In ischemic stroke, treatment options are limited. Therapeutic thrombolysis is restricted to the first few hours after stroke, and the utility of current platelet aggregation inhibitors, including GPIIb/IIIa receptor antagonists, and anticoagulants is counterbalanced by the risk of intracerebral bleeding complications. Numerous attempts to establish neuroprotection in ischemic stroke have been unfruitful. Thus, there is strong demand for novel treatment strategies. Major advances have been made in understanding the molecular functions of platelet receptors such as glycoprotein Ib (GPIb) and GPVI and their downstream signaling pathways that allow interference with their function. Inhibition of these receptors in the mouse stroke model of transient middle cerebral artery occlusion prevented infarctions without increasing the risk of intracerebral bleeding. Similarly, it is now clear that the intrinsic coagulation factor XII (FXII) and FXI play a functional role in thrombus formation and stabilization during stroke: their deficiency or blockade protects from cerebral ischemia without overtly affecting hemostasis. Based on the accumulating evidence that thrombus formation and hemostasis are not inevitably linked, new concepts for prevention and treatment of ischemic stroke may eventually emerge without the hazard of severe bleeding complications. This review discusses recent advances related to antithrombotic strategies in experimental stroke research.

Stroke is the second leading cause of death worldwide.1,2  Approximately 80% of strokes are caused by focal cerebral ischemia due to arterial occlusion, whereas up to 20% are caused by intracerebral hemorrhages.3,4  Extracranial artery stenoses are prone to destabilization and plaque rupture leading to cerebral thromboembolism.5  In approximately one-third of ischemic stroke patients, embolism to the brain originates from the heart, especially in atrial fibrillation.6  Thromboembolic occlusion of major or multiple smaller intracerebral arteries leads to focal impairment of the downstream blood flow, and to secondary thrombus formation within the cerebral microvasculature.

In the center of the ischemic territory, oxygen and glucose deprivation, neuronal depolarization, and Ca2+-mediated excitotoxicity induce necrotic and apoptotic cell death. In the penumbra region surrounding the infarct core, however, tissue is preserved for a certain time span depending on whether blood flow is restored.7  Since numerous agents that proved neuroprotective in experimental stroke failed in subsequent clinical trials,8  the only effective treatment option in acute ischemic stroke remains immediate thrombolysis. In this review, we will focus on the initiating event of stroke development, namely intravascular thrombus formation, and highlight promising novel molecular targets for its prevention and treatment.

Thrombolytic therapy

In acute thromboembolic stroke the principal treatment goal is to rapidly achieve recanalization of occluded intracerebral vessels. In the case of a permanent vessel occlusion, a complete infarct will inevitably develop. At present, early intravenous or intra-arterial thrombolysis are the only established therapeutic options.9,10  Less than 10% of patients are amenable to this treatment due to the limited time window of up to 3 to 6 hours after symptom onset because of the risk of severe intracerebral hemorrhage with later application.11  A trial to extend the therapeutic window up to 9 hours by use of recombinant desmoteplase, a novel plasminogen activator, failed.12  For unknown reasons, thrombolytic treatment leads to the dissolution of the vessel-occluding clots in some cases, but not in others. Moreover, secondary arterial reocclusion may follow a previously successful recanalization.13  Most importantly, patients may develop progressive stroke despite sustained early reperfusion of previously occluded major intracranial arteries, a process referred to as “reperfusion injury.” These observations suggest that reperfusion of occluded major arterial branches is a prerequisite for salvage of tissue, but does not inevitably guarantee prevention of infarct growth and clinical recovery.

Platelet inhibitors

There have been numerous attempts to improve stroke outcome by use of platelet aggregation inhibitors and anticoagulants. The antithrombotic effect of acetylsalicylic acid (ASA) is based on the irreversible inhibition of platelet cyclooxygenases 1 and 2, leading to reduced prostaglandin and thromboxane A2 synthesis. ASA has been evaluated within 48 hours of stroke onset in 2 large trials.14,15  There was a moderate, but statistically significant benefit on stroke outcome. It was assumed, yet not based on solid data, that the primary effect of ASA might be due to prevention of early stroke recurrence rather than limiting the neurologic consequences of the initial stroke per se.16  Although the beneficial role of platelet aggregation inhibitors including ASA, ASA in combination with dipyridamole, and the platelet P2Y12 receptor inhibitor clopidogrel in stroke prevention is well established,17,18  the multifaceted role of platelets in acute stroke development is unclear. This limited understanding extends to the mechanisms by which antiplatelet agents may act in preventing thrombus growth within the brain microvasculature.16,19 

The formation of a thrombus requires functional glycoprotein IIb (GPIIb)/GPIIIa, a heterodimeric receptor of the integrin family expressed at high density (50 000-80 000 copies/cell) on the platelet membrane.20  In resting platelets, GPIIb/IIIa exists in a low-affinity state and does not bind its ligands. During platelet activation, intracellular signals are generated that are integrated at defined checkpoints such as CalDAG-GEFI21  and culminate in the activation of talin-122,23  and kindlin-3.24  These bind to the intracellular tails of the integrin β3-subunit (GPIIIa) and induce a conformational change that involves both subunits of the complex. This “final common pathway” of platelet activation results in the exposure of the binding site(s) for a variety of ligands, most notably fibrinogen, von Willebrand factor (VWF), and fibronectin (inside-out signaling), which allows firm adhesion to the extracellular matrix and aggregation. Current strategies to inhibit GPIIb/IIIa include antibodies (abciximab), cyclic peptides adapted from a snake venom disintegrin (eptifibatide), and nonpeptide analogues of an RGD peptide (tirofiban and lamifiban), all of which directly inhibit ligand binding. Although the utility of intravenous GPIIb/IIIa inhibitors in acute coronary syndromes is well established,25  a recent phase 3 trial applying abciximab in acute ischemic stroke was prematurely stopped due to an increased intracranial hemorrhage rate and mortality, as well as lack of efficacy.26  Several studies also evaluated GPIIb/IIIa inhibitors in conjunction with thrombolytic therapy and described some benefit.27,29  Based on the available data, the use of GPIIb/IIIa inhibitors in acute stroke patients cannot be recommended at present.26,30 

Anticoagulants

Anticoagulation with warfarin targets the synthesis of coagulation factors II, VII, IX, and X and is effective in primary and secondary prophylaxis of thromboembolism to the brain in patients with atrial fibrillation.31  Although other anticoagulants, namely unfractionated heparin, low-molecular-weight heparin, or heparinoids that block FXa activity have frequently been used for acute stroke therapy within 48 hours,32,33  several randomized studies have been negative.15,34,35  A most recent trial did not find a significant advantage of low-molecular-weight heparin over ASA.36  The few studies that have addressed the potential benefit of anticoagulation immediately within the first hours after cerebral ischemia gave inconsistent results regarding clinical outcome and stroke recurrence but mostly found a significant increase in intracranial bleeding.36,,39  Consequently, the recently updated American Heart Association guidelines16 (p1680) state that “urgent anticoagulation with the goal of preventing early recurrent stroke, halting neurologic worsening, or improving outcomes is not recommended.” Because anticoagulation carries a significant risk for intracerebral bleeding in the setting of acute stroke,40,41  it remains a major challenge to develop novel anticoagulants and/or antiplatelet agents with a more favorable safety profile and better efficacy.42 

