thromboxane-a2 has been researched along with Thrombosis* in 210 studies
77 review(s) available for thromboxane-a2 and Thrombosis
Article | Year |
---|---|
The Effect of Aspirin on the Prevention of Pro-thrombotic States in Hospitalized COVID-19 Patients: Systematic Review.
Thromboembolic events are one of the important complications in COVID-19 patients, especially in severe cases. Aspirin affects platelet function by irreversibly inhibiting cyclooxygenase activity, reducing the risk of thrombosis. The current systematic review aimed to evaluate aspirin's effectiveness in preventing pro-thrombotic states in COVID-19 hospitalized patients.. The systematic search was done in PubMed/Medline, EMBASE, and Medrxiv until September 27, 2021. The following keywords were used: "COVID-19", "SARS-CoV-2", "2019 Novel Coronavirus", "Aspirin," and "Acetylsalicylic Acid.". Twelve studies were included. In COVID-19 patients, aspirin can reduce CRP, IL-6 levels, and platelet aggregation by inhibiting thromboxane A2. It can also improve antiviral immunity by hindering the biosynthesis of prostaglandins and lipoxin. Eight out of twelve articles indicated that aspirin provided a beneficial effect on COVID-19. Most studies consider lowered mechanical ventilation needs, ICU admission, illness severity, overt thrombosis, and clinical outcomes in COVID-19 patients receiving aspirin.. Aspirin as an antiplatelet and anti-inflammatory agent may reduce the mortality rates in hospitalized patients with severe COVID-19. Further observational studies are necessary to determine the effect of aspirin on the prevention of pro-thrombotic states in hospitalized COVID- 19 patients. Topics: Antiviral Agents; Aspirin; COVID-19 Drug Treatment; Humans; Interleukin-6; Lipoxins; Platelet Aggregation Inhibitors; Prostaglandin-Endoperoxide Synthases; Prostaglandins; SARS-CoV-2; Thrombosis; Thromboxane A2 | 2022 |
The key contribution of platelet and vascular arachidonic acid metabolism to the pathophysiology of atherothrombosis.
Arachidonic acid is one of the most abundant and ubiquitous ω-6 polyunsaturated fatty acid, present in esterified form in the membrane phospholipids of all mammalian cells and released from phospholipids by several phospholipases in response to various activating or inhibitory stimuli. Arachidonic acid is the precursor of a large number of enzymatically and non-enzymatically derived, biologically active autacoids, including prostaglandins (PGs), thromboxane (TX) A2, leukotrienes, and epoxyeicosatetraenoic acids (collectively called eicosanoids), endocannabinoids and isoprostanes, respectively. Eicosanoids are local modulators of the physiological functions and pathophysiological roles of blood vessels and platelets. For example, the importance of cyclooxygenase (COX)-1-derived TXA2 from activated platelets in contributing to primary haemostasis and atherothrombosis is demonstrated in animal and human models by the bleeding complications and cardioprotective effects associated with low-dose aspirin, a selective inhibitor of platelet COX-1. The relevance of vascular COX-2-derived prostacyclin (PGI2) in endothelial thromboresistance and atheroprotection is clearly shown by animal and human models and by the adverse cardiovascular effects exerted by COX-2 inhibitors in humans. A vast array of arachidonic acid-transforming enzymes, downstream synthases and isomerases, transmembrane receptors, and specificity in their tissue expression make arachidonic acid metabolism a fine-tuning system of vascular health and disease. Its pharmacological regulation is central in human cardiovascular diseases, as demonstrated by biochemical measurements and intervention trials. Topics: Animals; Arachidonic Acid; Atherosclerosis; Biomarkers; Blood Platelets; Cyclooxygenase Inhibitors; Endothelium, Vascular; Epoprostenol; Fibrinolytic Agents; Humans; Leukotriene Antagonists; Platelet Aggregation Inhibitors; Signal Transduction; Thrombosis; Thromboxane A2 | 2021 |
Mouse laser injury models: variations on a theme.
