Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 135(25): 2292-2301, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32157300

ABSTRACT

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.


Subject(s)
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
2.
J Clin Lab Anal ; 35(9): e23939, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34347925

ABSTRACT

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.


Subject(s)
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.
Blood ; 131(17): 1899-1902, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29467183

ABSTRACT

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.


Subject(s)
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
4.
Cell Biochem Funct ; 37(2): 56-61, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30701573

ABSTRACT

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.


Subject(s)
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
5.
J Biol Chem ; 292(45): 18608-18617, 2017 11 10.
Article in English | MEDLINE | ID: mdl-28924049

ABSTRACT

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.


Subject(s)
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
6.
Immunology ; 154(2): 322-328, 2018 06.
Article in English | MEDLINE | ID: mdl-29325217

ABSTRACT

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.


Subject(s)
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
7.
Brain Behav Immun ; 69: 255-263, 2018 03.
Article in English | MEDLINE | ID: mdl-29195783

ABSTRACT

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.


Subject(s)
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
8.
Ann Hematol ; 97(4): 641-654, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29350259

ABSTRACT

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.


Subject(s)
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
9.
Arterioscler Thromb Vasc Biol ; 36(9): 1821-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27417583

ABSTRACT

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.


Subject(s)
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
10.
Arterioscler Thromb Vasc Biol ; 35(4): 930-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25722431

ABSTRACT

OBJECTIVE: The hypothesis that hypertension induces a hypercoagulable state arises from the complications associated with hypertension: stroke and myocardial infarction. Here, we determine whether hypertension causes changes in the thrombin-generating capacity of the vascular wall. APPROACH AND RESULTS: We used spontaneously hypertensive rats (SHR) compared with Wistar rats. The addition of thoracic aortic rings of SHR to a Wistar or SHR plasma pool resulted in a greater increase in thrombin generation compared with equivalent rings from Wistar. This increase occurred in 12- but not 5-week-old rats and was prevented by an angiotensin II-converting enzyme inhibitor, indicating that established hypertension is required to induce increased thrombin generation within the vessel wall. Whereas no difference was observed for endothelial cells, thrombin formation was higher on aortic smooth muscle cells (SMCs) from SHR than on those from Wistar. Exposure of negatively charged phospholipids was higher on SHR than on Wistar rings, as well as on cultured SMCs. Tissue factor activity was higher in SHR SMCs. Twelve-week-old SHR exhibited accelerated FeCl3-induced thrombus formation in carotid arteries, and the resulting occlusive thrombi were disaggregated by blockade of glycoprotein Ibα-von Willebrand factor interactions. SHR SMCs were more sensitive to thrombin-induced proliferation than Wistar SMCs. This effect was totally abolished by a protease-activated receptor 1 inhibitor. CONCLUSIONS: The prothrombotic phenotype of the SHR vessel wall was due to the ability of SMCs to support greater thrombin generation and resulted in accelerated occlusive thrombus formation after arterial injury, which was sensitive to glycoprotein Ibα-von Willebrand factor inhibitors.


Subject(s)
Blood Coagulation , Hypertension/complications , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Thrombosis/etiology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Antihypertensive Agents/pharmacology , Aorta, Thoracic/metabolism , Aorta, Thoracic/physiopathology , Blood Coagulation/drug effects , Blood Pressure , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Fibrinolytic Agents/pharmacology , Hypertension/blood , Hypertension/drug therapy , Hypertension/genetics , Hypertension/physiopathology , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Myocytes, Smooth Muscle/drug effects , Phenotype , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/metabolism , Rats, Inbred SHR , Rats, Wistar , Receptor, PAR-1/antagonists & inhibitors , Receptor, PAR-1/metabolism , Thrombin/metabolism , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/genetics , Thrombosis/physiopathology , Thrombosis/prevention & control , Time Factors , Vascular Remodeling , von Willebrand Factor/antagonists & inhibitors , von Willebrand Factor/metabolism
11.
Ann Neurol ; 75(5): 670-83, 2014 May.
Article in English | MEDLINE | ID: mdl-24644058

