Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 97
1.
Pharmacotherapy ; 41(10): 828-836, 2021 10.
Article En | MEDLINE | ID: mdl-34478577

STUDY OBJECTIVE: This study aimed to determine the safety and efficacy of a novel GP Ib receptor inhibitor in patients with non-ST segment elevation myocardial infarction (NSTEMI) undergoing percutaneous coronary intervention (PCI). DESIGN AND SETTING: Multicenter, randomized, double-blind, placebo-controlled, dose-escalating, phase Ib-IIa clinical trial. Eligible patients were randomly assigned to the low-dose (n=20, 2 IU/60 kg), moderate-dose (n=20, 3 IU/60 kg), or high-dose anfibatide group (n=20, 5 IU/60 kg), or the placebo group (n=30). Anfibatide was administered for up to 48 hours along with standard aspirin and clopidogrel therapy. PATIENTS: Ninety patients with NSTEMI who underwent PCI at six academic hospitals in China. MEASUREMENTS AND MAIN RESULTS: All three doses of anfibatide showed dose-dependent antiplatelet activity as measured by ex vivo platelet aggregation at 5 minutes, 24 hours, and 48 hours during infusion, and 4 hours post-infusion compared with placebo. Higher inhibition of platelet aggregation occurred in all anfibatide groups compared with the placebo group. The post-procedural TIMI grade flow, myocardial blush grade, and corrected TIMI frame count were not significantly different among the four groups. Thirty-day mortality, non-fatal myocardial infarction, and major bleeding were rare and comparable between patients who received anfibatide and placebo. There was no significant difference in the platelet count among the groups during follow-up. CONCLUSIONS: This study shows that intravenous administration of the platelet receptor GP Ib antagonist anfibatide is feasible and safe to inhibit platelet aggregation without increasing the risk of bleeding and thrombocytopenia in patients with NSTEMI undergoing PCI.


Non-ST Elevated Myocardial Infarction , Platelet Aggregation Inhibitors , Platelet Glycoprotein GPIb-IX Complex , Double-Blind Method , Humans , Non-ST Elevated Myocardial Infarction/drug therapy , Percutaneous Coronary Intervention , Platelet Aggregation Inhibitors/adverse effects , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Treatment Outcome
2.
J Clin Lab Anal ; 35(9): e23939, 2021 Sep.
Article En | MEDLINE | ID: mdl-34347925

AIM: To test a novel method of assessment of platelet adhesion to a fibrinogen-coated surface in whole blood under flow conditions. METHODS: We developed a fluidic device that mimics blood flow in vessels. The method of detection of platelet adhesion is based on recording of a scattered laser light signal from a fibrinogen-covered surface. Testing was performed in platelet-rich plasma (PRP) and whole blood of healthy volunteers. Control measurements were performed, followed by tests with inhibition of platelet GPIIa/IIIb and GPIb receptors. Then, the same testing sequence was performed in whole blood of persons with autoimmune thrombocytopenia and type 3 von Willebrand disease. RESULTS: The change in intensity of scattered light was 2.7 (2.4; 4.1) times higher in whole blood (0.2 ± 0.08V, n = 7) than in PRP (0.05 ± 0.02 V, n = 7), p < 0.01. The blocking of GP IIb/IIIa receptors decreased the intensity of scattered light to 8.5 (6.5;12)%; the blocking of GPIb receptors decreased it to 34 (23;58)%, p < 0.01. In the whole blood of a person with autoimmune thrombocytopenia, the inhibition of GPIb receptors decreased platelet adhesion, but no effect was observed in type 3 von Willebrand disease. Inhibition of platelet GPIIb/IIIa receptors alone or combined inhibition of GPIb and GPIIb/IIIa receptors resulted in almost total suppression of adhesion in both cases. CONCLUSION: Our system effectively registers platelet adhesion to a fibrinogen-coated surface under controlled-flow conditions and may successfully be applied to the investigation of platelet adhesion kinetics.


Blood Platelets/metabolism , Fibrinogen/metabolism , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Healthy Volunteers , Humans , Kinetics , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors
3.
Sci Rep ; 11(1): 11663, 2021 06 03.
Article En | MEDLINE | ID: mdl-34083615

The interaction of platelet GPIbα with von Willebrand factor (VWF) is essential to initiate platelet adhesion and thrombosis, particularly under high shear stress conditions. However, no drug targeting GPIbα has been developed for clinical practice. Here we characterized anfibatide, a GPIbα antagonist purified from snake (Deinagkistrodon acutus) venom, and evaluated its interaction with GPIbα by surface plasmon resonance and in silico modeling. We demonstrated that anfibatide interferds with both VWF and thrombin binding, inhibited ristocetin/botrocetin- and low-dose thrombin-induced human platelet aggregation, and decreased thrombus volume and stability in blood flowing over collagen. In a single-center, randomized, and open-label phase I clinical trial, anfibatide was administered intravenously to 94 healthy volunteers either as a single dose bolus, or a bolus followed by a constant rate infusion of anfibatide for 24 h. Anfibatide inhibited VWF-mediated platelet aggregation without significantly altering bleeding time or coagulation. The inhibitory effects disappeared within 8 h after drug withdrawal. No thrombocytopenia or anti-anfibatide antibodies were detected, and no serious adverse events or allergic reactions were observed during the studies. Therefore, anfibatide was well-tolerated among healthy subjects. Interestingly, anfibatide exhibited pharmacologic effects in vivo at concentrations thousand-fold lower than in vitro, a phenomenon which deserves further investigation.Trial registration: Clinicaltrials.gov NCT01588132.