Animal models for focal ischemic stroke

For the study of ischemic stroke, several animal models have been developed.43  Most frequently, occlusion of major extracranial and intracranial arteries is applied in rodents or higher mammals leading to focal cerebral ischemia. Permanent occlusion of the middle cerebral artery (MCAO) at proximal sites, either by a suture or by an intraluminal thread, causes complete infarctions of the middle cerebral artery brain territory involving neocortex and basal ganglia. The clinical situation with vessel occlusion followed by resolution of clots and reperfusion can be mimicked in the MCAO paradigm by withdrawing of the intraluminal thread (so-called transient MCAO model; Figure 1). It has been firmly established that final infarct size depends on the prior occlusion time: ischemic periods less than 30 minutes will lead to infarctions of the caudate and putamen (basal ganglia) and only partly affect the neocortex because ischemic cortical tissue is salvaged by collateral blood supply and reperfusion.44  If reperfusion is delayed further, however, the size of neocortical infarctions will increase, because the surrounding penumbra is subsequently involved in the definite infarct area (Figure 1). One of the unanswered questions is why sufficient reflow does not guarantee salvage of brain tissue. The observation that reperfusion of major intracerebral arteries did not completely prevent further infarct growth led to the concept of a focal “no-reflow” within the brain microvasculature.45  It was shown that thrombus formation continues with accumulation of platelets and fibrin deposition despite removal of the vessel-occluding thread.46,48  These data indicate that ongoing thrombus formation within the brain during reperfusion is an important pathophysiologic step in stroke development that acts in concert with activation of endothelial cells and adhesion of leukocytes to the vessel wall.47,49 

Figure 1

Transient middle cerebral artery occlusion model (tMCAO) in the mouse. (A,B) Three coronal sections through the brain in individual animals. (A) A cerebral infarct at 24 hours after 30 minutes of tMCAO and (B,C) after 1 hour of tMCAO as revealed on tissue sections stained for 2,3,4-triphenyltetarzoliumchloride (TTC), a mitochondrial marker. Red areas represent vital brain tissue; white areas indicate cerebral infarctions. With short occlusion times of 30 minutes, infarcts are restricted to the basal ganglia (arrow in A), whereas prolonged occlusion leads to infarction of the entire MCA territory (B). (D) Infarct development can also be assessed in vivo by magnetic resonance imaging (MRI). Infarcts appear white on T2-w or diffusion-weighed MRI (D) and correspond closely to the extent of infarction seen on tissue sections (C). TTC scans were taken from an Epson Perfection 3200 Photo flatbed scanner (Seiko Epson, Nagano, Japan) at 600 dpi and processed using Epson Scan software. MRI was performed on a 17.6-Tesla ultrahigh field MR unit (Biospin; Bruker BioSpin, Ettlingen, Germany) using a custom-made dual channel surface coil designed for the examination of mouse heads (A063HACG; Rapid Biomedical, Würzburg, Germany). The image protocol comprised a coronal diffusion–weighted sequence (slice thickness, 0.5 mm). MR images were transferred to an external workstation (Leonardo; Siemens, Berlin, Germany) for data processing.

Figure 1

Transient middle cerebral artery occlusion model (tMCAO) in the mouse. (A,B) Three coronal sections through the brain in individual animals. (A) A cerebral infarct at 24 hours after 30 minutes of tMCAO and (B,C) after 1 hour of tMCAO as revealed on tissue sections stained for 2,3,4-triphenyltetarzoliumchloride (TTC), a mitochondrial marker. Red areas represent vital brain tissue; white areas indicate cerebral infarctions. With short occlusion times of 30 minutes, infarcts are restricted to the basal ganglia (arrow in A), whereas prolonged occlusion leads to infarction of the entire MCA territory (B). (D) Infarct development can also be assessed in vivo by magnetic resonance imaging (MRI). Infarcts appear white on T2-w or diffusion-weighed MRI (D) and correspond closely to the extent of infarction seen on tissue sections (C). TTC scans were taken from an Epson Perfection 3200 Photo flatbed scanner (Seiko Epson, Nagano, Japan) at 600 dpi and processed using Epson Scan software. MRI was performed on a 17.6-Tesla ultrahigh field MR unit (Biospin; Bruker BioSpin, Ettlingen, Germany) using a custom-made dual channel surface coil designed for the examination of mouse heads (A063HACG; Rapid Biomedical, Würzburg, Germany). The image protocol comprised a coronal diffusion–weighted sequence (slice thickness, 0.5 mm). MR images were transferred to an external workstation (Leonardo; Siemens, Berlin, Germany) for data processing.

Close modal

In contrast to focal cerebral ischemia, global ischemia refers to an interruption of the entire brain circulation typically seen during cardiac arrest. If transient, global ischemia causes delayed and selective neuronal death in hypoxia-susceptible brain areas without widespread necrosis. The underlying pathologic mechanisms are quite different between global ischemia and MCAO-induced focal ischemia, and therefore global ischemia is not further considered in this review. Cerebral photothrombosis has often been used as an alternative model to induce focal cerebral lesions, but recent studies have shown that the development of brain lesions after photothrombosis does not require intravascular thrombus formation.50,51  Recently, a promising novel mouse model of thromboembolic stroke was reported based on microinjection of murine thrombin,52  but, as with older similar clot models in mice, no data on the effect of antiplatelet treatment or anticoagulation are available yet. Moreover, embolic models are limited by variable infarct sizes, since it is difficult to anticipate which branch of the middle cerebral artery will finally be occluded by the inserted clot. All experimental studies included in this review were based on the transient MCAO (tMCAO) model in mice, which is the most widely used. Here, brain infarctions are initiated by mechanical, but not thromboembolic, occlusion of a major cerebral artery. Although a single model can cover only some aspects of human stroke, which is a complex and heterogeneous disease, the tMCAO model turned out to be useful in elucidating basic pathomechanisms of thrombus formation in the downstream microvasculature.

The role of platelets in experimental stroke

Pathologic platelet activity and platelet receptor-ligand interactions have been linked to cerebral ischemic events.48  By use of 111In-labeled platelets in a primate model of tMCAO, the del Zoppo group showed that platelets are deposited in the ischemic basal ganglia early during reperfusion, and electron microscopic examination demonstrated aggregates of degranulated platelets together with fibrin and leukocytes (Okada et al53 ). Accordingly, baboons treated with ticlopidine and heparin displayed a significant reduction in platelet deposition and microvascular occlusions in the ischemic basal ganglia.48  The advent of genetic methods that allow targeted manipulations in the mouse genome has paved the way for novel concepts of thrombus formation in mice54,55  that may help to identify important steps in the pathogenesis of human atherothrombosis and ischemic stroke. In the following sections, we will summarize the recent experimental evidence in support of a pathophysiologic role of platelet receptors GPIIb/IIIa, GPIb, and GPVI (Figure 2) and the involvement of the intrinsic coagulation cascade in focal cerebral ischemia. Novel drugs targeting these molecules may help to overcome the current limitations and hazards of conventional anticoagulation and platelet inhibition in acute ischemic stroke.56,57 

Figure 2

Model of platelet–vessel wall interaction. (A) The initial contact (tethering) of platelets to the extracellular matrix (ECM) is mediated predominantly by GPIbα-VWF interactions. The GPIbα-VWF interaction is essential at high shear rates (> 500 s−1). GPIbα may also interact with P-selectin exposed on activated endothelial cells and thereby contribute to platelet recruitment to the intact vessel wall. (B) At sites of vascular injury, GPVI-collagen interactions initiate cellular activation followed by shifting of integrins to high-affinity state and the release of secondarily acting agonists, most importantly ADP, ATP, and TxA2. GPIb-mediated signaling may amplify GPVI-induced activation pathways. In parallel, exposed tissue factor (TF) locally triggers the formation of thrombin (extrinsic pathway), which in addition to GPVI mediates cellular activation. On the growing thrombus, activation of FXII and FXI also leads to thrombin formation. (C) Activated GPIIb/IIIa (integrin αIIbβ3) together with β1 integrins (not shown) mediates firm adhesion by binding to VWF, fibronectin, and other ligands. Released ADP, ATP, and TxA2 amplify integrin activation on adherent platelets and mediate thrombus growth by activating additional platelets and fibrinogen binding to GPIIb/IIIa. (D) Adherent platelets may recruit leukocytes to the thrombus through GPIbα-MAC1 interactions. This scheme does not exclude the involvement of other receptor-ligand interactions.

Figure 2

Model of platelet–vessel wall interaction. (A) The initial contact (tethering) of platelets to the extracellular matrix (ECM) is mediated predominantly by GPIbα-VWF interactions. The GPIbα-VWF interaction is essential at high shear rates (> 500 s−1). GPIbα may also interact with P-selectin exposed on activated endothelial cells and thereby contribute to platelet recruitment to the intact vessel wall. (B) At sites of vascular injury, GPVI-collagen interactions initiate cellular activation followed by shifting of integrins to high-affinity state and the release of secondarily acting agonists, most importantly ADP, ATP, and TxA2. GPIb-mediated signaling may amplify GPVI-induced activation pathways. In parallel, exposed tissue factor (TF) locally triggers the formation of thrombin (extrinsic pathway), which in addition to GPVI mediates cellular activation. On the growing thrombus, activation of FXII and FXI also leads to thrombin formation. (C) Activated GPIIb/IIIa (integrin αIIbβ3) together with β1 integrins (not shown) mediates firm adhesion by binding to VWF, fibronectin, and other ligands. Released ADP, ATP, and TxA2 amplify integrin activation on adherent platelets and mediate thrombus growth by activating additional platelets and fibrinogen binding to GPIIb/IIIa. (D) Adherent platelets may recruit leukocytes to the thrombus through GPIbα-MAC1 interactions. This scheme does not exclude the involvement of other receptor-ligand interactions.

Close modal

Glycoprotein IIb/IIIa (integrin αIIbβ3).

In a seminal paper, Choudhri et al46  used pharmacologic blockade of GPIIb/IIIa in the tMCAO model as proof of concept that even if large arteries are recanalized after thromboembolic stroke, microvascular thrombosis continues to occur at distal sites. When the GPIIb/IIIa antagonist GPI 562 was administered to mice immediately before or after 1 hour of MCAO, platelet and fibrin accumulation as well as cerebral infarct volumes were reduced. This effect was dose-dependent, with an associated significant increase in the rate of intracerebral hemorrhages. Treatment of mice with an anti-GPIIb/IIIa antibody similarly led to reduced endothelial adhesion of leukocytes and platelets during reperfusion after tMCAO.58  In baboons, a 30% inhibition of platelet aggregation by the GPIIb/IIIa antagonist TP9201 was sufficient to regain complete microvascular patency after 3 hours of tMCAO, whereas near complete inhibition of platelet aggregation again led to large intracerebral hemorrhages.59  In a recent study,57  we reassessed efficacy and safety of anti-GPIIb/IIIa treatment in ischemic stroke using Fab2 fragments of the mouse GPIIb/IIIa-blocking mAb, JON/A, which completely inhibits ex vivo platelet aggregation and induces prolonged tail bleeding times.60  Most animals that had received 100 μg anti-GPIIb/IIIa Fab2, leading to a virtually complete receptor blockade, died due to intracerebral hemorrhage, and the few surviving animals exhibited infarct volumes of the same extent as seen in controls (Figure 3). A 78% and 68% receptor blockade improved survival rates, but failed to influence infarct volumes or neurologic outcome. By contrast, in GPIIb-deficient mice, cerebral infarct size was reduced at 24 hours after tMCAO, but no information on bleeding complications is available from this study.61  Excessive GPIIb/IIIa blockade appears to be inevitably associated with major bleeding complications in mice and reflects similar findings in stroke patients.26  Thus, rather than blocking this final common pathway of platelet activation, their aggregation, targeting platelet adhesion, and/or early signaling events may provide a promising therapeutic alternative.

Figure 3

The influence of platelet glycoprotein receptor blockade on stroke outcome. (A) Coronal 2,3,4-triphenyltetarzoliumchloride–stained sections at 24 hours after 1 hour of tMCAO in a sham-treated mouse. Note the large infarction of the entire middle cerebral artery territory, and the corresponding T2-w magnetic resonance image of the infarct in the bottom panel. (B) Blockade with anti-GPIb Fab significantly reduced infarct size. The arrow points to the small infarct within the basal ganglia, whereas the cerebral cortex is protected. The corresponding magnetic resonance image correctly depicts decreased infarct size (arrow in bottom panel). (C) Surprisingly, blockade of GPIIb/IIIa had no influence on the infarct size in surviving animals, and was associated with lethal intracerebral hemorrhage (ICH) in many animals (not shown). Importantly, no areas with signal loss indicating bleeding complications were seen in mice after GPIb-Fab blockade (bottom panel in B; compare with ICH in Figure 2C). TTC scans were taken from an Epson Perfection 3200 Photo flatbed scanner (Seiko Epson) at 600 dpi and processed using Epson Scan software (Seiko Epson). MRI was performed on a 1.5-Tesla MR device (Vision; Siemens) using a custom-made dual channel surface coil designed for the examination of mouse heads (A063HACG; Rapid Biomedical). The image protocol comprised a coronal 3D T2-weighted gradient echo–constructed interference in steady state sequence (slice thickness, 1 mm). MR images were transferred to an external workstation (Leonardo; Siemens) for data processing.