ABSTRACT

OBJECTIVE: Bacterial infection contributes to diverse noninfectious diseases and worsens outcome after stroke. Streptococcus pneumoniae, the most common infection in patients at risk of stroke, is a major cause of prolonged hospitalization and death of stroke patients, but how infection impacts clinical outcome is not known. METHODS: We induced sustained pulmonary infection by a human S. pneumoniae isolate in naive and comorbid rodents to investigate the effect of infection on vascular and inflammatory responses prior to and after cerebral ischemia. RESULTS: S. pneumoniae infection triggered atherogenesis, led to systemic induction of interleukin (IL) 1, and profoundly exacerbated (50-90%) ischemic brain injury in rats and mice, a response that was more severe in combination with old age and atherosclerosis. Systemic blockade of IL-1 with IL-1 receptor antagonist (IL-1Ra) fully reversed infection-induced exacerbation of brain injury and functional impairment caused by cerebral ischemia. We show that infection-induced systemic inflammation mediates its effects via increasing platelet activation and microvascular coagulation in the brain after cerebral ischemia, as confirmed by reduced brain injury in response to blockade of platelet glycoprotein (GP) Ibα. IL-1 and platelet-mediated signals converge on microglia, as both IL-1Ra and GPIbα blockade reversed the production of IL-1α by microglia in response to cerebral ischemia in infected animals. INTERPRETATION: S. pneumoniae infection augments atherosclerosis and exacerbates ischemic brain injury via IL-1 and platelet-mediated systemic inflammation. These mechanisms may contribute to diverse cardio- and cerebrovascular pathologies in humans.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Interleukin-1/adverse effects , Platelet Glycoprotein GPIb-IX Complex/adverse effects , Streptococcal Infections/metabolism , Streptococcal Infections/pathology , Streptococcus pneumoniae , Animals , Brain Ischemia/microbiology , Disease Progression , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Interleukin-1/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/metabolism , Microglia/microbiology , Microglia/pathology , Platelet Activation , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/physiology , Rats , Rats, Wistar , Streptococcal Infections/microbiology
12.
Blood ; 120(17): 3390-2, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-23100301

ABSTRACT

Two articles in this issue of Blood from Feys et al and Callewaert et al, respectively, have employed very similar and elegant strategies in attempts to ameliorate the symptoms of thrombotic thrombocytopenic purpura (TTP).


Subject(s)
Anemia, Hemolytic/drug therapy , Antibodies, Monoclonal/therapeutic use , Fibrinolytic Agents/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Purpura, Thrombotic Thrombocytopenic/drug therapy , von Willebrand Factor/antagonists & inhibitors , Animals
13.
Blood ; 120(17): 3611-4, 2012 Oct 25.
Article in English | MEDLINE | ID: mdl-22855603

ABSTRACT

The pathophysiology of thrombotic thrombocytopenic purpura (TTP) can be explained by the absence of active ADAMTS13, leading to ultra-large von Willebrand factor (UL-VWF) multimers spontaneously interacting with platelets. Preventing the formation of UL-VWF-platelet aggregates therefore is an attractive new treatment strategy. Here, we demonstrate that simultaneous administration of the inhibitory anti-VWF monoclonal antibody GBR600 and the inhibitory anti-ADAMTS13 antibody 3H9 to baboons (prevention group) precluded TTP onset as severe thrombocytopenia and hemolytic anemia were absent in these animals. In addition, partial VWF inhibition was not enough to prevent thrombocytopenia, demonstrating the specificity of this therapeutic strategy. GBR600 treatment of baboons during acute TTP (treatment group) resulted in a rapid recovery of severe thrombocytopenia similar to the platelet count increases observed in TTP patients treated by plasma exchange. Baboons in the control group only injected with 3H9 developed early stages of TTP as previously described. Hence, inhibiting VWF-GPIb interactions is an effective way to prevent and treat the early symptoms of acquired TTP in baboons.


Subject(s)
Anemia, Hemolytic/drug therapy , Antibodies, Monoclonal/therapeutic use , Fibrinolytic Agents/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Purpura, Thrombotic Thrombocytopenic/drug therapy , von Willebrand Factor/antagonists & inhibitors , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , Anemia, Hemolytic/complications , Anemia, Hemolytic/metabolism , Anemia, Hemolytic/pathology , Animals , Antibodies, Monoclonal/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/pathology , Disease Models, Animal , Fibrinolytic Agents/pharmacology , Papio , Platelet Aggregation/drug effects , Platelet Count , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Binding , Purpura, Thrombotic Thrombocytopenic/complications , Purpura, Thrombotic Thrombocytopenic/metabolism , Purpura, Thrombotic Thrombocytopenic/pathology , von Willebrand Factor/metabolism
14.
Circ Res ; 110(9): 1202-10, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22456181