A confluence of technological advances in genetic manipulation and molecular-based fluorescence imaging has led to the widespread adoption of laser injury models to study hemostasis and thrombosis in mice. In all animal models of hemostasis and thrombosis, detailing the nature of experimentally induced vascular injury is paramount in enabling appropriate interpretation of experimental results. A careful appraisal of the literature shows that direct laser-induced injury can result in variable degrees of vascular damage. This review will compare and contrast models of laser injury utilized in the field, with an emphasis on the mechanism and extent of injury, the use of laser injury in different vascular beds and the molecular mechanisms regulating the response to injury. All of these topics will be discussed in the context of how distinct applications of laser injury models may be viewed as representing thrombosis and/or hemostasis. Topics: Animals; Disease Models, Animal; Endothelial Cells; Femoral Artery; Hemostasis; Humans; Intravital Microscopy; Laser Therapy; Mice; Platelet Activation; Saphenous Vein; Thrombosis; Thromboxane A2; Vascular System Injuries | 2020 |
Platelet signaling: a complex interplay between inhibitory and activatory networks.
The role of platelets in hemostasis and thrombosis is dependent on a complex balance of activatory and inhibitory signaling pathways. Inhibitory signals released from the healthy vasculature suppress platelet activation in the absence of platelet receptor agonists. Activatory signals present at a site of injury initiate platelet activation and thrombus formation; subsequently, endogenous negative signaling regulators dampen activatory signals to control thrombus growth. Understanding the complex interplay between activatory and inhibitory signaling networks is an emerging challenge in the study of platelet biology, and necessitates a systematic approach to utilize experimental data effectively. In this review, we will explore the key points of platelet regulation and signaling that maintain platelets in a resting state, mediate activation to elicit thrombus formation, or provide negative feedback. Platelet signaling will be described in terms of key signaling molecules that are common to the pathways activated by platelet agonists and can be described as regulatory nodes for both positive and negative regulators. Topics: Animals; Antigens, CD; Apyrase; Cell Adhesion Molecules; Cyclic GMP-Dependent Protein Kinases; Gene Expression Regulation; Hemostasis; Humans; Integrins; Models, Biological; Platelet Activation; Receptors, Cell Surface; Signal Transduction; Thrombosis; Thromboxane A2; Type C Phospholipases | 2016 |
Oxidative stress-related mechanisms affecting response to aspirin in diabetes mellitus.
Type 2 diabetes mellitus (T2DM) is a major cardiovascular risk factor. Persistent platelet activation plays a key role in atherothrombosis in T2DM. However, current antiplatelet treatments appear less effective in T2DM patients vs nondiabetics at similar risk. A large body of evidence supports the contention that oxidative stress, which characterizes DM, may be responsible, at least in part, for less-than-expected response to aspirin, with multiple mechanisms acting at several levels. This review discusses the pathophysiological mechanisms related to oxidative stress and contributing to suboptimal aspirin action or responsiveness. These include: (1) mechanisms counteracting the antiplatelet effect of aspirin, such as reduced platelet sensitivity to the antiaggregating effects of NO, due to high-glucose-mediated oxidative stress; (2) mechanisms interfering with COX acetylation especially at the platelet level, e.g., lipid hydroperoxide-dependent impaired acetylating effects of aspirin; (3) mechanisms favoring platelet priming (lipid hydroperoxides) or activation (F2-isoprostanes, acting as partial agonists of thromboxane receptor), or aldose-reductase pathway-mediated oxidative stress, leading to enhanced platelet thromboxane A2 generation or thromboxane receptor activation; (4) mechanisms favoring platelet recruitment, such as aspirin-induced platelet isoprostane formation; (5) modulation of megakaryocyte generation and thrombopoiesis by oxidative HO-1 inhibition; and (6) aspirin-iron interactions, eventually resulting in impaired pharmacological activity of aspirin, lipoperoxide burden, and enhanced generation of hydroxyl radicals capable of promoting protein kinase C activation and platelet aggregation. Acknowledgment of oxidative stress as a major contributor, not only of vascular complications, but also of suboptimal response to antiplatelet agents in T2DM, may open the way to designing and testing novel antithrombotic strategies, specifically targeting oxidative stress-mediated mechanisms of less-than-expected response to aspirin. Topics: Aldehyde Reductase; Aspirin; Blood Platelets; Diabetes Mellitus, Type 2; F2-Isoprostanes; Glucose; Glycosylation; Humans; Lipid Peroxides; Nitric Oxide; Oxidative Stress; Peroxynitrous Acid; Platelet Activation; Platelet Aggregation Inhibitors; Thrombosis; Thromboxane A2 | 2015 |
Importance of platelet aggregation in patients with end-stage renal disease.