Figure 3

The influence of platelet glycoprotein receptor blockade on stroke outcome. (A) Coronal 2,3,4-triphenyltetarzoliumchloride–stained sections at 24 hours after 1 hour of tMCAO in a sham-treated mouse. Note the large infarction of the entire middle cerebral artery territory, and the corresponding T2-w magnetic resonance image of the infarct in the bottom panel. (B) Blockade with anti-GPIb Fab significantly reduced infarct size. The arrow points to the small infarct within the basal ganglia, whereas the cerebral cortex is protected. The corresponding magnetic resonance image correctly depicts decreased infarct size (arrow in bottom panel). (C) Surprisingly, blockade of GPIIb/IIIa had no influence on the infarct size in surviving animals, and was associated with lethal intracerebral hemorrhage (ICH) in many animals (not shown). Importantly, no areas with signal loss indicating bleeding complications were seen in mice after GPIb-Fab blockade (bottom panel in B; compare with ICH in Figure 2C). TTC scans were taken from an Epson Perfection 3200 Photo flatbed scanner (Seiko Epson) at 600 dpi and processed using Epson Scan software (Seiko Epson). MRI was performed on a 1.5-Tesla MR device (Vision; Siemens) using a custom-made dual channel surface coil designed for the examination of mouse heads (A063HACG; Rapid Biomedical). The image protocol comprised a coronal 3D T2-weighted gradient echo–constructed interference in steady state sequence (slice thickness, 1 mm). MR images were transferred to an external workstation (Leonardo; Siemens) for data processing.

Close modal

Glycoprotein Ib-V-IX.

The initial tethering of platelets at sites of vascular injury is mediated by GPIb-V-IX, a structurally unique receptor complex exclusively expressed in platelets and megakaryocytes (Figure 2). In humans, lack or dysfunction of this receptor has been associated with the Bernard-Soulier syndrome, a congenital bleeding disorder characterized by mild thrombocytopenia, giant platelets, platelet inability to adhere to subendothelial matrices, and a dramatically prolonged bleeding time.62,63  This phenotype has been reproduced in mice lacking functional GPIb-V-IX.54,64  The binding of GPIbα to the A1 domain of VWF is the principal interaction capable of and necessary for tethering platelets to the vessel wall at high shear flow conditions (> ∼500 s−1), whereas this interaction may not be relevant at lower shear rates.65  Although sufficient to support platelet binding, this adhesive interaction is characterized by a rapid dissociation rate and thus cannot mediate irreversible adhesion by itself. Rather, the interaction keeps platelets in close contact with the matrix, while the cells continuously translocate in the direction of blood flow. The specific requirement for GPIbα for platelet adhesion under conditions of high shear, such as found in diseased arteries, makes this receptor a potentially attractive target for pharmacologic inhibition of pathologic thrombus formation. Inhibition of the VWF-binding site on GPIbα with Fab fragments of the antibody p0p/B in wild-type mice abrogated platelet tethering and adhesion in a model of mechanically induced arterial thrombosis.66  Such mice have prolonged tail bleeding times but do not show signs of spontaneous hemorrhage.57,66  The central role of GPIbα in arterial thrombus formation was later confirmed and extended in a study showing that transgenic mice expressing GPIbα in which the extracellular domain was replaced by that of the human interleukin-4 receptor (GPIb-TG) are completely unable to produce intravascular thrombi.67 

A crucial role of GPIb in stroke development has recently been elucidated in experimental focal cerebral ischemia.57  Complete blockade of the VWF-binding site of GPIbα by intravenous injection of 100 μg Fab fragments of p0p/B into mice before tMCAO led to a reduction of stroke volumes of approximately 60%. Importantly, delayed application of anti-GPIb Fab 1 hour after MCAO was likewise effective (Figure 3). Although tail bleeding times were strongly elevated in anti-GPIb Fab–treated mice, no increase in intracerebral hemorrhages was detected. Together, this indicated that GPIbα is critically involved in the pathogenesis of ischemic stroke, but not required to prevent bleeding at sites of ischemia/reperfusion damage in the brain, and supported the previous notion that there is no clear correlation between bleeding time and bleeding risk.68  This surprising result was confirmed shortly afterward by Goerge et al,69  who found that local inflammation in the brain (induced by tMCAO) and other tissues triggers bleeding in the absence of platelets. In the presence of platelets, bleeding was prevented, and this protective effect was unexpectedly also seen in mice lacking functional GPIbα. Thus, it appears that the mechanisms by which platelets contribute to the pathogenesis of ischemic brain injury are different from those required to maintain vascular integrity after ischemia/reperfusion in this organ. These observations not only demonstrate that GPIb is a central player in murine experimental stroke but also raise the intriguing possibility that strong platelet inhibition can be achieved without significantly increasing the risk of (spontaneous) intracerebral bleeding.

Besides its principal ligand, VWF, GPIbα also binds thrombin, high-molecular-weight kininogen, factor XII, Mac-1 (a β2 integrin expressed in neutrophils and monocytes [CD11b/CD18]), and P-selectin.70  The corresponding binding sites are located in the N-terminal region of the receptor, but their precise local arrangement is unknown. It is not entirely clear at present which of the GPIbα interactions are critical for thrombus formation in stroke. However, allelic variants of platelet GPIbα causing enhanced VWF/GPIb interactions are associated with an increased risk of ischemic stroke,71  and increased serum levels of VWF have been recognized as an independent stroke risk factor.72  In support of this, we found in a most recent investigation that VWF-deficient mice are protected against cerebral ischemia, although to a lesser extent than mice treated with anti-GPIbα antibodies (C.K., H. Deckmyn, B.N., and G.S., unpublished observation, June 2008). This indicates that different ligands of GPIb may be involved in the development of infarcts in this model.

Previous studies have shown that mice deficient in Mac-1 are less susceptible to cerebral ischemia/reperfusion injury.73  This protection was associated with reduced neutrophil infiltration after tMCAO, but the exact contribution of Mac-1 to the pathology is unclear. Therefore, it is tempting to speculate that Mac-1–GPIb interactions could mediate platelet-leukocyte adhesion, promoting inflammation at sites of thrombosis after cerebral ischemia. Moreover, GPIbα binds to P-selectin.74  During cerebral ischemia, increased surface P-selectin expression was noted on endothelial cells and platelets as early as 1 hour after reperfusion, and inhibition of P-selectin improved stroke outcome, indicating that this interaction is also functionally relevant.75  Taken together, GPIbα plays a central role as a receptor mediating complex platelet-platelet, platelet-endothelium, and platelet-leukocyte interactions, all of which may be critical in secondary infarct growth after tMCAO in rodents. Fab fragments of the monoclonal antibody 6B4, raised against human GPIbα, exhibited a powerful antithrombotic effect in baboons by blocking the GPIbα-binding site for VWF without significant prolongation of the skin bleeding time.76  This antibody was recently humanized by variable-domain resurfacing guided by computer modeling77  and may provide an important tool to study the role of GPIbα in human thrombotic diseases, including stroke.

Glycoprotein VI.

Although GPIb-VWF interactions can elicit intracellular signals,78  these are generally considered very weak compared with other stimuli, most notably subendothelial collagens, which are exposed to the cells at sites of endothelial damage. Among the numerous collagen receptors expressed in platelets, GPVI is of central importance for cellular activation and subsequent firm arrest.79  GPVI, a 62-kDa type I transmembrane receptor of the Ig superfamily,80  is exclusively expressed in platelets and megakaryocytes.81  It noncovalently associates with the Fc receptor (FcR) γ-chain, and the complex signals through tyrosine phosphorylation cascades leading to calcium mobilization, degranulation, activation of GPIIb/IIIa, and aggregation.79  Platelets in which GPVI has been depleted by in vivo administration of antibodies against the receptor do not respond to collagen.79,81  Several reports have demonstrated a profound antithrombotic effect of such GPVI inhibition after arterial wall injury and collagen-induced thromboembolism,81,83  which is associated with a very moderate increase in tail bleeding.81  In tMCAO, treatment of mice with the anti-GPVI antibody JAQ1 significantly reduced the brain infarct volumes at day 1 after tMACO57  but did not increase the incidence of intracerebral hemorrhages. This indicates that platelet/collagen interactions via GPVI may also be involved in stroke development in this model. However, GPVI depletion was less effective than GPIb blockade and did not affect clinical outcome variables, suggesting that other platelet agonists contribute to thrombus formation in the tMCAO model. Among these, thrombin is the most powerful initiator of platelet adhesion and thrombus formation sufficient to drive these processes independently of collagen under certain experimental conditions.84 

The observation that inhibitors of GPIb or GPVI function provide significant protection from ischemic brain injury suggests that signaling pathways downstream of these receptors could be promising therapeutic targets. This has recently been confirmed by the analysis of mice lacking stromal interaction molecule 1 (STIM1), a key regulator of agonist-induced Ca2+ entry in immune cells and platelets.85,86  Platelets derived from these mice display selective defects in cellular activation downstream of GPVI and shear-resistant adhesion and thrombus formation in vitro and in vivo.87  In contrast, activation in response to G protein–coupled agonists, such as thrombin, is largely preserved, which indicates that STIM1-mediated signaling events are particularly important for the GPIb-GPVI-ITAM pathway in platelets. Mice with STIM1-deficient platelets showed profound reduction in secondary infarct growth after tMCAO, but no increase in intracerebral hemorrhages.87 

The intrinsic coagulation pathway as a novel target for stroke prevention

The role of coagulation factors XI (FXI) and XII in thrombus stability and hemostasis.

Hemostasis and pathologic thrombus formation occluding coronary or cerebral arteries in myocardial infarction and stroke, respectively, have long been considered to share identical molecular pathways. According to this concept, treatment of thrombosis would be possible only at the expense of impaired hemostasis. However, this dogma has recently been challenged.55,56,88  Two distinct pathways for initiating plasmatic coagulation exist, triggered either by vessel wall (extrinsic) or blood-borne (intrinsic) factors and converge on a common pathway leading to thrombin and fibrin formation. The extrinsic pathway is initiated by exposure of subendothelial tissue factor upon vessel injury (Figure 2). Tissue factor activates the plasma protease factor VIIa. The intrinsic pathway of coagulation is initiated when coagulation factor XII (FXII, Hageman factor) comes into contact with negatively charged surfaces (contact activation). Because individuals with hereditary FXII deficiency do not show an abnormal bleeding phenotype, FXII has been considered dispensable for proper hemostasis.89  Recent investigations, however, revealed a role of FXII in pathologic thrombus formation and stability.88  FXII-deficient mice, similar to FXII-deficient patients, exhibit a prolonged activated partial thromboplastin time but no bleeding tendency. Importantly, FXII-deficient mice showed impaired formation and stabilization of thrombi in different models of arterial thrombosis.88  These unexpected findings indicate that it is possible to target thrombus formation without ensuing bleeding complications. The molecular mechanisms that activate FXII during pathologic thrombus formation in vivo await elucidation. One possible mechanism has recently been proposed by Kannemeier et al, who demonstrated that extracellular RNA, but not DNA, augments (auto)activation of FXII and FXI and thereby acts as a potent trigger of coagulation in vivo.90  It is at present not clear whether this pathway is the predominant mechanism of FXII activation or whether other, presumably polyanionic, molecules also contribute to this process.

The significance of the intrinsic coagulation system for stroke development.

FXII-deficient mice are also protected from cerebral ischemia.56  After tMCAO, infarct volumes were 50% less in FXII-deficient compared with wild-type mice at 24 hours, and FXII mutants developed significantly less neurologic impairment. Follow-up magnetic resonance imaging (MRI) on days 3 and 7 after tMCAO revealed that this protective effect was sustained over time. Infarcts in FXII-deficient mice were restricted to the basal ganglia, and fibrin deposition in the microvasculature of cortical vessels was markedly reduced. Administration of the FXIIa inhibitor D-Pro-Phe-Arg-chloromethyl ketone (PCK) to wild-type mice similarly conferred protection from stroke. Because FXI-deficient mice were similarly protected from experimental stroke as FXII-deficient mice, the intrinsic coagulation pathway appears to be critically involved in infarct development.56  A recent epidemiologic study disclosed protection against cerebrovascular events in Jewish patients with severe congenital FXI deficiency, indicating that our experimental results in mice may be relevant for the human situation.91  Of note, FXII knockout mice and wild-type mice treated with FXII inhibitors had a prolonged activated partial thromboplastin time, but did not display excessive bleeding during surgery. Moreover, serial MRI using blood-sensitive sequences did not show an increased frequency of intracerebral hemorrhages. Importantly, pharmacologic blockade of factor IX downstream of factors XII and XI was similarly protective after tMCAO.92  However, factor IX deficiency reflects human hemophilia type B, which is associated with a significant bleeding phenotype.93  This can be explained by the fact that factor IX is also activated by the factor VIIa–tissue factor complex and thereby participates in the extrinsic pathway of coagulation.94  Intracerebral hemorrhage is the most feared complication of coumarins, the conventional anticoagulant today. It appears that FXII inhibition could be a novel target for safer anticoagulation and stroke prevention, since FXII is an essential component of pathologic thrombus formation but not of physiologic hemostasis.95 

Basic molecular biology has uncovered the functions of several platelet receptors such as GPIb and GPVI and downstream signaling pathways in thrombus formation. The fact that inhibition of these platelet receptors/signaling pathways is not associated with increased intracerebral bleeding after tMCAO may open up new avenues for stroke treatment in the future. Novel therapeutic approaches are eagerly awaited in view of the recent negative experience with GPIIb/IIIa receptor inhibition26  and the limited access of stroke patients to thrombolytic treatment. Therefore, it is very promising that antibodies against human GPIb effectively prevented thrombus formation after peripheral vessel injury in baboons76  and that humanized Fab′ fragments against GPIbα have been generated by molecular engineering.77  Before these can be clinically tested in stroke patients, they await proof of safety and efficacy in a translational primate model. This is important to rule out species-specific effects restricted to rodents, as seen in many other potential therapeutics in the past.8,96 

Besides the prevention and treatment of cerebral ischemia resulting from spontaneous thromboembolism, iatrogenic stroke might become another area of application for the novel antithrombotics described. Frequently performed routine procedures such as angiography, angioplasty, or heart surgery are accompanied by a significant incidence of (clinically silent) cerebral thromboemboli, which above a certain threshold can cause severe cognitive dysfunction or even large brain infarction.97  Now, the role of the intrinsic pathway of coagulation in pathologic thrombus formation has been unraveled56,88  and may provide novel options for anticoagulation during these potentially proembolic interventions.95  In balancing risk and benefit of any new stroke prophylaxis or treatment, the reduction of the bleeding risk is as essential as preventing thrombus formation and improving reperfusion. Highly selective FXII inhibitors are currently being developed for the application in humans.98  They may help to control and limit thromboembolic complications with a better safety profile than coumarins or heparins in which the therapeutic benefit has often been neutralized by excess bleeding complications.

