integrin chain. J Biol Chem. 1998;273:7981–7.PubMedCrossRef Ni H, Li A, Simonsen N, Wilkins JA. Integrin activation by dithiothreitol or Mn2+ induces a ligand-occupied conformation and exposure of a novel NH2-terminal regulatory site on the beta1 integrin chain. J Biol Chem. 1998;273:7981–7.PubMedCrossRef
128.
Zurück zum Zitat Yan B, Smith JW. A redox site involved in integrin activation. J Biol Chem. 2000;275:39964–72.PubMedCrossRef Yan B, Smith JW. A redox site involved in integrin activation. J Biol Chem. 2000;275:39964–72.PubMedCrossRef
129.
130.
Zurück zum Zitat Manickam N, Ahmad SS, Essex DW. Vicinal thiols are required for activation of the alphaIIbbeta3 platelet integrin. J Thromb Haemost. 2011;9:1207–15.PubMedCrossRef Manickam N, Ahmad SS, Essex DW. Vicinal thiols are required for activation of the alphaIIbbeta3 platelet integrin. J Thromb Haemost. 2011;9:1207–15.PubMedCrossRef
131.
Zurück zum Zitat Wang L, Wu Y, Zhou J, Ahmad SS, Mutus B, Garbi N, et al. Platelet-derived ERp57 mediates platelet incorporation into a growing thrombus by regulation of the alphaIIbbeta3 integrin. Blood. 2013;122:3642–50.PubMedPubMedCentralCrossRef Wang L, Wu Y, Zhou J, Ahmad SS, Mutus B, Garbi N, et al. Platelet-derived ERp57 mediates platelet incorporation into a growing thrombus by regulation of the alphaIIbbeta3 integrin. Blood. 2013;122:3642–50.PubMedPubMedCentralCrossRef
132.
Zurück zum Zitat Carrim N, Zhu G, Reddy E, Xu M, Xu X, Wang Y, et al. Integrin PSI domain has endogenous thiol isomerase function and is a novel target for anti-thrombotic therapy. J Thromb Haemost. 2015;13(Supplement S2):60. Carrim N, Zhu G, Reddy E, Xu M, Xu X, Wang Y, et al. Integrin PSI domain has endogenous thiol isomerase function and is a novel target for anti-thrombotic therapy. J Thromb Haemost. 2015;13(Supplement S2):60.
133.
Zurück zum Zitat Wang X, Palasubramaniam J, Gkanatsas Y, Hohmann JD, Westein E, Kanojia R, et al. Towards effective and safe thrombolysis and thromboprophylaxis: preclinical testing of a novel antibody-targeted recombinant plasminogen activator directed against activated platelets. Circ Res. 2014;114:1083–93.PubMedCrossRef Wang X, Palasubramaniam J, Gkanatsas Y, Hohmann JD, Westein E, Kanojia R, et al. Towards effective and safe thrombolysis and thromboprophylaxis: preclinical testing of a novel antibody-targeted recombinant plasminogen activator directed against activated platelets. Circ Res. 2014;114:1083–93.PubMedCrossRef
134.
Zurück zum Zitat Fuentes RE, Zaitsev S, Ahn HS, Hayes V, Kowalska M, Lambert MP, et al. A chimeric platelet-targeted urokinase prodrug selectively blocks new thrombus formation. J Clin Invest. 2016;126:483–94.PubMedCrossRef Fuentes RE, Zaitsev S, Ahn HS, Hayes V, Kowalska M, Lambert MP, et al. A chimeric platelet-targeted urokinase prodrug selectively blocks new thrombus formation. J Clin Invest. 2016;126:483–94.PubMedCrossRef
135.
Zurück zum Zitat Schaff M, Tang C, Maurer E, Bourdon C, Receveur N, Eckly A, et al. Integrin alpha6beta1 is the main receptor for vascular laminins and plays a role in platelet adhesion, activation, and arterial thrombosis. Circulation. 2013;128:541–52.PubMedCrossRef Schaff M, Tang C, Maurer E, Bourdon C, Receveur N, Eckly A, et al. Integrin alpha6beta1 is the main receptor for vascular laminins and plays a role in platelet adhesion, activation, and arterial thrombosis. Circulation. 2013;128:541–52.PubMedCrossRef
136.
Zurück zum Zitat Miller MW, Basra S, Kulp DW, Billings PC, Choi S, Beavers MP, et al. Small-molecule inhibitors of integrin alpha2beta1 that prevent pathological thrombus formation via an allosteric mechanism. Proc Natl Acad Sci U S A. 2009;106:719–24.PubMedPubMedCentralCrossRef Miller MW, Basra S, Kulp DW, Billings PC, Choi S, Beavers MP, et al. Small-molecule inhibitors of integrin alpha2beta1 that prevent pathological thrombus formation via an allosteric mechanism. Proc Natl Acad Sci U S A. 2009;106:719–24.PubMedPubMedCentralCrossRef
137.
Zurück zum Zitat Marcinkiewicz C, Lobb RR, Marcinkiewicz MM, Daniel JL, Smith JB, Dangelmaier C, et al. Isolation and characterization of EMS16, a C-lectin type protein from Echis multisquamatus venom, a potent and selective inhibitor of the alpha2beta1 integrin. Biochemistry. 2000;39:9859–67.PubMedCrossRef Marcinkiewicz C, Lobb RR, Marcinkiewicz MM, Daniel JL, Smith JB, Dangelmaier C, et al. Isolation and characterization of EMS16, a C-lectin type protein from Echis multisquamatus venom, a potent and selective inhibitor of the alpha2beta1 integrin. Biochemistry. 2000;39:9859–67.PubMedCrossRef
138.
Zurück zum Zitat Arlinghaus FT, Momic T, Ammar NA, Shai E, Spectre G, Varon D, et al. Identification of alpha2beta1 integrin inhibitor VP-i with anti-platelet properties in the venom of Vipera palaestinae. Toxicon. 2013;64:96–105.PubMedCrossRef Arlinghaus FT, Momic T, Ammar NA, Shai E, Spectre G, Varon D, et al. Identification of alpha2beta1 integrin inhibitor VP-i with anti-platelet properties in the venom of Vipera palaestinae. Toxicon. 2013;64:96–105.PubMedCrossRef
139.
Zurück zum Zitat Piotrowicz RS, Orchekowski RP, Nugent DJ, Yamada KY, Kunicki TJ. Glycoprotein Ic-IIa functions as an activation-independent fibronectin receptor on human platelets. J Cell Biol. 1988;106:1359–64.PubMedCrossRef Piotrowicz RS, Orchekowski RP, Nugent DJ, Yamada KY, Kunicki TJ. Glycoprotein Ic-IIa functions as an activation-independent fibronectin receptor on human platelets. J Cell Biol. 1988;106:1359–64.PubMedCrossRef
140.
Zurück zum Zitat Przyklenk K, Frelinger 3rd AL, Linden MD, Whittaker P, Li Y, Barnard MR, et al. Targeted inhibition of the serotonin 5HT2A receptor improves coronary patency in an in vivo model of recurrent thrombosis. J Thromb Haemost. 2010;8:331–40.PubMedCrossRef Przyklenk K, Frelinger 3rd AL, Linden MD, Whittaker P, Li Y, Barnard MR, et al. Targeted inhibition of the serotonin 5HT2A receptor improves coronary patency in an in vivo model of recurrent thrombosis. J Thromb Haemost. 2010;8:331–40.PubMedCrossRef
142.
Zurück zum Zitat Cameron-Vendrig A, Reheman A, Siraj MA, Xu XR, Wang Y, Lei X, et al. Glucagon-like peptide 1 receptor activation attenuates platelet aggregation and thrombosis. Diabetes. 2016;65:1714–23.PubMedCrossRef Cameron-Vendrig A, Reheman A, Siraj MA, Xu XR, Wang Y, Lei X, et al. Glucagon-like peptide 1 receptor activation attenuates platelet aggregation and thrombosis. Diabetes. 2016;65:1714–23.PubMedCrossRef
143.
Zurück zum Zitat Monami M, Dicembrini I, Nardini C, Fiordelli I, Mannucci E. Effects of glucagon-like peptide-1 receptor agonists on cardiovascular risk: a meta-analysis of randomized clinical trials. Diabetes Obes Metab. 2014;16:38–47.PubMedCrossRef Monami M, Dicembrini I, Nardini C, Fiordelli I, Mannucci E. Effects of glucagon-like peptide-1 receptor agonists on cardiovascular risk: a meta-analysis of randomized clinical trials. Diabetes Obes Metab. 2014;16:38–47.PubMedCrossRef
145.
Zurück zum Zitat Kumar A, Villani MP, Patel UK, Keith Jr JC, Schaub RG. Recombinant soluble form of PSGL-1 accelerates thrombolysis and prevents reocclusion in a porcine model. Circulation. 1999;99:1363–9.PubMedCrossRef Kumar A, Villani MP, Patel UK, Keith Jr JC, Schaub RG. Recombinant soluble form of PSGL-1 accelerates thrombolysis and prevents reocclusion in a porcine model. Circulation. 1999;99:1363–9.PubMedCrossRef
146.
Zurück zum Zitat Bedard PW, Clerin V, Sushkova N, Tchernychev B, Antrilli T, Resmini C, et al. Characterization of the novel P-selectin inhibitor PSI-697 [2-(4-chlorobenzyl)-3-hydroxy-7,8,9,10-tetrahydrobenzo[h] quinoline-4-carboxylic acid] in vitro and in rodent models of vascular inflammation and thrombosis. J Pharmacol Exp Ther. 2008;324:497–506.PubMedCrossRef Bedard PW, Clerin V, Sushkova N, Tchernychev B, Antrilli T, Resmini C, et al. Characterization of the novel P-selectin inhibitor PSI-697 [2-(4-chlorobenzyl)-3-hydroxy-7,8,9,10-tetrahydrobenzo[h] quinoline-4-carboxylic acid] in vitro and in rodent models of vascular inflammation and thrombosis. J Pharmacol Exp Ther. 2008;324:497–506.PubMedCrossRef
147.
Zurück zum Zitat Meier TR, Myers Jr DD, Wrobleski SK, Zajkowski PJ, Hawley AE, Bedard PW, et al. Prophylactic P-selectin inhibition with PSI-421 promotes resolution of venous thrombosis without anticoagulation. Thromb Haemost. 2008;99:343–51.PubMed Meier TR, Myers Jr DD, Wrobleski SK, Zajkowski PJ, Hawley AE, Bedard PW, et al. Prophylactic P-selectin inhibition with PSI-421 promotes resolution of venous thrombosis without anticoagulation. Thromb Haemost. 2008;99:343–51.PubMed
148.
Zurück zum Zitat Kolandaivelu K, Bhatt DL. Novel antiplatelet therapies. In: Michelson AD, editor. Platelets. 3rd ed. Amsterdam: Academic Press/Elsevier; 2013. p. 1185–213.CrossRef Kolandaivelu K, Bhatt DL. Novel antiplatelet therapies. In: Michelson AD, editor. Platelets. 3rd ed. Amsterdam: Academic Press/Elsevier; 2013. p. 1185–213.CrossRef
149.
Zurück zum Zitat Conde ID, Kleiman NS. Soluble CD40 ligand in acute coronary syndromes. N Engl J Med. 2003;348:2575–7.PubMedCrossRef Conde ID, Kleiman NS. Soluble CD40 ligand in acute coronary syndromes. N Engl J Med. 2003;348:2575–7.PubMedCrossRef
150.
151.
Zurück zum Zitat Lin J, Kakkar V, Lu X. Essential roles of toll-like receptors in atherosclerosis. Curr Med Chem. 2016;23:431–54.PubMedCrossRef Lin J, Kakkar V, Lu X. Essential roles of toll-like receptors in atherosclerosis. Curr Med Chem. 2016;23:431–54.PubMedCrossRef
152.
Zurück zum Zitat Hovland A, Jonasson L, Garred P, Yndestad A, Aukrust P, Lappegard KT, et al. The complement system and toll-like receptors as integrated players in the pathophysiology of atherosclerosis. Atherosclerosis. 2015;241:480–94.PubMedCrossRef Hovland A, Jonasson L, Garred P, Yndestad A, Aukrust P, Lappegard KT, et al. The complement system and toll-like receptors as integrated players in the pathophysiology of atherosclerosis. Atherosclerosis. 2015;241:480–94.PubMedCrossRef
153.
Zurück zum Zitat Beckman MG, Hooper WC, Critchley SE, Ortel TL. Venous thromboembolism: a public health concern. Am J Prev Med. 2010;38:S495–501.PubMedCrossRef Beckman MG, Hooper WC, Critchley SE, Ortel TL. Venous thromboembolism: a public health concern. Am J Prev Med. 2010;38:S495–501.PubMedCrossRef
154.
Zurück zum Zitat Husain M, Aameron-Vendrig A, Ni H. Methods for inhibiting platelet aggregation using glp-1 receptor agonists. Google Patents; 2014. WO2014066992. Husain M, Aameron-Vendrig A, Ni H. Methods for inhibiting platelet aggregation using glp-1 receptor agonists. Google Patents; 2014. WO2014066992.
Metadaten
Titel
Platelets and platelet adhesion molecules: novel mechanisms of thrombosis and anti-thrombotic therapies
verfasst von
Xiaohong Ruby Xu
Naadiya Carrim
Miguel Antonio Dias Neves
Thomas McKeown
Tyler W. Stratton
Rodrigo Matos Pinto Coelho
Xi Lei
Pingguo Chen
Jianhua Xu
Xiangrong Dai
Benjamin Xiaoyi Li
Heyu Ni
Publikationsdatum
01.10.2016
Verlag
BioMed Central
Erschienen in
Thrombosis Journal / Ausgabe Sonderheft 1/2016
Elektronische ISSN: 1477-9560
DOI
https://doi.org/10.1186/s12959-016-0100-6

Weitere Artikel der Sonderheft 1/2016

Thrombosis Journal 1/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.