ABSTRACT

RATIONALE: Multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis (EAE), are inflammatory disorders of the central nervous system (CNS). The function of platelets in inflammatory and autoimmune pathologies is thus far poorly defined. OBJECTIVE: We addressed the role of platelets in mediating CNS inflammation in EAE. METHODS AND RESULTS: We found that platelets were present in human MS lesions as well as in the CNS of mice subjected to EAE but not in the CNS from control nondiseased mice. Platelet depletion at the effector-inflammatory phase of EAE in mice resulted in significantly ameliorated disease development and progression. EAE suppression on platelet depletion was associated with reduced recruitment of leukocytes to the inflamed CNS, as assessed by intravital microscopy, and with a blunted inflammatory response. The platelet-specific receptor glycoprotein Ibα (GPIbα) promotes both platelet adhesion and inflammatory actions of platelets and targeting of GPIbα attenuated EAE in mice. Moreover, targeting another platelet adhesion receptor, glycoprotein IIb/IIIa (GPIIb/IIIa), also reduced EAE severity in mice. CONCLUSIONS: Platelets contribute to the pathogenesis of EAE by promoting CNS inflammation. Targeting platelets may therefore represent an important new therapeutic approach for MS treatment.


Subject(s)
Blood Platelets/metabolism , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/blood , Leukocytes/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Blood Platelets/drug effects , Blood Platelets/immunology , Cells, Cultured , Central Nervous System/drug effects , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Humans , Inflammation Mediators/metabolism , Leukocytes/drug effects , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/blood , Mice , Mice, Inbred C57BL , Platelet Adhesiveness , Platelet Aggregation Inhibitors/pharmacology , 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 , Time Factors
15.
Arterioscler Thromb Vasc Biol ; 33(11): 2529-37, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24051142

ABSTRACT

OBJECTIVE: Poor prognosis of sepsis is associated with bacterial lipopolysaccharide (LPS)-induced intravascular inflammation, microvascular thrombosis, thrombocytopenia, and disseminated intravascular coagulation. Platelets are critical for thrombosis, and there has been increasing evidence of the importance of platelets in endotoxemia. The platelet adhesion receptor, the glycoprotein Ib-IX complex (GPIb-IX), mediates platelet adhesion to inflammatory vascular endothelium and exposed subendothelium. Thus, we have investigated the role of GPIb-IX in LPS-induced platelet adhesion, thrombosis, and thrombocytopenia. APPROACH AND RESULTS: LPS-induced mortality is significantly decreased in mice expressing a functionally deficient mutant of GPIbα. Furthermore, we have developed a micellar peptide inhibitor, MPαC (C13H27CONH-SIRYSGHpSL), which selectively inhibits the von Willebrand factor -binding function of GPIb-IX and GPIb-IX-mediated platelet adhesion under flow without affecting GPIb-IX-independent platelet activation. MPαC inhibits platelet adhesion to LPS-stimulated endothelial cells in vitro and alleviates LPS-induced thrombosis in glomeruli in mice. Importantly, MPαC reduces mortality in LPS-challenged mice, suggesting a protective effect of this inhibitor during endotoxemia. Interestingly, MPαC, but not the integrin antagonist, Integrilin, alleviated LPS-induced thrombocytopenia. CONCLUSIONS: These data indicate an important role for the platelet adhesion receptor GPIb-IX in LPS-induced thrombosis and thrombocytopenia, and suggest the potential of targeting GPIb as an antiplatelet strategy in managing endotoxemia.


Subject(s)
Endotoxemia/metabolism , Membrane Glycoproteins/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombocytopenia/metabolism , Thrombosis/metabolism , Animals , Endothelium, Vascular/metabolism , Endotoxemia/drug therapy , Endotoxemia/mortality , Humans , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Peptides/pharmacology , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/physiology , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Thrombocytopenia/drug therapy , Thrombocytopenia/mortality , Thrombosis/drug therapy , Thrombosis/mortality , von Willebrand Factor/metabolism
16.
Bull Exp Biol Med ; 157(1): 25-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24913571

ABSTRACT

A short-term open prospective study examined 62 patients at the terminal stage of chronic renal failure. The experimental group received erythropoietin in a total dose of about 40,000 U. The expression of glycoproteins IIb-IIIa, IIb, and Ib was enhanced, the content of LPO products was elevated, and SOD and catalase activities were reduced in platelets from patients with chronic renal failure. Administration of erythropoietin partially restored free radical oxidation and expression of glycoproteins IIb-IIIa, IIb, and Ib in platelets. A significant correlation was revealed between the expression of platelet receptors on the one hand, and content of LPO products and SOD and catalase activities, on the other hand.