Blood Platelets/drug effects , Blood Platelets/metabolism , Crotalid Venoms/therapeutic use , Fibrinolytic Agents/therapeutic use , Lectins, C-Type/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Snake Venoms/therapeutic use , Animals , Blood Coagulation/drug effects , Crotalid Venoms/chemistry , Crotalid Venoms/isolation & purification , Crotalid Venoms/pharmacokinetics , Crotalinae , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/isolation & purification , Fibrinolytic Agents/pharmacokinetics , Healthy Volunteers , Humans , Lectins, C-Type/chemistry , Lectins, C-Type/isolation & purification , Models, Molecular , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Count , Platelet Glycoprotein GPIb-IX Complex/chemistry , Protein Binding , Protein Conformation , Ristocetin/pharmacology , Snake Venoms/chemistry , Snake Venoms/isolation & purification , Snake Venoms/pharmacokinetics , Structure-Activity Relationship , Thrombin/pharmacology , Thrombosis/prevention & control , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism
4.
Biomed Pharmacother ; 140: 111698, 2021 Aug.
Article En | MEDLINE | ID: mdl-34029954

Glycoprotein (GP) Ib is a platelet membrane receptor complex exposed to vascular injury, proposed as an effective target for stroke therapy. Previously, we have observed that the GPIb antagonist anfibatide (ANF) could mitigate blood-brain barrier (BBB) disruption following cerebral ischemia/reperfusion (CI/R) injury. The current study was designed to investigate whether the amelioration of the BBB by ANF is mediated via the Epac signaling pathway. A murine model of CI/R injury was induced following 90 min of transient middle cerebral artery occlusion (MCAO). ANF (4 µg/kg) was intravenously injected 1 h after reperfusion. Herein, ANF ameliorated BBB disruption, increased the expression of tight junction proteins, suppressed F-actin cytoskeleton rearrangement, decreased the permeability of the ischemic brain tissue, and relieved brain edema. ANF-treated mice had smaller infarct volumes and less severe neurological deficits than the MCAO mice. Moreover, the effects of ANF and Epac1 agonists were very similar in the MCAO mice. Epac activation with a cAMP analog, 8-CPT-2'-O-Me-cAMP, mitigated the breakdown of BBB function and CI/R injury. The Epac specific antagonist, ESI-09, worsened barrier damage and cerebral impairment, antagonizing the protective effects afforded by ANF. In addition, ANF upregulated the expression of Epac1 protein in the ischemic cerebral cortex. Collectively, our results indicate that the protective effect of ANF on the BBB after CI/R could be attributed to the activation of the Epac pathway.


Blood Platelets/drug effects , Blood-Brain Barrier/drug effects , Brain Ischemia/drug therapy , Guanine Nucleotide Exchange Factors/metabolism , Ischemic Stroke/drug therapy , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Animals , Blood Platelets/metabolism , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Brain Edema/drug therapy , Brain Edema/metabolism , Brain Ischemia/metabolism , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/metabolism , Ischemic Stroke/metabolism , Male , Mice , Mice, Inbred C57BL , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Signal Transduction/drug effects
5.
Stroke Vasc Neurol ; 5(2): 185-197, 2020 06.
Article En | MEDLINE | ID: mdl-32606086

Arterial thrombosis is in part contributed by excessive platelet aggregation, which can lead to blood clotting and subsequent heart attack and stroke. Platelets are sensitive to the haemodynamic environment. Rapid haemodynamcis and disturbed blood flow, which occur in vessels with growing thrombi and atherosclerotic plaques or is caused by medical device implantation and intervention, promotes platelet aggregation and thrombus formation. In such situations, conventional antiplatelet drugs often have suboptimal efficacy and a serious side effect of excessive bleeding. Investigating the mechanisms of platelet biomechanical activation provides insights distinct from the classic views of agonist-stimulated platelet thrombus formation. In this work, we review the recent discoveries underlying haemodynamic force-reinforced platelet binding and mechanosensing primarily mediated by three platelet receptors: glycoprotein Ib (GPIb), glycoprotein IIb/IIIa (GPIIb/IIIa) and glycoprotein VI (GPVI), and their implications for development of antithrombotic 'mechano-medicine' .


Arterial Occlusive Diseases/drug therapy , Blood Coagulation/drug effects , Blood Platelets/drug effects , Fibrinolytic Agents/therapeutic use , Hemodynamics , Mechanotransduction, Cellular/drug effects , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Thrombosis/drug therapy , Animals , Arterial Occlusive Diseases/blood , Arterial Occlusive Diseases/physiopathology , Blood Platelets/metabolism , Fibrinolytic Agents/adverse effects , Humans , Molecular Targeted Therapy , Platelet Aggregation Inhibitors/adverse effects , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/metabolism , Stress, Mechanical , Thrombosis/blood , Thrombosis/diagnosis
6.
Blood ; 135(25): 2292-2301, 2020 06 18.
Article En | MEDLINE | ID: mdl-32157300

Immune thrombocytopenia (ITP) is an acquired bleeding disorder characterized by antibody-mediated platelet destruction. Different mechanisms have been suggested to explain accelerated platelet clearance and impaired thrombopoiesis, but the pathophysiology of ITP has yet to be fully delineated. In this study, we tested 2 mouse models of immune-mediated thrombocytopenia using the rat anti-mouse GPIbα monoclonal antibody 5A7, generated in our laboratory. After a single IV administration of high-dose (2 mg/kg) 5A7, opsonized platelets were rapidly cleared from the circulation into the spleen and liver; this was associated with rapid upregulation of thrombopoietin (TPO) messenger RNA. In contrast, subcutaneous administration of low-dose 5A7 (0.08-0.16 mg/kg) every 3 days gradually lowered the platelet count; in this case, opsonized platelets were observed only in the spleen, and TPO levels remained unaltered. Interestingly, in both models, the 5A7 antibody was found on the surface of, as well as internalized to, bone marrow megakaryocytes. Consequently, platelets generated in the chronic phase of repeated subcutaneous 5A7 administration model showed reduced GPIbα membrane expression on their surface. Our findings indicate that evaluation of platelet surface GPIbα relative to platelet size may be a useful marker to support the diagnosis of anti-GPIbα antibody-induced ITP.


Antibodies, Monoclonal/immunology , Platelet Glycoprotein GPIb-IX Complex/immunology , Purpura, Thrombocytopenic, Idiopathic/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/toxicity , Antigen-Antibody Reactions , Blood Platelets/immunology , Disease Models, Animal , Injections, Intravenous , Injections, Subcutaneous , Liver/metabolism , Mice , Mice, Inbred C57BL , Opsonin Proteins/immunology , Platelet Aggregation/immunology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Purpura, Thrombocytopenic, Idiopathic/etiology , RNA, Messenger/biosynthesis , Rats , Spleen/pathology , Thrombopoietin/biosynthesis , Thrombopoietin/genetics , Up-Regulation
7.
Phytomedicine ; 59: 152903, 2019 Jun.
Article En | MEDLINE | ID: mdl-30981188

BACKGROUND: The prevalence of procoagulant state under prolonged hypoxic exposures and the complications and lack of specificity associated with use of existing anti-thrombotic agents have necessitated the search for safer and natural therapeutics. Codonopsis, a widely studied medicinal herb, has been reported to decrease whole blood viscosity but the bioactive ingredients involved, and their mechanism of action therein however remain to be investigated. PURPOSE: The present study aimed at evaluating the efficacy of C. clematidea root extract and mechanism of action of its bioactive constituent flavonoid, Kaempferol, in ameliorating hypobaric hypoxia induced procoagulant state. METHODS: Fingerprinting analysis of methanolic extract of C. clematidea root was performed by RP-HPLC. In vitro toxicity study was conducted using HUVEC cell line and in vivo acute and sub-acute toxicity were done according to OECD guidelines (section-4, number-420 and 407 respectively). Adult male Sprague-Dawley rats weighing 230-250 g were exposed to global hypoxia simulating an altitude of 7600 m (282 mmHg), in animal decompression chamber for 3, 7, 14 and 21 days for in vivo studies. Dose optimisation of the extract was done by quantification of Thromboxane A2 in the serum of hypoxic rats. C. clematidea root extract was also evaluated for its in vitro and in vivo antioxidant properties. Procoagulant changes were studied by biochemical plasma coagulation assays and expression analysis of the signalling molecules of the platelet activation cascade like vWF, platelet activation marker CD41, GpIb-IX-V (CD42), Lyn kinase, p-PI3K, p-ERK and p-PLCγ were conducted to investigate C. clematidea mediated signalling mechanisms. RESULTS: Methanolic extract of C. clematidea root showed improved antioxidant status and improvement in bleeding time and in vitro coagulation assays like pT, aPTT, INR. Decreased concentrations of D-Dimers along with that of platelet activation marker CD41 and serum concentration of Thromboxane A2 were observed in C. clematidea root extract supplemented hypoxic animals. Phosphorylation of Lyn kinase, was reduced despite increase in concentration of activating ligand vWF. CONCLUSION: C. clematidea root extract was effective in preventing hypoxia induced platelet activation and resultant procoagulant state by inhibiting Lyn kinase, a serine threonine kinase effector of vWF signalling cascade.


Codonopsis/chemistry , Hypoxia/complications , Plant Extracts/pharmacology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , src-Family Kinases/metabolism , Animals , Blood Coagulation/drug effects , Enzyme Activation/drug effects , Kaempferols/pharmacology , Male , Methanol/chemistry , Phosphorylation/drug effects , Plant Extracts/chemistry , Plant Roots/chemistry , Platelet Activation/drug effects , Platelet Glycoprotein GPIb-IX Complex/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Thromboxane A2/blood
8.
Nat Commun ; 10(1): 1204, 2019 03 13.
Article En | MEDLINE | ID: mdl-30867419

Platelets contract forcefully after their activation, contributing to the strength and stability of platelet aggregates and fibrin clots during blood coagulation. Viscoelastic approaches can be used to assess platelet-induced clot strengthening, but they require thrombin and fibrin generation and are unable to measure platelet forces directly. Here, we report a rapid, microfluidic approach for measuring the contractile force of platelet aggregates for the detection of platelet dysfunction. We find that platelet forces are significantly reduced when blood samples are treated with inhibitors of myosin, GPIb-IX-V, integrin αIIbß3, P2Y12, or thromboxane generation. Clinically, we find that platelet forces are measurably lower in cardiology patients taking aspirin. We also find that measuring platelet forces can identify Emergency Department trauma patients who subsequently require blood transfusions. Together, these findings indicate that microfluidic quantification of platelet forces may be a rapid and useful approach for monitoring both antiplatelet therapy and traumatic bleeding risk.


Blood Platelets/physiology , Hemorrhage/diagnosis , Microfluidics/methods , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/physiology , Wounds and Injuries/complications , Adult , Aspirin/pharmacology , Aspirin/therapeutic use , Blood Coagulation/drug effects , Blood Platelets/drug effects , Computer Simulation , Cross-Sectional Studies , Drug Monitoring/methods , Female , Hemorrhage/etiology , Hemorrhage/therapy , Humans , Male , Middle Aged , Myosins/antagonists & inhibitors , Myosins/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/metabolism , Prognosis , Thromboxane A2/metabolism , Wounds and Injuries/blood , Wounds and Injuries/therapy
9.
Cell Biochem Funct ; 37(2): 56-61, 2019 Mar.
Article En | MEDLINE | ID: mdl-30701573

Neutrophils participate in the regulation of pathogens by phagocytosis as well as by generating neutrophil extracellular traps (NETs). Antiphospholipid antibodies, particularly those targeting beta-2-glycoprotein I (ß2GPI), stimulate monocytes, platelets, and endothelial cells with prothrombotic participation. This study aimed to explore NET generation in response to anti-ß2GPI/ß2GPI. A series of experiments involving the separation of primary human leukocytes, NETosis quantification using propidium iodide, exploration of NETosis by fluorescence microscopy, western blotting, examination of free Zn2+ using FluoZin-3, and reactive oxygen species (ROS) examination with dihydrorhodamine 123 were performed in this study. We found that anti-ß2GPI/ß2GPI triggered NETosis, resembling phorbol 12-myristate 13-acetate (PMA)-induced NETosis in magnitude and morphology. The anti-ß2 GPI/ß2 GPI complex in isolation stimulated NETs without relying on p38, protein kinase B (AKT), extracellular signal-related kinase (ERK) 1/2, and zinc signals. NET generation was unaffected by the NADPH oxidase suppressor DP1. The anti-ß2 GPI/ß2 GPI complex stimulated ROS generation without relying on NADPH oxidase, which may participate in NET generation triggered via the anti-ß2 GPI/ß2 GPI complex. In summary, our results indicate that the anti-ß2 GPI/ß2 GPI complex reinforced NET generation by relying on ROS. THE SIGNIFICANCE OF THE PAPER IN THE CONTEXT OF CURRENT KNOWLEDGE: Neutrophils as one of the first lines of defence and essential in the response to pathogen invasion. They eradicate bacteria via phagocytosis or by releasing antimicrobial proteins in degranulation. In this study, we explored the capability of anti-ß2 GPI/ß2 GPI to stimulate NETosis, demonstrating that anti-ß2 GPI/ß2 GPI is a promising method for triggering NET. Anti-ß2 GPI/ß2 GPI induced ROS generation without relying on NADPH oxidase, which contributes to NETosis independently of ERK1/2, Zn2+ , or AKT. Our results showed that anti-ß2GPI/ß2GPI triggered NETosis, resembling PMA-induced NETosis in magnitude as well as morphology. The anti-ß2 GPI/ß2 GPI complex in isolation stimulated NETs without relying on p38, AKT, ERK1/2, or zinc signals. The anti-ß2 GPI/ß2 GPI complex stimulated ROS generation without relying on NADPH oxidase, which may participate in NET generation triggered via the anti-ß2 GPI/ß2 GPI complex.


Antibodies/pharmacology , Extracellular Traps/metabolism , MAP Kinase Signaling System/drug effects , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/metabolism , Reactive Oxygen Species/metabolism , Humans
10.
Blood ; 131(17): 1899-1902, 2018 04 26.
Article En | MEDLINE | ID: mdl-29467183

Current antithrombotic drugs, including widely used antiplatelet agents and anticoagulants, are associated with significant bleeding risk. Emerging experimental evidence suggests that the molecular and cellular mechanisms of hemostasis and thrombosis can be separated, thereby increasing the possibility of new antithrombotic therapeutic targets with reduced bleeding risk. We review new coagulation and platelet targets and highlight the interaction between integrin αMß2 (Mac-1, CD11b/CD18) on leukocytes and GPIbα on platelets that seems to distinguish thrombosis from hemostasis.


Drug Discovery , Fibrinolytic Agents , Hemorrhage/prevention & control , Hemostasis/drug effects , Thrombosis/drug therapy , Animals , Blood Platelets/metabolism , Fibrinolytic Agents/adverse effects , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/therapeutic use , Hemorrhage/metabolism , Humans , Integrins/antagonists & inhibitors , Integrins/metabolism , Leukocytes/metabolism , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombosis/metabolism , Thrombosis/pathology
11.
Immunology ; 154(2): 322-328, 2018 06.
Article En | MEDLINE | ID: mdl-29325217

Neutrophils are key components of the innate immune response, providing host defence against infection and being recruited to non-microbial injury sites. Platelets act as a trigger for neutrophil extravasation to inflammatory sites but mechanisms and tissue-specific aspects of these interactions are currently unclear. Here, we use bacterial endotoxin in mice to trigger an innate inflammatory response in different tissues and measure neutrophil invasion with or without platelet reduction. We show that platelets are essential for neutrophil infiltration to the brain, peritoneum and skin. Neutrophil numbers do not rise above basal levels in the peritoneum and skin and are decreased (~60%) in the brain when platelet numbers are reduced. In contrast neutrophil infiltration in the lung is unaffected by platelet reduction, up-regulation of CXCL-1 (2·4-fold) and CCL5 (1·4-fold) acting as a compensatory mechanism in platelet-reduced mice during lung inflammation. In brain inflammation targeting platelet receptor GPIbα results in a significant decrease (44%) in platelet-mediated neutrophil invasion, while maintaining platelet numbers in the circulation. These results suggest that therapeutic blockade of platelet GPIbα could limit the harmful effects of excessive inflammation while minimizing haemorrhagic complications of platelet reduction in the brain. The data also demonstrate the ability to target damaging brain inflammation in stroke and related disorders without compromising lung immunity and hence risk of pneumonia, a major complication post stroke. In summary, our data reveal an important role for platelets in neutrophil infiltration to various tissues, including the brain, and so implicate platelets as a key, targetable component of cerebrovascular inflammatory disease or injury.


Blood Platelets/drug effects , Blood Platelets/metabolism , Brain/immunology , Brain/metabolism , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/metabolism , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Animals , Antibodies, Monoclonal/pharmacology , Brain/pathology , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Disease Models, Animal , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/immunology , Mice
12.
Transl Stroke Res ; 9(5): 493-498, 2018 10.
Article En | MEDLINE | ID: mdl-29322481

In acute ischemic stroke (AIS), there is an alarming discrepancy between recanalization rates of up to 70% by combined recombinant tissue-type plasminogen activator (rt-PA) therapy and mechanical thrombectomy, and no clinical benefit in at least every second stroke patient. This is partly due to ischemia/reperfusion (I/R) injury. In a translational approach, we used mice lacking dense- (Unc13d-/-) or α-granules (Nbeal2-/-) and mice after blocking of platelet glycoprotein receptor (GP) Ib conferring protection from I/R injury. These mice underwent transient middle cerebral artery occlusion (tMCAO) and, as in the clinic, were treated with rt-PA. Our data show that rt-PA treatment is still safe in conjunction with selected anti-platelet therapies and pave the way for eagerly awaited additive treatment options in acute human stroke.


Fibrinolytic Agents/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Stroke/therapy , Thrombectomy/methods , Animals , Blood Proteins/genetics , Blood Proteins/metabolism , Brain Ischemia/complications , Disease Models, Animal , Hemoglobins/metabolism , Intracranial Hemorrhages/etiology , Magnetic Resonance Imaging , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Platelet Glycoprotein GPIb-IX Complex/metabolism , Statistics, Nonparametric , Stroke/diagnostic imaging , Stroke/etiology , Tissue Plasminogen Activator/therapeutic use , Treatment Outcome
13.
Ann Hematol ; 97(4): 641-654, 2018 Apr.
Article En | MEDLINE | ID: mdl-29350259

Anti-GPIIb/IIIa-mediated complement activation has been reported to be important in the pathogenesis of immune thrombocytopenia (ITP). However, the role of the complement system and the involved regulatory mechanism remain equivocal. Beta2-glycoprotein I (ß2-GPI), known as the main target for antiphospholipid autoantibodies, has been demonstrated as a complement regulator. Here, we investigated the complement-regulatory role of ß2-GPI in anti-GPIIb/IIIa-mediated ITP. Plasma complement activation and enhanced complement activation capacity (CAC) were found in ITP patients with anti-GPIIb/IIIa antibodies in vivo and in vitro. Diminished plasma levels of ß2-GPI were shown in patients of this group, which was inversely correlated with C5b-9 deposition. C5b-9 generation was inhibited by approximate physiological concentrations of ß2-GPI, in a dose-dependent manner. Inhibition of C3a generation by ß2-GPI and the existence of ß2-GPI/C3 complexes in plasma indicated a regulation on the level of the C3 convertase. Furthermore, ß2-GPI down-regulated the phosphorylation levels of c-Jun N-terminal kinase (JNK) and cleavage of BH3 interacting domain death agonist (Bid) and ultimately harbored platelet lysis. Our findings may provide a novel link between diminished plasma levels of ß2-GPI and enhanced complement activation, indicating ß2-GPI as a potential diagnostic biomarker and therapeutic target in the treatment of anti-GPIIb/IIIa-mediated ITP.


Complement Activation , Down-Regulation , Isoantibodies/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Purpura, Thrombocytopenic, Idiopathic/metabolism , beta 2-Glycoprotein I/metabolism , Adult , Aged , Biomarkers/blood , Blood Platelets/immunology , Blood Platelets/metabolism , Blood Platelets/pathology , China/epidemiology , Complement C3-C5 Convertases/metabolism , Complement C3a/metabolism , Female , Humans , Male , Middle Aged , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/metabolism , Purpura, Thrombocytopenic, Idiopathic/immunology , Purpura, Thrombocytopenic, Idiopathic/pathology , Purpura, Thrombocytopenic, Idiopathic/physiopathology , Risk , Thrombocytopenia/blood , Thrombocytopenia/immunology , Thrombocytopenia/metabolism , Thrombosis/epidemiology , Thrombosis/etiology , Young Adult , beta 2-Glycoprotein I/blood
14.
Brain Behav Immun ; 69: 255-263, 2018 03.
Article En | MEDLINE | ID: mdl-29195783

Blood-brain barrier (BBB) disruption, thrombus formation and immune-mediated inflammation are important steps in the pathophysiology of cerebral ischemia-reperfusion injury but are still inaccessible to therapeutic interventions. Recent studies have provided increasing evidence that blocking of platelet glycoprotein (GP) receptor Ib might represent a novel target in treating acute ischemic stroke. This research was conducted to explore the therapeutic efficacy and potential mechanisms of GPIbα inhibitor (anfibatide) in a model of brain ischemia-reperfusion injury in mice. Male mice underwent 90 min of right middle cerebral artery occlusion (MCAO) followed by 24 h of reperfusion. Anfibatide (1, 2, 4 ug/kg) or tirofiban were administered intravenously 1 h after reperfusion. The results showed that anfibatide could significantly reduce infarct volumes, increase the number of intact neuronal cells and improve neurobehavioral function. Moreover, anfibatide could reduce post ischemic BBB damage by attenuating increased paracellular permeability in the ischemia hemisphere significantly. Stroke-induced increases in activity and protein expression of macrophage-1 antigen (MAC-1) and P-selectin were also reduced by anfibatide intervention. Finally, anfibatide exerted antithrombotic effects upon stroke by decreased the number of microthrombi formation. This is the first demonstration of anfibatide's efficacy in protecting the BBB integrity and decreasing neutrophil inflammation response mediated by MAC-1 besides microthrombus formation inhibition in the brain during reperfusion. Anfibatide, as a promising anti-thrombo-inflammation agent, could be beneficial for the treatment of ischemic stroke.


Blood-Brain Barrier/drug effects , Crotalid Venoms/therapeutic use , Fibrinolytic Agents/therapeutic use , Lectins, C-Type/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Reperfusion Injury/drug therapy , Tirofiban/therapeutic use , Animals , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Crotalid Venoms/pharmacology , Fibrinolytic Agents/pharmacology , Infarction, Middle Cerebral Artery/metabolism , Male , Mice , Neurons/drug effects , Neurons/metabolism , Reperfusion Injury/metabolism , Tirofiban/pharmacology
15.
J Biol Chem ; 292(45): 18608-18617, 2017 11 10.
Article En | MEDLINE | ID: mdl-28924049

The plasma protein von Willebrand factor (VWF) is essential for hemostasis initiation at sites of vascular injury. The platelet-binding A1 domain of VWF is connected to the VWF N-terminally located D'D3 domain through a relatively unstructured amino acid sequence, called here the N-terminal linker. This region has previously been shown to inhibit the binding of VWF to the platelet surface receptor glycoprotein Ibα (GpIbα). However, the molecular mechanism underlying the inhibitory function of the N-terminal linker has not been elucidated. Here, we show that an aspartate at position 1261 is the most critical residue of the N-terminal linker for inhibiting binding of the VWF A1 domain to GpIbα on platelets in blood flow. Through a combination of molecular dynamics simulations, mutagenesis, and A1-GpIbα binding experiments, we identified a network of salt bridges between Asp1261 and the rest of A1 that lock the N-terminal linker in place such that it reduces binding to GpIbα. Mutations aimed at disrupting any of these salt bridges activated binding unless the mutated residue also formed a salt bridge with GpIbα, in which case the mutations inhibited the binding. These results show that interactions between charged amino acid residues are important both to directly stabilize the A1-GpIbα complex and to indirectly destabilize the complex through the N-terminal linker.


Aspartic Acid/chemistry , Blood Flow Velocity , Blood Platelets/metabolism , Models, Molecular , Platelet Glycoprotein GPIb-IX Complex/metabolism , von Willebrand Factor/metabolism , Amino Acid Substitution , Binding Sites , Cell Adhesion , Gene Deletion , Humans , Microspheres , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/chemistry , Platelet Glycoprotein GPIb-IX Complex/genetics , Point Mutation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Static Electricity , von Willebrand Factor/antagonists & inhibitors , von Willebrand Factor/chemistry , von Willebrand Factor/genetics
16.
Thromb Haemost ; 117(7): 1249-1257, 2017 06 28.
Article En | MEDLINE | ID: mdl-28597906

Anti-platelet drugs reduce arterial thrombosis after plaque rupture and erosion, prevent stent thrombosis and are used to prevent and treat myocardial infarction and ischaemic stroke. Some of them may also be helpful in treating less frequent diseases such as thrombotic thrombocytopenic purpura. The present concise review aims to cover current and future developments of anti-platelet drugs interfering with the interaction of von Willebrand factor (VWF) with glycoprotein (GP) Ibα, and directed against GPVI, GPIIb/IIIa (integrin αIIbß3), the thrombin receptor PAR-1, and the ADP receptor P2Y12. The high expectations of having novel antiplatelet drugs which selectively inhibit arterial thrombosis without interfering with normal haemostasis could possibly be met in the near future.


Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/antagonists & inhibitors , Blood Platelets/drug effects , Blood Platelets/metabolism , Humans , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Purinergic P2Y Receptor Antagonists/therapeutic use , Receptor, PAR-1/antagonists & inhibitors , Thrombin/antagonists & inhibitors , Thrombosis/blood , Thrombosis/prevention & control , von Willebrand Factor/antagonists & inhibitors
17.
Arterioscler Thromb Vasc Biol ; 36(9): 1821-8, 2016 09.
Article En | MEDLINE | ID: mdl-27417583

OBJECTIVE: The platelet storage lesion accelerates platelet clearance after transfusion, but the underlying molecular mechanism remains elusive. Although inhibiting sheddase activity hampers clearance of platelets with storage lesion, the target platelet protein responsible for ectodomain shedding-induced clearance is not definitively identified. Monoclonal antibody 5G6 was developed recently to bind specifically human platelet receptor glycoprotein (GP)Ibα and inhibit its shedding but not shedding of other receptors. Here, the role of GPIbα shedding in platelet clearance after transfusion was addressed. APPROACH AND RESULTS: Both human leukoreduced apheresis-derived platelets and transgenic mouse platelets expressing human GPIbα were stored at room temperature in the presence and absence of 5G6 Fab fragment. At various time points, aliquots of stored platelets were analyzed and compared. 5G6 Fab inhibited GPIbα shedding in both platelets during storage and preserved higher level of GPIbα on the platelet surface. Compared with age-matched control platelets, 5G6 Fab-stored platelets exhibited similar levels of platelet activation, degranulation, and agonist-induced aggregation. 5G6 Fab-stored human GPIbα platelets exhibited significantly higher post-transfusion recovery and in vivo hemostatic function in recipient mice than control platelets. Consistently, 5G6 Fab-stored, 8-day-old human platelets produced similar improvement in post-transfusion recovery in immunodeficient mice and in ex vivo thrombus formation over collagen under shear flow. CONCLUSIONS: Specific inhibition of GPIbα shedding in the stored platelets improves post-transfusion platelet recovery and hemostatic function, providing clear evidence for GPIbα shedding as a cause of platelet clearance. These results suggest that specific inhibition of GPIbα shedding may be used to optimize platelet storage conditions.


Antibodies, Monoclonal/pharmacology , Blood Platelets/drug effects , Hemostasis/drug effects , Immunoglobulin Fab Fragments/pharmacology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Transfusion , Animals , Blood Component Removal , Blood Platelets/metabolism , Cell Degranulation/drug effects , Genotype , Humans , Mice, Inbred C57BL , Mice, SCID , Mice, Transgenic , Phenotype , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Transfusion/adverse effects , Time Factors
18.
Thromb Haemost ; 116(1): 87-95, 2016 07 04.
Article En | MEDLINE | ID: mdl-27052467

Multimerin 1 (MMRN1) is a massive, homopolymeric protein that is stored in platelets and endothelial cells for activation-induced release. In vitro, MMRN1 binds to the outer surfaces of activated platelets and endothelial cells, the extracellular matrix (including collagen) and von Willebrand factor (VWF) to support platelet adhesive functions. VWF associates with MMRN1 at high shear, not static conditions, suggesting that shear exposes cryptic sites within VWF that support MMRN1 binding. Modified ELISA and surface plasmon resonance were used to study the structural features of VWF that support MMRN1 binding, and determine the affinities for VWF-MMRN1 binding. High shear microfluidic platelet adhesion assays determined the functional consequences for VWF-MMRN1 binding. VWF binding to MMRN1 was enhanced by shear exposure and ristocetin, and required VWF A1A2A3 region, specifically the A1 and A3 domains. VWF A1A2A3 bound to MMRN1 with a physiologically relevant binding affinity (KD: 2.0 ± 0.4 nM), whereas the individual VWF A1 (KD: 39.3 ± 7.7 nM) and A3 domains (KD: 229 ± 114 nM) bound to MMRN1 with lower affinities. VWF A1A2A3 was also sufficient to support the adhesion of resting platelets to MMRN1 at high shear, by a mechanism dependent on VWF-GPIbα binding. Our study provides new information on the molecular basis of MMRN1 binding to VWF, and its role in supporting platelet adhesion at high shear. We propose that at sites of vessel injury, MMRN1 that is released following activation of platelets and endothelial cells, binds to VWF A1A2A3 region to support platelet adhesion at arterial shear rates.


Blood Proteins/metabolism , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism , Blood Platelets/drug effects , Blood Platelets/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , In Vitro Techniques , Microfluidic Analytical Techniques , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Platelet Activation , Platelet Adhesiveness , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Binding/drug effects , Protein Domains , Recombinant Proteins/metabolism , Ristocetin/pharmacology , Surface Plasmon Resonance , von Willebrand Factor/genetics
19.
Sci Rep ; 5: 11730, 2015 Jul 06.
Article En | MEDLINE | ID: mdl-26144864

Agkisacucetin extracted from the venom of Agkistrodon acutus has been demonstrated to be a promising antithrombotic drug candidate in clinical studies due to its function as a novel platelet membrane glycoprotein (GP) Ib inhibitor. Agkisacucetin is a heterodimeric protein composed of α- and ß-subunits with seven disulphide bonds. Both subunits form inactive homodimeric products, which cause difficulties for recombinant production. In this study, Agkisacucetin α- and ß-subunits were inserted sequentially into the chromosome of Pichia pastoris at the mutant histidinol dehydrogenase gene and ribosomal DNA repeat sites, respectively. By optimizing the gene copies and productivity of each subunit by drug screening, we successfully obtained a recombinant strain with balanced expression of the two subunits. Using this strain, a yield greater than 100 mg/L recombinant Agkisacucetin in fed-batch fermentation was reached. The recombinant Agkisacucetin possessed extremely similar binding affinity to recombinant GPIb and human platelets in in vitro assays, and its ristocetin-induced platelet aggregation activity ex vivo was identical to that of the extracted native Agkisacucetin, demonstrating that the yeast-derived Agkisacucetin could be an effective alternative to native Agkisacucetin. Moreover, this study provides an effective strategy for balancing the expression and production of heterodimeric proteins in P. pastoris.


Antineoplastic Agents/metabolism , Crotalid Venoms/metabolism , Agkistrodon/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Batch Cell Culture Techniques , Crotalid Venoms/chemistry , Crotalid Venoms/genetics , Dimerization , Humans , Molecular Sequence Data , Pichia/metabolism , Plasmids/genetics , Plasmids/metabolism , Platelet Aggregation/drug effects , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Ristocetin/pharmacology
20.
Br J Pharmacol ; 172(15): 3904-16, 2015 Aug.
Article En | MEDLINE | ID: mdl-25917571

BACKGROUND AND PURPOSE: Ischaemic stroke is a serious disease with limited therapy options. Glycoprotein (GP)Ib binding to von Willebrand factor (vWF) exposed at vascular injury initiates platelet adhesion and contributes to platelet aggregation. GPIb has been suggested as an effective target for antithrombotic therapy in stroke. Anfibatide is a GPIb antagonist derived from snake venom and we investigated its protective effect on experimental brain ischaemia in mice. EXPERIMENTAL APPROACH: Focal cerebral ischaemia was induced by 90 min of transient middle cerebral artery occlusion (MCAO). These mice were then treated with anfibatide (4, 2, 1 µg·kg(-1) ), injected i.v., after 90 min of MCAO, followed by 1 h of reperfusion. Tirofiban, a GPIIb/IIIα antagonist, was used as a positive control. KEY RESULTS: Twenty-four hours after MCAO, anfibatide-treated mice showed significantly improved ischaemic lesions in a dose-dependent manner. The mice had smaller infarct volumes, less severe neurological deficits and histopathology of cerebrum tissues compared with the untreated MCAO mice. Moreover, anfibatide decreased the amount of GPIbα, vWF and accumulation of fibrin(ogen) in the vasculature of the ischaemic hemisphere. Tirofiban had similar effects on infarct size and fibrin(ogen) deposition compared with the MCAO group. Importantly, the anfibatide-treated mice showed a lower incidence of intracerebral haemorrhage and shorter tail bleeding time compared with the tirofiban-treated mice. CONCLUSIONS AND IMPLICATIONS: Our data indicate anfibatide is a safe GPIb antagonist that exerts a protective effect on cerebral ischaemia and reperfusion injury. Anfibatide is a promising candidate that could be beneficial for the treatment of ischaemic stroke.


Brain Ischemia/prevention & control , Crotalid Venoms/pharmacology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Protective Agents/pharmacology , Reperfusion Injury/prevention & control , Stroke/prevention & control , Animals , Bleeding Time , Blood Platelets/drug effects , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Count , Cerebral Hemorrhage/prevention & control , Dose-Response Relationship, Drug , Fibrin/metabolism , Infarction, Middle Cerebral Artery , Lectins, C-Type , Male , Mice , Platelet Glycoprotein GPIb-IX Complex/metabolism , Stroke/pathology , Tirofiban , Tyrosine/analogs & derivatives , Tyrosine/pharmacology , von Willebrand Factor/metabolism
...