We thank Profs Wolfgang Müllges, Klaus Toyka, and Ulrich Walter (University of Würzburg) for valuable comments.

Our own work cited in this review was supported by the Deutsche Forschungsgemeinschaft (Bonn, Germany; SFB 688, A1, A3, B1), the Rudolf Virchow Center, and institutional funds of the state of Bavaria, Germany.

Contribution: G.S., C.K., and B.N. wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Guido Stoll, Department of Neurology, University of Würzburg, Josef-Schneider-Str 11, D-97080 Würzburg, Germany; e-mail: stoll_g@klinik.uni-wuerzburg.de; or Bernhard Nieswandt, Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine, Zinklesweg 10; 97080 Würzburg, Germany; e-mail: bernhard.nieswandt@virchow.uni-wuerzburg.de.

1
Lopez
AD
Mathers
CD
Ezzati
M
Jamison
DT
Murray
CJL
Global and regional burden of disease and risk factors, 2001: systematic analysis of population health data.
Lancet
2006
367
1747
1757
2
Carro
JJ
Huybrechts
KF
Duchesne
I
Management patterns and costs of acute ischemic stroke: an international study.
Stroke
2000
31
582
590
3
Feigin
VL
Lawes
CM
Bennet
DA
Anderson
CS
Stroke epidemiology: a review of population-based studies of incidence, prevalence, and case-fatality in the late 20th century.
Lancet Neurol
2003
2
43
53
4
Prospective Studies Collaboration
Lewington
S
Whitlock
G
et al
Blood cholesterol and vascular mortality by age, sex, and blood pressure: a meta-analysis of individual data from 61 prospective studies with 55,000 vascular deaths.
Lancet
2007
370
1829
1839
5
Stoll
G
Bendszus
M
Inflammation and atherosclerosis: novel insights into plaque formation and destabilization.
Stroke
2006
37
1923
1932
6
on behalf of the guideline development group for the NICE national clinical guideline for management of atrial fibrillation in primary and secondary care
Hughes
M
Lip
GYH
Stroke and thrombembolism in atrial fibrillation: a systemic review of stroke risk factors, risk stratification schema and cost effectiveness data.
Thromb Haemost
2008
99
295
304
7
Dirnagl
U
Iadecola
C
Moskowitz
MA
Pathobiology of ischaemic stroke:an integrated view.
Trends Neurosci
1999
22
391
397
8
O'Collins
VE
Macleod
MR
Donnan
GA
Horky
LL
van der Worp
BH
Howells
DW
1,026 experimental treatments in acute stroke.
Ann Neurol
2006
59
467
477
9
The National Institute of Neurological Disorders and Stroke rt-PA Stroke Study Group
Tissue plasminogen activator for acute ischemic stroke.
N Engl J Med
1995
333
1581
1588
10
Choi
JH
Bateman
BT
Mangla
S
et al
Endovascular recanalization therapy in acute ischemic stroke.
Stroke
2006
37
419
424
11
Adams
H
Adams
R
del Zoppo
G
Goldstein
LB
Guidelines for the early management of patients with ischemic stroke.
Stroke
2005
36
916
921
12
Desmoteplase in Acute Ischemic Stroke-2.
Accessed March 3, 2008
13
Heo
JH
Lee
KY
Kim
SH
Kim
DI
Immediate reocclusion following a successful thrombolysis in acute stroke: a pilot study.
Neurology
2003
60
1684
1687
14
CAST (Chinese Acute Stroke Trial) Collaborative group
CAST: randomised placebo-controlled trial of early aspirin use in 20,000 patients with acute ischemic stroke.
Lancet
1997
349
1641
1649
15
International Stroke Trial Collaborative Group
The International Stroke Trial (IST): a randomised trial of aspirin, subcutaneous heparin, both, or neither among 19435 patients with acute ischemic stroke.
Lancet
1997
349
1569
1581
16
Adams
HP
del Zoppo
G
Alberts
MJ
et al
Guidelines for the early management of adults with ischemic stroke.
Stroke
2007
38
1655
1711
17
CAPRIE Steering Committee
A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk for ischemic events (CAPRIE).
Lancet
1996
348
1329
1339
18
Verro
P
Gorelick
PB
Nguyen
D
Aspirin plus dipyridamole versus aspirin for prevention of vascular events after stroke or TIA: a meta-analysis.
Stroke
2008
39
1358
1363
19
Sandercock
P
Gubitz
G
Foley
P
Counsell
C
Antiplatelet therapy for acute ischaemic stroke.
Cochrane Database Syst Rev
2008
2
1
72
20
Shattil
SJ
Ginsberg
MH
Integrin signalling in vascular biology.
J Clin Invest
1997
100
1
5
21
Crittenden
JR
Bergmeier
W
Zhang
Y
et al
CalDAG-GEFI integrates signalling for platelet aggregation and thrombus formation.
Nat Med
2004
10
982
986
22
Nieswandt
B
Moser
M
Pleines
I
et al
Loss of talin in platelets abrogates integrin activation, platelet aggregation, and thrombus formation in vitro and in vivo.
J Exp Med
2007
204
3113
3118
23
Petrich
BG
Marchese
P
Ruggeri
ZM
et al
Talin is required for integrin-mediated platelet function in hemostasis and thrombosis.
J Exp Med
2007
204
3103
3111
24
Moser
M
Nieswandt
B
Ussar
S
Pozgajova
M
Fässler
R
Kindlin-3 is essential for integrin activation and platelet aggregation.
Nat Med
2008
14
325
330
25
Bhatt
DL
Topol
EJ
Current role of platelet glycoprotein IIb/IIIa inhibitors in acute coronary syndromes.
JAMA
2000
284
1549
1558
26
Adams
HP
Effron
MB
Torner
J
et al
Emergency administration of abciximab for treatment of patients with acute ischemic stroke: results of an international phase III trial.
Stroke
2008
39
87
99
27
Seitz
RJ
Meisel
S
Moll
M
Wittsack
HJ
Junghans
U
Siebler
M
The effect of combined thrombolysis with rtPA and tirofiban on ischemic brain lesions.
Neurology
2004
62
2110
2112
28
Eckert
B
Koch
C
Thomalla
G
et al
Aggressive therapy with intravenous abciximab and intra-arterial rtPA and additional PTA/stenting improves clinical outcome in acute vertebrobasilar occlusion: combined local fibrinolysis and intravenous abciximab in acute vertebrobasilar stroke treatment (FAST): results of a multicenter study.
Stroke
2005
36
1160
1165
29
Abou-Chebl
A
Bajzer
CT
Kreiger
DW
Furlan
AJ
Yaday
JS
Multimodal therapy for the treatment of severe ischemic stroke combining GPIIb/IIIa antagonists and angioplasty after failure of thrombolysis.
Stroke
2005
36
2286
2288
30
Ciccone
A
Abraha
I
Santilli
I
Glycoprotein IIb-IIIa inhibitors for acute ischemic stroke.
Stroke
2007
38
1113
1114
31
Andersen
KK
Olsen
TS
Reduced poststroke mortality in patients with stroke and atrial fibrillation treated with anticoagulants: results from a dutch quality-control registry of 22179 patients with ischemic stroke.
Stroke
2007
38
259
263
32
Adams
HP
Emergent use of anticoagulation for treatment of patients with ischemic stroke.
Stroke
2002
33
856
861
33
Al-Sadat
A
Sunbulli
M
Chaturvedi
S
Use of intravenous heparin by North American neurologists: do the data matter?
Stroke
2002
33
1574
1577
34
Publications Committee for the Trial of ORG 10172 in Acute Stroke Treatment Investigators (TOAST)
Low molecular weight heparinoid, ORG 10172 (danaparoid), and outcome after acute ischemic stroke: a randomized controlled trial.
JAMA
1998
279
1265
1272
35
Bath
PM
Lindenstrom
E
Boysen
G
et al
Tinzaparin in acute ischemic stroke (TAIST): a randomized aspirin-controlled trial.
Lancet
2001
358
702
710
36
Wong
KS
Chen
C
Ng
PW
et al
Low-molecular-weight heparin compared with aspirin for the treatment of acute ischemic stroke in Asian patients with large occlusive disease: a randomised study.
Lancet Neurol
2007
6
407
413
37
Camerlingo
M
Salvi
P
Belloni
G
et al
Intravenous heparin started within the first 3 hours after onset of symptoms as a treatment for acute nonlacunar hemispheric cerebral infarctions.
Stroke
2005
36
2415
2420
38
Chamorro
A
Busse
O
Obach
V
et al
The rapid anticoagulation prevents ischemic damage study in acute stroke: final results from the writing commitee.
Cerebrovasc Dis
2005
19
402
440
39
Paciaroni
M
Agnelli
G
Micheli
S
Caso
V
Efficacy and safety of anticoagulant treatment in acute cardioembolic stroke: a meta-analysis of randomized controlled trials.
Stroke
2007
38
423
430
40
Benatar
M
Heparin use in acute ischaemic stroke: does evidence change practice?
Q J Med
2005
98
147
152
41
Gubitz
G
Sandercock
P
Counsell
C
Anticoagulants for acute ischaemic stroke.
Cochrane Database Syst Rev
2004
3
1
49
42
Hart
RG
Tonarellin
SB
Pearce
LA
Avoiding central nervous system bleeding during antithrombotic therapy.
Stroke
2005
36
1588
1593
43
Durukan
A
Strbian
D
Tatlisumak
T
Rodent models of ischemic stroke: a useful tool for stroke drug development.
Curr Pharmaceutical Design
2008
14
359
370
44
Memezawa
H
Smith
ML
Siesjö
BK
Penumbral tissues salvaged by reperfusion following middle cerebral artery occlusion in rats.
Stroke
1992
23
552
559
45
Connolly
ES
Winfree
CJ
Springer
TA
et al
Cerebral protection in homozygous null ICAM-1 mice after middle cerebral artery occlusion: role of neutrophil adhesion in the pathogenesis of stroke.
J Clin Invest
1996
97
209
216
46
Choudhri
TF
Hoh
BL
Zerwes
HG
et al
Reduced microvascular thrombosis and improved outcome in acute murine stroke by inhibiting GP IIb/IIIa receptor-mediated platelet aggregation.
J Clin Invest
1998
102
1301
1310
47
Zhang
ZG
Zhang
L
Tsang
W
et al
Dynamic platelet accumulation at the site of the occluded middle cerebral artery and in downstream microvessels is associated with loss of microvascular integrity after embolic middle cerebral artery occlusion.
Brain Res
2001
912
181
194
48
Del Zoppo
GJ
The role of platelets in ischemic stroke.
Neurology
1998
51
suppl 3
S9
S14
49
Del Zoppo
GJ
Mabuchi
T
Cerebral microvessel responses to focal ischemia.
J Cereb Blood Flow Metab
2003
23
879
894
50
Frederix
K
Chauhan
AK
Kisucka
J
et al
Platelet adhesion receptors do not modulate infarct volume after photochemically induced stroke in mice.
Brain Res
2007
1185
239
245
51
Kleinschnitz
C
Braeuninger
S
Pham
M
et al
Blocking of platelets or intrinsic coagulation pathway-driven thrombosis does not prevent cerebral infarctions induced by photothrombosis.
Stroke
2008
39
1262
1268
52
Orset
C
Macrez
R
Young
AR
et al
Mouse model of in situ thromboembolic stroke and reperfusion.
Stroke
2007
38
2771
2778
53
Okada
Y
Copeland
BR
Fitridge
R
Koziol
JA
del Zoppo
GL
Fibrin contributes to microvascular obstructions and parenchymal changes during early focal cerebral ischemia and reperfusion.
Stroke
1994
25
1847
1853
54
Sachs
UJH
Nieswandt
B
In vivo thrombus formation in murine models.
Circ Res
2007
100
979
991
55
Gailani
D
Renné
T
The intrinsic pathway of coagulation: a target for treating thromboembolic disease?
J Thromb Hemost
2007
5
733
741
56
Kleinschnitz
C
Stoll
G
Bendszus
M
et al
Targeting coagulation factor XII provides protection from pathological thrombosis in cerebral ischemia without interfering with hemostasis.
J Exp Med
2006
203
513
518
57
Kleinschnitz
C
Pozgajova
M
Pham
M
Bendszus
M
Nieswandt
B
Stoll
G
Targeting platlets in acute experimental stroke: impact of glycoprotein Ib, VI, IIb/IIa blockade on infarct size, functional outcome, and intracranial bleeding.
Circulation
2007
115
2323
2330
58
Ishikawa
M
Cooper
D
Arumugam
TV
Zhang
JH
Nanda
A
Granger
DN
Platelet-leukocyte-endothelial cell interactions after middle cerebral artery occlusion and reperfusion.
J Cereb Blood Flow Metab
2004
24
907
914
59
Abumiya
T
Fitridge
R
Mazur
C
et al
Integrin alphaIIbbeta3 inhibitor preserves microvascular patency in experimental acute focal cerebral ischemia.
Stroke
2000
31
1402
1410
60
Bergmeier
W
Schulte
V
Brockhoff
G
Bier
U
Zirngibl
H
Nieswandt
B
Flow cytometric detection of activated mouse integrin alphaIIbbeta3 with a novel monoclonal antibody.
Cytometry
2002
48
80
86
61
Massberg
S
Schürzinger
K
Lorenz
M
et al
Platelet adhesion via glycoprotein IIb integrin is critical for atheroprogression and focal cerebral ischemia: an in vivo study in mice lacking glycoprotein IIb.
Circulation
2005
112
1180
1188
62
López
JA
Andrews
RK
Afshar-Kharghan
V
Berndt
MC
Bernard-Soulier syndrome.
Blood
1998
92
2366
2373
63
Andrews
RK
Berndt
MC
Platelet physiology and thrombosis.
Thromb Res
2004
114
447
453
64
Ware
J
Russell
S
Ruggeri
ZM
Generation and rescue of a murine model of platelet dysfunction: the Bernhard-Soulier syndrome.
Proc Natl Acad Sci U S A
2000
97
2803
2808
65
Savage
B
Saldivar
E
Ruggeri
ZM
Initiation of platelet adhesion by arrest onto fibrinogen or translocation on von Willebrand factor.
Cell
1996
84
289
297
66
Massberg
S
Gawaz
M
Grüner
S
et al
A crucial role of glycoprotein VI for platelet recruitment to the injured arterial wall in vivo.
J Exp Med
2003
197
41
49
67
Bergmeier
W
Piffath
CL
Goerge
T
et al
The role of platelet adhesion receptor GPIbalpha far exceeds that of its main ligand, von Willebrand factor, in arterial thrombosis.
Proc Natl Acad Sci U S A
2006
103
16900
16905
68
Rodgers
RP
Levin
J
A critical reappraisal of the bleeding time.
Semin Thromb Hemost
1990
16
1
20
69
Goerge
T
Ho-Tin-Noe
B
Carbo
C
et al
Inflammation induces hemorrhage in thrombocytopenia.
Blood
2008
111
4958
4964
70
Berndt
MC
Shen
Y
Dopheide
SM
Gardiner
EE
Andrews
RK
The vascular biology of the glycoprotein Ib-IX-V complex.
Thromb Hemost
2001
86
178
188
71
Maguire
JM
Thakkinstian
A
Sturm
J
et al
Polymorphisms in platelet glycoprotein 1balpha and factor VII and risk of ischemic stroke: a meta-analysis.
Stroke
2008
39
1710
1716
72
Bongers
TN
de Maat
MP
van Goor
ML
et al
High von Willebrand factor levels increase the risk of first ischemic stroke: influence of ADAMTS13, inflammation, and genetic variability.
Stroke
2006
37
2672
2677
73
Soriano
SG
Coxon
A
Wang
YF
et al
Mice deficient in Mac-1 (CD11b/CD18) are less susceptible to cerebral ischemia/reperfusion injury.
Stroke
1999
30
134
139
74
Romo
GM
Dong
JF
Schade
AJ
et al
The glycoprotein Ib-IX-V complex is a platelet counterreceptor for p-selectin.
J Exp Med
1999
190
803
814
75
Okada
Y
Copeland
BR
Mori
E
Tung
MM
Thomas
WS
del Zoppo
GJ
P-selectin and intercellular adhesion molecule-1 expression after focal cerebral brain ischemia and reperfusion.
Stroke
1994
25
202
211
76
Wu
D
Vanhoorelbeke
K
Cauwenberghs
N
et al
Inhibition of the von Willebrand (vWF)-collagen interaction by an antihuman VWF monoclonal antibody results in abolition of in vivo arterial platelet thrombus formation in baboons.
Blood
2002
99
3623
3628
77
Fontayne
A
Vanhoorelbeke
K
Pareyn
I
et al
Rational humanization of the powerful antithrombotic anti-GPIbalpha antibody 6B4.
Thromb Haemost
2006
96
671
684
78
Jackson
SP
Nesbitt
WS
Kulkarni
S
Signaling events underlying thrombus formation.
J Thromb Haemost
2003
1
1602
1612
79
Nieswandt
B
Watson
SP
Platelet-collagen interaction: is GPVI the central receptor?
Blood
2003
102
449
461
80
Clemetson
JM
Polgar
J
Magnenat
E
Wells
TN
Clemetson
KJ
The platelet collagen receptor glycoprotein VI is a member of the immunoglobulin superfamily closely related to FcalphaR and the natural killer recepotrs.
J Biol Chem
1999
274
29019
29024
81
Nieswandt
B
Schulte
V
Bergmeier
W
et al
Long-term antithrombotic protection by in vivo depletion of platelet glycoprotein VI in mice.
J Exp Med
2001
193
459
469
82
Massberg
S
Gawaz
M
Grüner
S
et al
A crucial role of glycoprotein VI for platelet recruitment to the injured arterial wall in vitro.
J Exp Med
2003
197
41
49
83
Grüner
S
Prostredna
M
Schulte
V
et al
Multiple integrin-ligand interactions synergize in shear-resistant platelet adhesion at sites of arterial injury in vivo.
Blood
2003
102
4021
4027
84
Furie
B
Furie
BC
In vivo thrombus formation.
J Thromb Hemost
2007
5
suppl 1
12
17
85
Feske
S
Calcium signalling in lymphocyte activation and disease.
Nat Rev Immunol
2007
7
690
702
86
Grosse
J
Braun
A
Varga-Szabo
D
et al
An EF hand mutation in STIM 1 causes premature platelet activation and bleeding in Mice.
J Clin Invest
2007
117
3540
3550
87
Varga-Szabo
D
Braun
A
Kleinschnitz
C
et al
The calcium sensor STIM1 is an essential mediator of arterial thrombosis and ischemic brain infarction.
J Exp Med
2008
205
1583
1591
88
Renné
T
Pozgajova
M
Grüner
S
et al
Defective thrombus formation in mice lacking coagulation factor XII.
J Exp Med
2005
202
271
281
89
Mackman
N
Role of tissue factor in hemostasis, thrombosis, and vascular development.
Arterioscler Thromb Vasc Biol
2004
24
1015
1022
90
Kannemeier
C
Shibamiya
A
Nakazawa
F
et al
Extracellular RNA constitutes a natural procoagulant cofactor in blood coagulation.
Proc Natl Acad Sci U S A
2007
104
6388
6393
91
Salomon
O
Steinberg
DM
Koren-Mong
N
Tanne
D
Seligsohn
U
Reduced incidence of ischemic stroke in patients with severe factor XI deficiency.
Blood
2008
111
4113
4117
92
Choudhri
TF
Hoh
BL
Prestigiacomo
CJ
et al
Targeted inhibition of intrinsic coagulation limits cerebral injury in stroke without increasing intracerebral hemorrhage.
J Exp Med
1999
190
91
99
93
Salomon
O
Steinberg
DM
Seligsohn
U
Variable bleeding manifestations characterize different types of surgery in patients with severe factor XI deficiency enabling parsimonious use of replacement therapy.
Haemophilia
2006
12
490
493
94
Osterud
B
Rapaport
SI
Activation of factor IX by the reaction product of tissue factor and factor VII: additional pathway for initiating blood coagulation.
Proc Natl Acad Sci U S A
1977
74
5260
5264
95
Coleman
JW
Are hemoastasis and thrombosis two sides of the same coin.
J Exp Med
2006
203
493
495
96
Mocco
J
Mack
WJ
Ducruet
AF
et al
Preclinical evaluation of the neuroprotective effect of soluble complement receptor type 1 in a nonhuman primate model of reperfused stroke.
J Neurosurg
2006
105
595
601
97
Bendszus
M
Stoll
G
Silent cerebral ischemia: hidden fingerprints of invasive medical procedures.
Lancet Neurol
2006
5
364
372
98
Robert
S
Bertolla
C
Masereel
B
Dogné
JM
Pochet
L
Novel 3-carboxamide-coumarins as potent and selective FXIIa inhibitors.
J Med Chem
2008
51
3077
3080
Sign in via your Institution