Subject(s)
Blood Platelets/drug effects , Erythropoietin/therapeutic use , Free Radicals/antagonists & inhibitors , Gene Expression/drug effects , Kidney Failure, Chronic/drug therapy , Blood Platelets/metabolism , Catalase/blood , Drug Administration Schedule , Female , Free Radicals/metabolism , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Lipid Peroxidation/drug effects , Male , Middle Aged , Oxidation-Reduction , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoprotein IIb/genetics , Platelet Membrane Glycoprotein IIb/metabolism , Prospective Studies , Superoxide Dismutase/blood
17.
Blood ; 117(18): 4999-5008, 2011 May 05.
Article in English | MEDLINE | ID: mdl-21398580

ABSTRACT

The membrane-anchored CX3C chemokine fractalkine (FKN) is expressed on activated endothelium and is associated with the development of atherosclerosis. The potential of FKN in mediating platelet adhesion beyond platelet activation remains unexplored to date. A flow-based adhesion assay was used to study the adhesion of platelets to immobilized FKN under physiologic flow conditions. Platelet adhesion to von Willebrand factor (VWF) was increased in the presence of FKN at 600 inverse seconds. Additional platelet adhesion to FKN coimmobilized with VWF was dependent on the FKN receptor CX3CR1 and activation of glycoprotein (GP) IIb/IIIa. The number of platelets rolling on VWF was likewise enhanced in the presence of FKN. The enhancement of rolling on FKN and VWF was insensitive to anti-CX3CR1 antibody but was fully inhibited by neutralizing GPIbα function. The extracellular domain of GPIbα was covalently coupled to fluorescent microspheres, and microsphere binding was significantly higher in the presence of FKN. Platelet adhesion to activated endothelium in vitro and to intact human arteries was substantially increased in an FKN-dependent manner. These data demonstrate that endothelial expressed FKN activates platelets via its cognate receptor CX3CR1, whereas platelet adhesion is predominantly mediated by GPIbα and independent of CX3CR1.


Subject(s)
Chemokine CX3CL1/physiology , Platelet Adhesiveness/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , Platelet Membrane Glycoproteins/physiology , von Willebrand Factor/physiology , Arteries/physiology , CX3C Chemokine Receptor 1 , Endothelial Cells/physiology , Hemorheology , Humans , In Vitro Techniques , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Receptors, Cytokine/antagonists & inhibitors , Receptors, Cytokine/physiology , Receptors, HIV/antagonists & inhibitors , Receptors, HIV/physiology
18.
Handb Exp Pharmacol ; (210): 287-309, 2012.
Article in English | MEDLINE | ID: mdl-22918736

ABSTRACT

The formation of platelet-rich thrombi, a critical step in the pathogenesis of atherothrombotic events, is a multistep process involving several components, among which von Willebrand Factor (VWF) plays a central role. Ruptured atherosclerotic plaques expose subendothelial matrix proteins which bind VWF that represents a bridge between the injured blood vessel and activated platelets, playing a crucial role in platelet adhesion and aggregation, especially in conditions of high-shear rate. Due to these peculiarities, the binding of VWF to GPIbα is an attractive drug target. Here we summarize the present knowledge on the different classes of drugs targeting the VWF-GPIb interaction and we give an account of their level of clinical development. In particular, the following compounds are discussed: AJW200, an IgG4 humanized monoclonal antibody against VWF-A1; 82D6A3, a monoclonal antibody against VWF-A3; ALX-0081 and ALX-0681, bivalent humanized nanobodies targeting the VWF-A1 domain; ARC1779 and its advanced formulation ARC15105, second-generation aptamers that bind the VWF-A1 domain; h6B4-Fab, a murine monoclonal antibody, and GPG-290, a recombinant chimeric protein, both directed against GPIbα.


Subject(s)
Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , von Willebrand Factor/antagonists & inhibitors , Animals , Aptamers, Nucleotide/therapeutic use , Humans , Platelet Activation , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/physiology , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Thrombosis/drug therapy , von Willebrand Factor/chemistry , von Willebrand Factor/physiology
19.
Pharmacotherapy ; 41(10): 828-836, 2021 10.
Article in English | MEDLINE | ID: mdl-34478577

ABSTRACT

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.


Subject(s)
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
20.
Sci Rep ; 11(1): 11663, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083615

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL