Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Arterioscler Thromb Vasc Biol ; 43(6): 1031-1040, 2023 06.
Article in English | MEDLINE | ID: mdl-37078286

ABSTRACT

BACKGROUND: Current clinical imaging of thromboembolic diseases often relies on indirect detection of thrombi, which may delay diagnosis and ultimately the institution of beneficial, potentially lifesaving treatment. Therefore, the development of targeting tools that facilitate the rapid, specific, and direct imaging of thrombi using molecular imaging is highly sought after. One potential molecular target is FXIIa (factor XIIa), which initiates the intrinsic coagulation pathway but also activates the kallikrein-kinin system, thereby initiating coagulation and inflammatory/immune responses. As FXII (factor XII) is dispensable for normal hemostasis, its activated form (FXIIa) represents an ideal molecular target for diagnostic and therapeutic approaches, the latter combining diagnosis/identification of thrombi and effective antithrombotic therapy. METHODS: We conjugated an FXIIa-specific antibody, 3F7, to a near-infrared (NIR) fluorophore and demonstrated binding to FeCl3-induced carotid thrombosis with 3-dimensional fluorescence emission computed tomography/computed tomography and 2-dimensional fluorescence imaging. We further demonstrated ex vivo imaging of thromboplastin-induced pulmonary embolism and detection of FXIIa in human thrombi produced in vitro. RESULTS: We demonstrated imaging of carotid thrombosis by fluorescence emission computed tomography/computed tomography and measured a significant fold increase in signal between healthy and control vessels from mice injected with 3F7-NIR compared with mice injected with nontargeted probe (P=0.002) ex vivo. In a model of pulmonary embolism, we measured increased NIR signal in lungs from mice injected with 3F7-NIR compared with mice injected with nontargeted probe (P=0.0008) and healthy lungs from mice injected with 3F7-NIR (P=0.021). CONCLUSIONS: Overall, we demonstrate that FXIIa targeting is highly suitable for the specific detection of venous and arterial thrombi. This approach will allow direct, specific, and early imaging of thrombosis in preclinical imaging modalities and may facilitate monitoring of antithrombotic treatment in vivo.


Subject(s)
Carotid Artery Thrombosis , Pulmonary Embolism , Thrombosis , Mice , Humans , Animals , Blood Coagulation , Thrombosis/diagnostic imaging , Factor XII/metabolism , Factor XIIa/metabolism , Molecular Imaging
2.
Transplant Proc ; 53(2): 612-619, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33279259

ABSTRACT

BACKGROUND: Use of donation after circulatory death (DCD) hearts is becoming more prevalent in cardiac transplantation. However, there is no standardized approach to myocardial preservation, and little data exists on ultrastructural changes in DCD hearts. We have previously identified increased proapoptotic and proinflammatory activity in brain dead donor (BDD) hearts that subsequently exhibit primary graft failure and lower levels in DCD left atrial tissue. This study further investigates these markers and correlates them with cardiac function in DCD hearts. METHODS: This prospective study used donor hearts deemed unsuitable for transplant after gaining institutional ethics approval; 11 human hearts were obtained from 5 DCD donors and 6 BDDs. All hearts were preserved by continuous microperfusion for 4 hours with a cold crystalloid solution and then were evaluated on a blood perfusion bench rig. After 4 hours perfusion and working assessment, tissues from all cardiac chambers were stored for later messenger RNA (mRNA) analysis for proapoptotic and proinflammatory markers. RESULTS: Significantly raised levels of caspase-1, BNIP3, and NADPH oxidase mRNA expression were identified in cardiac chambers from BDD hearts compared to DCD hearts, and these differences were exaggerated in older donors. In the pooled analysis, lower expression of caspase-1, NF-κB1, and BNIP3 mRNA correlated with developed pressure at 1 hour after reperfusion in the right ventricle, but not the left. CONCLUSION: Compared to BDD hearts, DCD hearts exhibit less stimulation of proapoptotic cascades and reactive oxygen species, potentially reducing their susceptibility to ischemic reperfusion injury.


Subject(s)
Brain Death , Death , Heart Transplantation , Heart , Transplants/pathology , Aged , Female , Humans , Male , Middle Aged , Prospective Studies , Tissue Donors
3.
Int J Mol Sci ; 20(12)2019 Jun 18.
Article in English | MEDLINE | ID: mdl-31216638

ABSTRACT

Thrombus formation in hemostasis or thrombotic disease is initiated by the rapid adhesion, activation, and aggregation of circulating platelets in flowing blood. At arterial or pathological shear rates, for example due to vascular stenosis or circulatory support devices, platelets may be exposed to highly pulsatile blood flow, while even under constant flow platelets are exposed to pulsation due to thrombus growth or changes in vessel geometry. The aim of this study is to investigate platelet thrombus formation dynamics within flow conditions consisting of either constant or variable shear. Human platelets in anticoagulated whole blood were exposed ex vivo to collagen type I-coated microchannels subjected to constant shear in straight channels or variable shear gradients using different stenosis geometries (50%, 70%, and 90% by area). Base wall shears between 1800 and 6600 s-1, and peak wall shears of 3700 to 29,000 s-1 within stenoses were investigated, representing arterial-pathological shear conditions. Computational flow-field simulations and stenosis platelet thrombi total volume, average volume, and surface coverage were analysed. Interestingly, shear gradients dramatically changed platelet thrombi formation compared to constant base shear alone. Such shear gradients extended the range of shear at which thrombi were formed, that is, platelets became hyperthrombotic within shear gradients. Furthermore, individual healthy donors displayed quantifiable differences in extent/formation of thrombi within shear gradients, with implications for future development and testing of antiplatelet agents. In conclusion, here, we demonstrate a specific contribution of blood flow shear gradients to thrombus formation, and provide a novel platform for platelet functional testing under shear conditions.


Subject(s)
Biomechanical Phenomena , Shear Strength , Thrombosis/etiology , Algorithms , Blood Coagulation , Blood Platelets/metabolism , Constriction, Pathologic , Humans , Models, Biological , Platelet Adhesiveness , Platelet Aggregation , Thrombosis/metabolism , Thrombosis/pathology
4.
Int Wound J ; 16(3): 788-792, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30864220

ABSTRACT

The overall quality of evidence of autologous platelet-rich plasma (PRP) for treating chronic wounds remains low. While further well-designed clinical studies are clearly required to convincingly demonstrate the efficacy of autologous PRP in improved healing of venous leg ulcers (VLUs) and other chronic wounds, there is also an increasing need to better define the underlying mechanisms of action and whether positive outcomes can be predicted based on the analysis of PRP. This brief review will discuss the current understanding of autologous PRP in VLUs and whether molecular evaluation of PRP at the time of collection could potentially be informative to clinical outcomes. Benefits of the autologous PRP treatment strategy include that PRP is easily accessible and is relatively inexpensive and safe. Better understanding of the mechanisms involved could improve treatment, enable supplementation, and/or lead to gains in product development. Analysis of PRP could also add value to future clinical trials on efficacy and potentially personalised treatment regimens.


Subject(s)
Blood Transfusion, Autologous/methods , Platelet-Rich Plasma , Varicose Ulcer/therapy , Wound Healing/physiology , Humans , Treatment Outcome
5.
Blood Rev ; 32(5): 349-360, 2018 09.
Article in English | MEDLINE | ID: mdl-29496356

ABSTRACT

Hematological cancer survivors are highly vulnerable to cardiometabolic complications impacting long-term health status, quality of life and survival. Elevated risk of diabetes and cardiovascular disease arises not only from the effects of the cancers themselves, but also from the toxic effects of cancer therapies, and deconditioning arising from reduced physical activity levels. Regular physical activity can circumvent or reverse adverse effects on the heart, skeletal muscle, vasculature and blood cells, through a combination of systemic and molecular mechanisms. We review the link between hematological cancers and cardiometabolic risk with a focus on adult survivors, including the contributing mechanisms and discuss the potential for physical activity interventions, which may act to oppose the negative effects of both physical deconditioning and therapies (conventional and targeted) on metabolic and growth signaling (kinase) pathways in the heart and beyond. In this context, we focus particularly on strategies targeting reducing and breaking up sedentary time and provide recommendations for future research.


Subject(s)
Heart Diseases/etiology , Heart Diseases/physiopathology , Hematologic Neoplasms/complications , Metabolic Diseases/etiology , Metabolic Diseases/metabolism , Animals , Combined Modality Therapy/adverse effects , Combined Modality Therapy/methods , Disease Management , Energy Metabolism , Heart Function Tests , Hematologic Neoplasms/therapy , Humans
6.
Redox Biol ; 14: 126-130, 2018 04.
Article in English | MEDLINE | ID: mdl-28888895

ABSTRACT

Reactive oxygen species (ROS) are generated within activated platelets and play an important role in regulating platelet responses to collagen and collagen-mediated thrombus formation. As a major collagen receptor, platelet-specific glycoprotein (GP)VI is a member of the immunoglobulin (Ig) superfamily, with two extracellular Ig domains, a mucin domain, a transmembrane domain and a cytoplasmic tail. GPVI forms a functional complex with the Fc receptor γ-chain (FcRγ) that, following receptor dimerization, signals via an intracellular immunoreceptor tyrosine-based activation motif (ITAM), leading to rapid activation of Src family kinase signaling pathways. Our previous studies demonstrated that an unpaired thiol in the cytoplasmic tail of GPVI undergoes rapid oxidation to form GPVI homodimers in response to ligand binding, indicating an oxidative submembranous environment in platelets after GPVI stimulation. Using a redox-sensitive fluorescent dye (H2DCF-DA) in a flow cytometric assay to measure changes in intracellular ROS, we showed generation of ROS downstream of GPVI consists of two distinct phases: an initial Syk-independent burst followed by additional Syk-dependent generation. In this review, we will discuss recent findings on the regulation of platelet function by ROS, focusing on GPVI-dependent platelet activation and thrombus formation.


Subject(s)
Blood Platelets/metabolism , Reactive Oxygen Species/metabolism , Thrombosis/pathology , Blood Platelets/cytology , Blood Platelets/drug effects , Collagen/metabolism , Humans , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/metabolism , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/toxicity , Thrombosis/drug therapy , Thrombosis/metabolism
7.
Diabetes ; 66(1): 7-13, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27999100

ABSTRACT

Adults with diabetes are 2-4 times more likely to suffer from heart disease or ischemic stroke than adults without diabetes, yet standard antiplatelet therapy, which is the cornerstone for primary and secondary prevention of cardiovascular disease, fails in many patients with diabetes. Three independent but often interrelated variables that contribute to platelet hyperreactivity-high blood glucose, oxidative stress, and elevated vascular shear forces-coexist in patients with diabetes, creating a perilous concurrence of risk factors for cardiovascular events. Recent research has focused attention on the platelet-specific collagen receptor glycoprotein VI (GPVI) as a potential antithrombotic target. Signaling events downstream of GPVI are influenced by hyperglycemia, oxidative stress, and shear stress. Importantly, drugs targeting these GPVI signaling pathways are already in existence. The potential to repurpose existing drugs is a high-gain strategy for yielding new antiplatelet agents and could have particular benefit in individuals with diabetes.


Subject(s)
Cardiovascular Diseases/blood , Cardiovascular Diseases/prevention & control , Diabetes Mellitus/blood , Diabetes Mellitus/metabolism , Platelet Membrane Glycoproteins/metabolism , Animals , Blood Glucose/drug effects , Blood Platelets/drug effects , Blood Platelets/physiology , Cardiovascular Diseases/metabolism , Diabetes Mellitus/physiopathology , Humans , Oxidative Stress/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Signal Transduction/drug effects
8.
Platelets ; 27(5): 402-9, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26943229

ABSTRACT

The physiological functions and cellular signaling of Class II phosphoinositide 3-kinases (PI3Ks) remain largely unknown. Platelets express two Class II PI3Ks: PI3KC2α and PI3KC2ß. PI3KC2α deficiency was recently reported to cause disruption of the internal membrane reserve structure of platelets (open canalicular system, OCS) that results in dysregulated platelet adhesion and impaired arterial thrombosis in vivo. Notably, these effects on platelets occurred despite normal agonist-induced 3-phosphorylated phosphoinositide (3-PPI) production and cellular activation in PI3KC2α-deficient platelets. However, the potential compensatory actions of PI3KC2ß in platelets have not yet been investigated. Here, we report the first mice deficient in both PI3KC2α and PI3KC2ß (no Class II PI3Ks in platelets) and reveal a nonredundant role for PI3KC2α in mouse platelet structure and function. Specifically, we show that the disrupted OCS and impaired thrombus stability observed in PI3KC2α-deficient platelets does not occur in PI3KC2ß-deficient platelets and is not exaggerated in platelets taken from mice deficient in both enzymes. Furthermore, detailed examination of 3-PPI production in platelets from this series of mice revealed no changes in either unactivated or activated platelets, including those with a complete lack of Class II PI3Ks. These findings indicate a nonredundant role for PI3KC2α in regulating platelet structure and function, and suggest that Class II PI3Ks do not significantly contribute to the acute agonist-induced production of 3-PPIs in these cells.


Subject(s)
Blood Platelets/metabolism , Class II Phosphatidylinositol 3-Kinases/deficiency , Thrombosis/blood , Thrombosis/genetics , Animals , Blood Platelets/ultrastructure , Class II Phosphatidylinositol 3-Kinases/genetics , Class II Phosphatidylinositol 3-Kinases/metabolism , Class III Phosphatidylinositol 3-Kinases/genetics , Class III Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Gene Expression Regulation , Mice , Mice, Knockout , Platelet Adhesiveness , Platelet Count , Platelet Function Tests
9.
Biochemistry ; 55(8): 1187-94, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26840909

ABSTRACT

The primary platelet collagen receptor, glycoprotein VI (GPVI), plays an important role in platelet activation and thrombosis. The ectodomain of human GPVI (sGPVI) is proteolytically shed from human platelets by a-disintegrin-and-metalloproteinase 10 (ADAM10). In this study, we used a novel ADAM10-sensitive fluorescence resonance energy transfer sensor to analyze ADAM10-mediated shedding of GPVI from human platelets in response to the exposure of GPVI ligands collagen-related peptide (10 µg/mL), collagen (10 µg/mL), and convulxin (0.1 µg/mL) to shear stress (1000-10000 s(-1), 5 min), or a generic activator of metalloproteinases, N-ethylmaleimide (NEM, 5 mM). Elevated shear, NEM, or ligand engagement of GPVI all induced shedding of GPVI, as detected by release of sGPVI; however, only shear or NEM significantly increased ADAM10 enzyme activity. ADAM10 activity was also detectable on the surface of thrombi formed on a collagen-coated surface under flow conditions. Our findings indicate different mechanisms regulate shear- and ligand-induced shedding and shear forces found within the vasculature can regulate ADAM10 activity.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Blood Platelets/cytology , Disintegrins/metabolism , Membrane Proteins/metabolism , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , ADAM10 Protein , Blood Coagulation , Blood Platelets/metabolism , Carrier Proteins/metabolism , Collagen/metabolism , Crotalid Venoms/metabolism , Humans , Lectins, C-Type/metabolism , Peptides/metabolism , Thrombosis/metabolism
10.
PLoS One ; 10(12): e0144860, 2015.
Article in English | MEDLINE | ID: mdl-26660525

ABSTRACT

Platelets can become activated in response to changes in flow-induced shear; however, the underlying molecular mechanisms are not clearly understood. Here we present new techniques for experimentally measuring the flow-induced shear rate experienced by platelets prior to adhering to a thrombus. We examined the dynamics of blood flow around experimentally grown thrombus geometries using a novel combination of experimental (ex vivo) and numerical (in silico) methodologies. Using a microcapillary system, platelet aggregate formation was analysed at elevated shear rates in the presence of coagulation inhibitors, where thrombus formation is predominantly platelet-dependent. These approaches permit the resolution and quantification of thrombus parameters at the scale of individual platelets (2 µm) in order to quantify real time thrombus development. Using our new techniques we can correlate the shear rate experienced by platelets with the extent of platelet adhesion and aggregation. The techniques presented offer the unique capacity to determine the flow properties for a temporally evolving thrombus field in real time.


Subject(s)
Blood Platelets/cytology , Models, Statistical , Stress, Mechanical , Thrombosis/blood , Anticoagulants/pharmacology , Blood Flow Velocity , Blood Platelets/drug effects , Cells, Cultured , Hirudins/pharmacology , Humans , Imaging, Three-Dimensional , Kinetics , Microscopy, Confocal , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects
11.
AIDS ; 29(17): 2309-13, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26544702

ABSTRACT

BACKGROUND: Current abacavir exposure has been reported to be associated with cardiovascular disease. Changes in platelet reactivity could plausibly explain the clinically observed pattern of association. OBJECTIVE: To determine if platelet reactivity changed following abacavir exposure and whether this effect was reversible on cessation of the drug. METHODS: In an open-label, interventional study abacavir, 600 mg daily, was added to a suppressive antiretroviral regimen in 20 adult HIV-positive men. Platelet function, estimated by the phosphorylated vasodilator-stimulated phosphoprotein (P-VASP) assay and through measurement of the expression and shedding of platelet-specific receptors, was assessed at baseline, following 15 days of abacavir and at completion of a 28-day washout period. RESULTS: The VASP-index decreased significantly from 79.1% [interquartile range (IQR) 47.8-87.6] to 32.6% (IQR -11.5-51.0) following 15 days of abacavir administration (P = 0.010), and returned to baseline levels following the washout period (day 43 =76.3%; IQR 40.7-92.3). There was no change in resting (prostaglandin E1 alone) P-VASP but a slight increase in P-VASP within stimulated platelets (prostaglandin E1 and adenosine diphosphate). Integrin ß3 levels decreased significantly [208.5 ng/ml (IQR 177.0-231.1) to 177.5 ng/ml (IQR 151.7-205) P < 0.001] and there was a nonsignificant trend towards decreased soluble glycoprotein VI levels [baseline; 72.5 ng/ml (95% CI 58.3-81.5) vs. day 15; 45.0 ng/ml (95% CI 33.0-98.2) P = 0.79]. CONCLUSION: Abacavir led to reversible changes in platelet function and structure. The clinical implications of these changes are uncertain; they may represent negative feedback mechanisms in response to an abacavir-associated prothrombotic state.


Subject(s)
Anti-HIV Agents/administration & dosage , Anti-HIV Agents/adverse effects , Dideoxynucleosides/administration & dosage , Dideoxynucleosides/adverse effects , HIV Infections/drug therapy , Platelet Activation/drug effects , Adult , Humans , Male , Middle Aged
12.
Redox Biol ; 6: 640-647, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26569550

ABSTRACT

BACKGROUND: Platelets are essential for maintaining haemostasis and play a key role in the pathogenesis of cardiovascular disease. Upon ligation of platelet receptors through subendothelial matrix proteins, intracellular reactive oxygen species (ROS) are generated, further amplifying the platelet activation response. Thrombin, a potent platelet activator, can signal through GPIbα and protease-activated receptor (PAR) 1 and PAR4 on human platelets, and recently has been implicated in the generation of ROS. While ROS are known to have key roles in intra-platelet signalling and subsequent platelet activation, the precise receptors and signalling pathways involved in thrombin-induced ROS generation have yet to be fully elucidated. OBJECTIVE: To investigate the relative contribution of platelet GPIbα and PARs to thrombin-induced reactive oxygen species (ROS) generation. METHODS AND RESULTS: Highly specific antagonists targeting PAR1 and PAR4, and the GPIbα-cleaving enzyme, Naja kaouthia (Nk) protease, were used in quantitative flow cytometry assays of thrombin-induced ROS production. Antagonists of PAR4 but not PAR1, inhibited thrombin-derived ROS generation. Removal of the GPIbα ligand binding region attenuated PAR4-induced and completely inhibited thrombin-induced ROS formation. Similarly, PAR4 deficiency in mice abolished thrombin-induced ROS generation. Additionally, GPIbα and PAR4-dependent ROS formation were shown to be mediated through focal adhesion kinase (FAK) and NADPH oxidase 1 (NOX1) proteins. CONCLUSIONS: Both GPIbα and PAR4 are required for thrombin-induced ROS formation, suggesting a novel functional cooperation between GPIbα and PAR4. Our study identifies a novel role for PAR4 in mediating thrombin-induced ROS production that was not shared by PAR1. This suggests an independent signalling pathway in platelet activation that may be targeted therapeutically.


Subject(s)
Blood Platelets/enzymology , Platelet Glycoprotein GPIb-IX Complex/physiology , Reactive Oxygen Species/metabolism , Receptors, Thrombin/physiology , Thrombin/physiology , Animals , COS Cells , Chlorocebus aethiops , Focal Adhesion Kinase 1/metabolism , Humans , Mice , NADPH Oxidase 1 , NADPH Oxidases/metabolism , Receptor, PAR-1/metabolism
13.
J Exp Med ; 212(2): 129-37, 2015 Feb 09.
Article in English | MEDLINE | ID: mdl-25646267

ABSTRACT

Nucleotide-based drug candidates such as antisense oligonucleotides, aptamers, immunoreceptor-activating nucleotides, or (anti)microRNAs hold great therapeutic promise for many human diseases. Phosphorothioate (PS) backbone modification of nucleotide-based drugs is common practice to protect these promising drug candidates from rapid degradation by plasma and intracellular nucleases. Effects of the changes in physicochemical properties associated with PS modification on platelets have not been elucidated so far. Here we report the unexpected binding of PS-modified oligonucleotides to platelets eliciting strong platelet activation, signaling, reactive oxygen species generation, adhesion, spreading, aggregation, and thrombus formation in vitro and in vivo. Mechanistically, the platelet-specific receptor glycoprotein VI (GPVI) mediates these platelet-activating effects. Notably, platelets from GPVI function-deficient patients do not exhibit binding of PS-modified oligonucleotides, and platelet activation is fully abolished. Our data demonstrate a novel, unexpected, PS backbone-dependent, platelet-activating effect of nucleotide-based drug candidates mediated by GPVI. This unforeseen effect should be considered in the ongoing development programs for the broad range of upcoming and promising DNA/RNA therapeutics.


Subject(s)
Blood Platelets/drug effects , Phosphorothioate Oligonucleotides/pharmacology , Platelet Activation/drug effects , Animals , Blood Platelets/metabolism , Disease Models, Animal , Humans , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , Mesenteric Vascular Occlusion/drug therapy , Mesenteric Vascular Occlusion/metabolism , Mice , Models, Molecular , Molecular Conformation , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Phosphorothioate Oligonucleotides/chemistry , Phosphorothioate Oligonucleotides/metabolism , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/metabolism , Protein Binding , Pulmonary Embolism/drug therapy , Pulmonary Embolism/metabolism , Pulmonary Embolism/pathology , RNA Interference , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
14.
Blood Rev ; 29(3): 179-89, 2015 May.
Article in English | MEDLINE | ID: mdl-25467961

ABSTRACT

Twenty-three years after the discovery of the first thrombin receptor, now known as protease-activated receptor 1 (PAR1), the first drug targeting this receptor is available for human use. The PAR1 inhibitor, vorapaxar (Zontivity, MSD), was recently approved by the FDA for use in the USA for the prevention of thrombotic cardiovascular events in patients with a history of myocardial infarction or peripheral artery disease. In this review, we detail the rationale, development, as well as the clinical significance and considerations of vorapaxar, the original PAR antagonist and the latest anti-platelet agent in the pharmaco-armoury against arterial thrombosis.


Subject(s)
Lactones/therapeutic use , Platelet Aggregation Inhibitors/therapeutic use , Pyridines/therapeutic use , Receptor, PAR-1/antagonists & inhibitors , Blood Platelets/drug effects , Blood Platelets/metabolism , Clinical Trials as Topic , Drug Approval , Humans , Lactones/pharmacology , Molecular Targeted Therapy , Platelet Aggregation Inhibitors/pharmacology , Pyridines/pharmacology , Thrombosis/blood , Thrombosis/drug therapy , Treatment Outcome , United States , United States Food and Drug Administration
15.
Thromb Haemost ; 112(4): 659-65, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25265341

ABSTRACT

In addition to playing a central role in normal haemostasis, platelets make important contributions to host inflammatory and immune responses to injury or infection. Under pathophysiological conditions where platelet function is not tightly controlled, platelets also play critical roles in pathogenic processes underlying cardiovascular disease, uncontrolled inflammation, coagulopathy and in tumour metastasis. Neutrophil extracellular traps (NETs) are webs of histone-modified nuclear material extruded from activated neutrophils during inflammatory responses and these degranulation events can be directly triggered by platelet/neutrophil engagement. Emerging research describes how NETs influence platelet function, particularly in the setting of infection and inflammation. Especially intriguing is the potential for platelet-driven coagulation to be modulated by NETs in plasma and interstitial spaces. These findings also reveal new perspectives related to improved therapy for venous thrombosis.


Subject(s)
Blood Platelets/physiology , Extracellular Traps/physiology , Infections/blood , Neutrophil Activation/physiology , Neutrophils/physiology , Animals , Blood Coagulation , Blood Platelets/metabolism , Cell Nucleus/metabolism , Hemostasis , Histones/chemistry , Humans , Inflammation , Leukocytes/metabolism , Neutrophils/metabolism , Venous Thrombosis/metabolism
16.
Thromb Haemost ; 112(4): 659-665, 2014 12.
Article in English | MEDLINE | ID: mdl-25144936

ABSTRACT

In addition to playing a central role in normal haemostasis, platelets make important contributions to host inflammatory and immune responses to injury or infection. Under pathophysiological conditions where platelet function is not tightly controlled, platelets also play critical roles in pathogenic processes underlying cardiovascular disease, uncontrolled inflammation, coagulopathy and in tumour metastasis. Neutrophil extracellular traps (NETs) are webs of histone-modified nuclear material extruded from activated neutrophils during inflammatory responses and these degranulation events can be directly triggered by platelet/neutrophil engagement. Emerging research describes how NETs influence platelet function, particularly in the setting of infection and inflammation. Especially intriguing is the potential for platelet-driven coagulation to be modulated by NETs in plasma and interstitial spaces. These findings also reveal new perspectives related to improved therapy for venous thrombosis.

17.
J Blood Med ; 5: 59-68, 2014.
Article in English | MEDLINE | ID: mdl-24899824

ABSTRACT

While platelet activation is essential to maintain blood vessel patency and minimize loss of blood upon injury, untimely or excessive activity can lead to unwanted platelet activation and aggregation. Resultant thrombosis has the potential to block blood vessels, causing myocardial infarction or stroke. To tackle this major cause of mortality, clinical therapies that target platelet responsiveness (antiplatelet therapy) can successfully reduce cardiovascular events, especially in people at higher risk; however, all current antiplatelet therapies carry an increased probability of bleeding. This review will evaluate new and emerging targets for antithrombotics, focusing particularly on platelet glycoprotein VI, as blockade or depletion of this platelet-specific receptor conveys benefits in experimental models of thrombosis and thromboinflammation without causing major bleeding complications.

18.
Acta Haematol ; 128(4): 233-41, 2012.
Article in English | MEDLINE | ID: mdl-22922528

ABSTRACT

INTRODUCTION: Ligands acting at the platelet collagen receptor, glycoprotein (GP)VI, induce intracellular FcRγ/Syk-dependent signaling pathways and Syk-dependent or Syk-independent generation of intracellular reactive oxygen species (ROS). Additional signaling-dependent or signaling-independent pathways lead to metalloproteinase-mediated shedding of GPVI. AIM: Analysis of platelet GPVI expression and signaling in a patient with a collagen-selective defect associated with myelodysplastic syndrome (MDS) uniquely demonstrates divergent pathways leading to ROS generation and Syk phosphorylation in human platelets. METHODS: Surface expression of GPVI and ligand-induced ROS generation was quantitated by flow cytometry. GPVI shedding and Syk phosphorylation were analyzed by Western blot. RESULTS: Despite platelet count/size and GPVI surface expression within normal ranges, platelet-rich plasma showed no aggregation in response to collagen or GPVI-selective agonist collagen-related peptide, but aggregated in response to other agonists, consistent with dysfunctional GPVI signaling. We observed rapid GPVI-dependent Syk-independent ROS generation and disulfide-dependent GPVI homodimerization, but not Syk-dependent ROS or ligand-induced shedding. Temporal analysis showed a gradual decline in platelet count and the appearance of ligand-induced phosphorylation of an ∼40-kDa Syk fragment. CONCLUSIONS: These studies show that GPVI ligation in platelets induces intracellular ROS production independent of either Syk activation or divergent pathways leading to platelet aggregation or ectodomain shedding.


Subject(s)
Myelodysplastic Syndromes/physiopathology , Platelet Membrane Glycoproteins/physiology , Reactive Oxygen Species/metabolism , Receptors, Collagen/physiology , Signal Transduction/physiology , Aged , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Protein-Tyrosine Kinases/metabolism , Syk Kinase
19.
Blood ; 119(18): 4311-20, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22431567

ABSTRACT

Ligand-induced ectodomain shedding of glycoprotein VI (GPVI) is a metalloproteinase-dependent event. We examined whether shear force, in the absence of GPVI ligand, was sufficient to induce shedding of GPVI. Human-citrated platelet-rich plasma or washed platelets were subjected to increasing shear rates in a cone-plate viscometer, and levels of intact and cleaved GPVI were examined by Western blot and ELISA. Pathophysiologic shear rates (3000-10 000 seconds(-1)) induced platelet aggregation and metalloproteinase-dependent appearance of soluble GPVI ectodomain, and GPVI platelet remnant. Shedding of GPVI continued after transient exposure to shear. Blockade of α(IIb)ß(3), GPIbα, or intracellular signaling inhibited shear-induced platelet aggregation but minimally affected shear-induced shedding of GPVI. Shear-induced GPVI shedding also occurred in platelet-rich plasma or washed platelets isolated from a von Willebrand disease type 3 patient with no detectable VWF, implying that shear-induced activation of platelet metalloproteinases can occur in the absence of GPVI and GPIbα ligands. Significantly elevated levels of sGPVI were observed in 10 patients with stable angina pectoris, with well-defined single vessel coronary artery disease and mean intracoronary shear estimates at 2935 seconds(-1) (peak shear, 19 224 seconds(-1)). Loss of GPVI in platelets exposed to shear has potential implications for the stability of a forming thrombus at arterial shear rates.


Subject(s)
Blood Platelets/chemistry , Coronary Stenosis/blood , Hemorheology , Platelet Membrane Glycoproteins/chemistry , Stress, Mechanical , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/physiology , ADAM10 Protein , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/physiology , Angina, Stable/blood , Blood Viscosity , Collagen/physiology , Coronary Stenosis/genetics , Dipeptides/pharmacology , Down-Regulation , Female , Humans , Hydroxamic Acids/pharmacology , Membrane Glycoproteins/physiology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Middle Aged , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Platelet Glycoprotein GPIb-IX Complex , Platelet Membrane Glycoproteins/biosynthesis , Platelet Membrane Glycoproteins/genetics , Platelet-Rich Plasma , Protein Structure, Tertiary , von Willebrand Disease, Type 3/blood
20.
J Infect Dis ; 204(9): 1403-12, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21921203

ABSTRACT

BACKGROUND: Recently, 3 new members of the genus Bocavirus, human bocavirus 2 (HBoV2), human bocavirus 3 (HBoV3), and human bocavirus 4 (HBoV4), were discovered. HBoV2-4 occur mainly in the gastrointestinal tract but rarely in the respiratory tract, contrary to human bocavirus 1 (HBoV1). METHODS: To investigate HBoV1-4 seroepidemiology among 195 adults and 252 wheezing children, we conducted immunoglobulin G (IgG) and immunoglobulin M (IgM) enzyme immunoassays with recombinant viruslike particles (VLPs). The children's sera were also tested for HBoV1-4 DNA by quantitative polymerase chain reaction (qPCR). RESULTS: Both rabbit and human antibodies to HBoV1-4 VP2 VLPs were found to be cross-reactive. After depletion of HBoV1-reactive antibodies, the HBoV2-4 approximate seroprevalences in adults were 34%, 15%, and 2% and in children aged 1-2 years 25%, 10%, and 5%, respectively. After depletion of HBoV2-4-reactive antibodies, the HBoV1 seroprevalence among adults decreased from 96% to 59%. No cross-reactivity of human anti-HBoV IgG was observed with bovine parvovirus1, parvovirus B19 or PARV4. No child was HBoV2-4 viremic. CONCLUSIONS: HBoV2-4 infect humans less commonly and elicit weaker B-cell responses than HBoV1. In our study HBoV2-4 did not seem to have a major etiological role in wheezing. Cross-reactivity with HBoV2-4 IgG partially accounts for the high HBoV1 seroprevalences previously reported. Correction for cross-reactivity is a prerequisite for VLP-based HBoV seroepidemiology.


Subject(s)
Antibodies, Viral/blood , Human bocavirus/immunology , Parvoviridae Infections/epidemiology , Adolescent , Adult , Child , Child, Preschool , Cross Reactions , DNA, Viral/blood , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , Infant , Infant, Newborn , Parvoviridae Infections/virology , Polymerase Chain Reaction , Seroepidemiologic Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...