The exact etiology of the conflicting hemostatic disorder in the advanced stage of chronic renal disease, i.e. prothrombotic versus bleeding tendency, is not completely understood. Abnormal platelet function in patients with renal failure is not caused by high concentrations of urea, although the presence of fibrinogen fragments may prevent binding of normal fibrinogen and formation of platelet aggregates. Hemostatic abnormalities in end-stage kidney disease may be affected, to some extent, by the choice of renal replacement therapy. Patients on hemodialysis have an increased risk of thrombotic events, primarily due to the release of thromboxane A2 and adenosine diphosphate into the circulation, as well as platelet degranulation. Some activation of platelets occurs due to the exposure of blood to the roller pump segment, but microbubbles may also play a role. Renal transplantation is the treatment of choice for patients with end-stage renal disease. Immunosuppressive therapy is associated with an increased risk of thromboembolic complications. Additional research is required to identify the potential benefits of different immunosuppressive therapies in relation to platelet aggregation, keeping in mind the long- term need for immunosuppression in renal transplant patients. Topics: Adenosine Diphosphate; Blood Platelet Disorders; Humans; Kidney Failure, Chronic; Kidney Transplantation; Platelet Aggregation; Renal Dialysis; Renal Replacement Therapy; Risk Factors; Thromboembolism; Thrombosis; Thromboxane A2 | 2013 |
Pharmacodynamic properties of antiplatelet agents: current knowledge and future perspectives.
Platelets play an important role in atherothrombotic disease. The currently available antiplatelet drugs target key steps of platelet activation including thromboxane A(2) synthesis, ADP-mediated signaling, and glycoprotein IIb/IIIa-mediated platelet aggregation. The improvement of our understanding on the pharmacokinetic and pharmacodynamic characteristics of these drugs enables the tailoring of the most appropriate anti-thrombotic therapy to the individual patient and risk situation in the daily clinical practice. However, current antiplatelet therapies are associated with increased bleeding risk. Thus, further research on platelet functions may give rise to numerous new antiplatelet agents with high anti-thrombotic efficiency and low adverse hemorrhagic side effects. Topics: Blood Platelets; Clinical Trials as Topic; Humans; Platelet Aggregation; Platelet Aggregation Inhibitors; Platelet Glycoprotein GPIIb-IIIa Complex; Thrombosis; Thromboxane A2 | 2012 |
The role of platelets in athero-thrombotic events.
The crucial role of platelets in primary hemostasis and repair of injured endothelium is well established, as is their role in atherothrombosis. No other single cell type is responsible for as much morbidity and mortality, since death from ischemic heart disease or stroke is by far the leading cause of death worldwide. There is no doubt that our understanding of atherothrombosis has guided current antithrombotic strategies that have dramatically reduced ischemic complications and cardiovascular mortality within the last decades. Yet the rate of ischemic complications after optimal revascularization therapy remains disappointingly high. There is still a strong need for new and smart antiplatelet drugs. The ideal antithrombotic drug would spare physiological platelet function, hemostasis and vascular repair in order to avoid bleeding complications, but would exclusively target the pathological atherothrombotic process. As platelet activity might be determined early in the bone marrow, this review starts with insights into the birth of platelets, describes the essential and primary role of platelets in hemostasis with new evidence in signaling cascades, and closes with the deleterious role of platelets in atherosclerosis and atherothrombosis, with a focus on acute coronary syndromes. Topics: Adenosine Diphosphate; Animals; Atherosclerosis; Blood Platelets; Coronary Artery Disease; Coronary Thrombosis; Fibrinolytic Agents; Hemostasis; Humans; Plaque, Atherosclerotic; Platelet Activation; Platelet Aggregation Inhibitors; Thrombin; Thrombosis; Thromboxane A2 | 2012 |
Anti-platelet therapy: cyclo-oxygenase inhibition and the use of aspirin with particular regard to dual anti-platelet therapy.
Aspirin and P2Y(12) antagonists are commonly used anti-platelet agents. Aspirin produces its effects through inhibition of thromboxane A(2) (TXA(2)) production, while P2Y(12) antagonists attenuate the secondary responses to ADP released by activated platelets. The anti-platelet effects of aspirin and a P2Y(12) antagonist are often considered to be separately additive. However, there is evidence of an overlap in effects, in that a high level of P2Y(12) receptor inhibition can blunt TXA(2) receptor signalling in platelets and reduce platelet production of TXA(2). Against this background, the addition of aspirin, particularly at higher doses, could cause significant reductions in the production of prostanoids in other tissues, e.g. prostaglandin I(2) from the blood vessel wall. This review summarizes the data from clinical studies in which dose-dependent effects of aspirin on prostanoid production have been evaluated by both plasma and urinary measures. It also addresses the biology underlying the cardiovascular effects of aspirin and its influences upon prostanoid production throughout the body. The review then considers whether, in the presence of newer, more refined P2Y(12) receptor antagonists, aspirin may offer less benefit than might have been predicted from earlier clinical trials using more variable P2Y(12) antagonists. The possibility is reflected upon, that when combined with a high level of P2Y(12) blockade the net effect of higher doses of aspirin could be removal of anti-thrombotic and vasodilating prostanoids and so a lessening of the anti-thrombotic effectiveness of the treatment. Topics: Aspirin; Blood Platelets; Clinical Trials as Topic; Cyclooxygenase Inhibitors; Dose-Response Relationship, Drug; Drug Interactions; Drug Therapy, Combination; Humans; Platelet Aggregation; Platelet Aggregation Inhibitors; Prostaglandins; Purinergic P2Y Receptor Antagonists; Thrombosis; Thromboxane A2 | 2011 |
Antiplatelet drugs--do we need new options? With a reappraisal of direct thromboxane inhibitors.
This review describes the current status of antiplatelet therapy in prevention of cardiovascular events of an atherothrombotic nature. The efficacy of aspirin clearly outweighs bleeding risk in secondary prevention, with the relevant exception of patients with peripheral arterial disease (PAD). In trials of primary prevention, aspirin has a limited advantage, which is challenged by the risk of major bleeding. A typical example is primary prevention in type 2 diabetes mellitus, in which a number of trials and a recent meta-analysis have confirmed these limitations. In various settings, clopidogrel has been shown to be marginally more effective than aspirin. Despite a non-negligible bleeding risk, the combination of aspirin-clopidogrel has provided satisfactory results in conditions at high thrombotic risk but rather disappointing results in the long-term treatment of chronic stable cardiovascular disease. The combination of aspirin-dipyridamole was shown to be superior to aspirin alone and equivalent to clopidogrel alone for secondary prevention in cerebrovascular patients. Limitations in the efficacy of antiplatelet agents are partly inherent in their mechanism of action and should not be considered simply as 'treatment failures'. Among other factors, individual variability of response to antiplatelet drugs also plays a meaningful role. Variability of response and 'resistance' may result from drug interactions, baseline and residual platelet hyperactivity, increased platelet turnover, pharmacogenetic factors and others. Poor biological response to aspirin and/or clopidogrel is also frequent in clinical settings such as diabetes, obesity and acute coronary syndromes. The correlation between biological resistance and impaired clinical efficacy of aspirin, and especially clopidogrel, is currently accepted, although with limitations due to the different methods used to assess platelet response. Indeed, the concept of individual 'tailoring' of antiplatelet regimens on the basis of previous laboratory or 'point of care' platelet function tests has been validated in a number of recent trials. The search for and validation of new antiplatelet agents with already known, or totally new, mechanisms of action have also been undertaken with increasing eagerness. Among new adenosine diphosphate receptor antagonists, prasugrel is already registered, and ticagrelor and cangrelor are being developed. New mechanisms being explored are blockade of thrombin-induced platelet Topics: Atherosclerosis; Clinical Trials as Topic; Diabetes Mellitus, Type 2; Drug Therapy, Combination; Humans; Platelet Aggregation Inhibitors; Thrombosis; Thromboxane A2; Treatment Outcome | 2010 |
Mechanisms of action, physiological effects, and complications of hypothermia.
Mild to moderate hypothermia (32-35 degrees C) is the first treatment with proven efficacy for postischemic neurological injury. In recent years important insights have been gained into the mechanisms underlying hypothermia's protective effects; in addition, physiological and pathophysiological changes associated with cooling have become better understood.. To discuss hypothermia's mechanisms of action, to review (patho)physiological changes associated with cooling, and to discuss potential side effects.. Review article.. None.. A myriad of destructive processes unfold in injured tissue following ischemia-reperfusion. These include excitotoxicty, neuroinflammation, apoptosis, free radical production, seizure activity, blood-brain barrier disruption, blood vessel leakage, cerebral thermopooling, and numerous others. The severity of this destructive cascade determines whether injured cells will survive or die. Hypothermia can inhibit or mitigate all of these mechanisms, while stimulating protective systems such as early gene activation. Hypothermia is also effective in mitigating intracranial hypertension and reducing brain edema. Side effects include immunosuppression with increased infection risk, cold diuresis and hypovolemia, electrolyte disorders, insulin resistance, impaired drug clearance, and mild coagulopathy. Targeted interventions are required to effectively manage these side effects. Hypothermia does not decrease myocardial contractility or induce hypotension if hypovolemia is corrected, and preliminary evidence suggests that it can be safely used in patients with cardiac shock. Cardiac output will decrease due to hypothermia-induced bradycardia, but given that metabolic rate also decreases the balance between supply and demand, is usually maintained or improved. In contrast to deep hypothermia ( Topics: Acidosis; Apoptosis; Body Temperature Regulation; Brain Edema; Brain Ischemia; Calpain; Critical Care; Epilepsy; Free Radicals; Genes, Immediate-Early; Humans; Hypothermia, Induced; Infections; Inflammation; Ion Pumps; Mitochondria; Reperfusion Injury; Thrombosis; Thromboxane A2 | 2009 |
COX-1 and vascular disease.
Since the discovery of the pivotal role of cyclooxygenase (COX) in the metabolism of arachidonic acid, vascular biologists have been confronted with the duality of this system. Indeed, one substrate (arachidonic acid) transformed by one enzyme (COX) yields end products (endoperoxides) that exist only very briefly before being metabolized to more stable prostanoids by a set of specific downstream synthases that were initially believed to be tissue specific. For instance, platelets contain mainly the synthase that produces thromboxane A(2) (a potent proaggregatory and vasoconstrictor substance), whereas endothelial cells contain mainly the enzyme that generates prostacyclin (an equally potent antiaggregatory and vasodilator substance). The overproduction of thromboxane A(2) by platelets leads to thrombosis; endothelial cells resist vascular occlusion by producing prostacyclin. This duality of the metabolism of arachidonic acid has dominated our thinking about atherothrombosis for decades, and rightfully still does. As scientific understanding progressed, it became evident that two isoforms of COX exist: COX-1 and COX-2. COX-1 was initially considered to be the "good," constitutive isoform, whereas COX-2 appeared to be mainly a "bad" inducible enzyme involved in inflammatory responses. However, more recently, the unexpected events resulting from the widespread use of selective COX-2 inhibitors has suggested that, from a cardiovascular point of view, the products of COX-2 exert a protective role and that this isoform cannot necessarily be regarded as "bad." Likewise, evidence has emerged that initiation of the metabolism of arachidonic acid by COX-1 is not necessarily a "good" thing in terms of vascular protection. This brief review focuses on the potential contribution of endothelial COX-1 to vascular dysfunction. It is based on a number of review articles, to which the reader will be referred in order to identify the original references to the statements made; these references are not cited here because of space limitations. Topics: Aging; Animals; Blood Platelets; Cyclooxygenase 1; Cyclooxygenase 2; Diabetes Mellitus; Endothelium, Vascular; Humans; Hypertension; Nitric Oxide Synthase; Platelet Aggregation Inhibitors; Prostaglandins; Thrombosis; Thromboxane A2; Vasoconstriction; Vasodilation | 2009 |
Effect of pharmaceutical interventions targeting thromboxane receptors and thromboxane synthase in cardiovascular and renal diseases.
The present review focuses on the roles of thromboxane A2 (TxA2) in arterial thrombosis, atherogenesis, vascular stent-related ischemic events and renal proteinuria. Particular emphasis is laid on therapeutic interventions targeting the TxA2 (TP) receptors and TxA2 synthase (TS), including dual TP-receptor antagonists and TS inhibitors. Their significant inhibitory efficacies on arterial thrombogenesis, atherogenesis, restenosis after stent placement, vasoconstriction and proteinuria indicate novel and improved treatments for cardiovascular and selected renal diseases. New therapeutic interventions of the TxA2 pathway may also be beneficial for patients with poor biological antiplatelet drug response, for example, to aspirin and/or clopidogrel. These new TP/TS agents offer novel improved treatments to efficiently and simultaneously interfere with thrombogenesis and atherogenesis, and to enlarge the existing panel of platelet inhibitors for efficient prophylaxis and treatment of arterial thrombosis and renal proteinuria. Topics: Aspirin; Cardiovascular Diseases; Clopidogrel; Coronary Artery Disease; Coronary Restenosis; Humans; Kidney Diseases; Platelet Aggregation Inhibitors; Proteinuria; Receptors, Thrombin; Thrombosis; Thromboxane A2; Thromboxane-A Synthase; Ticlopidine | 2009 |
Antiplatelet drugs.
Platelets play a major role in thromboembolic diseases, and so antiplatelet therapy remains crucial in treatment and prophylaxis. Upon vascular injury, platelets rapidly adhere to the exposed subendothelial matrix, after which they become activated, resulting in the recruitment of additional platelets from the circulation to eventually form a stable arterial platelet plug. Although controlled plug formation is desired for the prevention of excessive blood loss and for promoting wound healing, several pathological conditions may result in the formation of occlusive thrombi leading to severe clinical complications, including myocardial infarction and ischaemic stroke. Many antiplatelet approaches have been investigated, interfering with one or more of the different stages in thrombus formation. This review discusses antiplatelet agents that interfere with the three principal phases in thrombus formation: platelet adhesion, amplification of platelet activation and platelet aggregation. For each stage, novel experimental targets and clinically established antiplatelet strategies will be reviewed. Limitations and possible benefits will be discussed for each target. Topics: Blood Platelets; Humans; Platelet Adhesiveness; Platelet Aggregation; Platelet Aggregation Inhibitors; Platelet Membrane Glycoproteins; Receptors, Purinergic; Serotonin; Thrombin; Thrombosis; Thromboxane A2 | 2008 |
Variable platelet response to aspirin and clopidogrel in atherothrombotic disease.
Topics: Adenosine Diphosphate; Anti-Inflammatory Agents, Non-Steroidal; Aspirin; Atherosclerosis; Biomarkers; Biotransformation; Blood Coagulation Tests; Blood Platelets; Cardiovascular Diseases; Clopidogrel; Cyclooxygenase 1; Cyclooxygenase Inhibitors; Drug Interactions; Drug Resistance; Hemorrhage; Humans; Platelet Activation; Platelet Aggregation Inhibitors; Platelet Function Tests; Prospective Studies; Randomized Controlled Trials as Topic; Receptors, Purinergic P2; Receptors, Purinergic P2Y1; Receptors, Purinergic P2Y12; Thrombosis; Thromboxane A2; Thromboxane B2; Ticlopidine; Treatment Failure | 2007 |
Clinical implications of aspirin resistance.
Aspirin (acetylsalicylic acid) is one of the main therapeutic medications used in the prevention of thromboembolic vascular events. Aspirin exhibits its antiplatelet action by irreversibly inhibiting platelet cyclooxygenase-1 enzyme, thus preventing the production of thromboxane A2 (TXA2). Aspirin resistance, as measured in vitro, is the inability of aspirin to reduce platelet activation and aggregation by failure to suppress the platelet production of TXA2. Laboratory tests of platelet TXA2 production or platelet function dependent on TXA2 can detect aspirin resistance in vitro. The clinical implication of this laboratory definition has not yet been elucidated via prospective trials that have controlled for confounders, such as hypertension, diabetes and dyslipidemia. Large meta-analyses have found low-dose aspirin to be as effective as high-dose aspirin in preventing vascular events, making a dose-dependent improvement in laboratory response clinically irrelevant. Possible causes of aspirin resistance include poor compliance, inadequate dose, drug interactions, genetic polymorphisms of cyclooxygenase-1, increased platelet turnover and upregulation of non-platelet pathways of thromboxane production. However, there is currently no standardized approach to the diagnosis and no proven effective treatment for aspirin resistance. Further research exploring the mechanisms of aspirin resistance is needed in order to better define aspirin resistance, as well as to develop a standardized laboratory test that is specific and reliable, and can correlate with the clinical risk of vascular events. The intent of this paper is to review the literature discussing possible mechanisms, diagnostic testing and clinical trials of aspirin resistance and to discuss its clinical relevance as it pertains to cerebrovascular and cardiovascular disease. Topics: Aspirin; Cyclooxygenase 1; Drug Resistance; Enzyme Inhibitors; Heart Diseases; Humans; Ischemia; Platelet Activation; Platelet Aggregation Inhibitors; Platelet Function Tests; Thrombosis; Thromboxane A2 | 2007 |
Problem of the atherothrombotic potential of non-steroidal anti-inflammatory drugs.
Treatment of pain in rheumatoid arthritis must take into account the gastrointestinal and cardiovascular risk of individual patients. Adequate results are not yet available, and until they are, treatment recommendations must take into account, not only the more favourable gastrointestinal risk profile of selective COX-2 inhibitors, but also the potential atherothrombotic risk of any NSAID or selective COX-2 inhibitor treatment. Topics: Anti-Inflammatory Agents, Non-Steroidal; Atherosclerosis; Blood Platelets; Cyclooxygenase 2 Inhibitors; Cyclooxygenase Inhibitors; Humans; Thrombosis; Thromboxane A2 | 2006 |
Diet and aging: bearing on thrombosis and hemostasis.
Diet is one of the environmental factors that influences thrombosis and hemostasis. Macronutrients, micronutrients, and other bioactive food components alter the predisposition to thrombosis. The type and amount of dietary fat has been shown to alter thromboxane A2 production and platelet aggregation, bleeding time, factor VII, fibrinogen, tissue plasminogen activator (t-PA) and plasminogen activator inhibitor 1 (PAI-1). Both epidemiological studies and clinical trials indicate that the very long chain n-3 fatty acids lower thrombotic tendency and risk of heart disease. Other polyunsaturated fats and monounsaturated fat appear to have antithrombotic properties, but further studies are indicated. Hypercaloric diets and those with high glycemic loads are associated with elevations of PAI-1. Moderate consumption of alcohol is associated with decreased platelet aggregation. Low intakes of folate, vitamin B12, and vitamin B6 predispose to hyperhomocysteinemia, and the benefits of supplementation in decreasing vascular disease are under investigation. In a limited number of clinical and laboratory studies, vitamin E has been shown to decrease platelet aggregation and the concentration of PAI-1. Flavonoids and isoflavones appear to inhibit platelet aggregation at pharmacologic concentrations only. Nutritional status frequently declines with aging and may exacerbate the already increased risk for thrombosis. Diet presents an interesting area for research into thrombophilia, but additional work is indicated before specific recommendations are made. Topics: Aging; Alcohols; Bleeding Time; Carbohydrate Metabolism; Diet; Dietary Fats; Factor VII; Fatty Acids; Fatty Acids, Unsaturated; Fibrinogen; Hemostasis; Humans; Isoflavones; Niacin; Nutritional Physiological Phenomena; Plasminogen Activator Inhibitor 1; Platelet Aggregation; Risk; Thrombophilia; Thrombosis; Thromboxane A2; Tissue Plasminogen Activator | 2005 |
A critical appraisal of the phenomenon of aspirin resistance.
Aspirin is the mainstay antiplatelet treatment in patients with high risk of cardiovascular atherothrombotic events, and its beneficial effect is documented in several clinical trials. Nevertheless, the effectiveness of aspirin has been questioned by the emergence of the concept of 'aspirin resistance' (AR). This phenomenon, although lacking a precise definition, covers the fact that some patients do not exhibit the expected platelet inhibition by use of various techniques for measuring platelet function. In this critical review, we evaluate the methods used for measuring AR. We will discuss the available data regarding the prevalence and the clinical importance of the phenomenon. Finally, the potential mechanisms underlying AR are considered. Topics: Aspirin; Atherosclerosis; Cardiovascular Diseases; Drug Resistance; Humans; Platelet Aggregation; Platelet Aggregation Inhibitors; Platelet Function Tests; Thrombosis; Thromboxane A2; Thromboxane B2 | 2005 |
[Thromboxane A2 (TXA2), thromboxane B2 (TXB2)].
Topics: Asthma; Biomarkers; Cardiovascular Diseases; Humans; Immunoenzyme Techniques; Ischemia; Kidney Failure, Chronic; Radioimmunoassay; Reference Values; Specimen Handling; Thrombosis; Thromboxane A2; Thromboxane B2 | 2005 |
Low-dose aspirin for the prevention of atherothrombosis.
Topics: Arteriosclerosis; Aspirin; Cyclooxygenase 1; Cyclooxygenase Inhibitors; Drug Interactions; Drug Resistance; Gastrointestinal Hemorrhage; Humans; Male; Myocardial Infarction; Platelet Aggregation Inhibitors; Risk; Stroke; Thrombosis; Thromboxane A2 | 2005 |
[Icosapentanoic acid (EPA), docosahexanoic acid (DHA)].
Topics: Arrhythmias, Cardiac; Arteriosclerosis; Biomarkers; Cholesterol, VLDL; Coronary Restenosis; Docosahexaenoic Acids; Eicosapentaenoic Acid; Humans; Hyperlipidemias; Hypertension; Liver; Reference Values; Thrombosis; Thromboxane A2 | 2004 |
Platelet-collagen interaction: is GPVI the central receptor?
At sites of vascular injury, platelets come into contact with subendothelial collagen, which triggers their activation and the formation of a hemostatic plug. Besides glycoprotein Ib (GPIb) and alphaIIbbeta3 integrin, which indirectly interact with collagen via von Willebrand factor (VWF), several collagen receptors have been identified on platelets, most notably alpha2beta1 integrin and the immunoglobulin (Ig) superfamily member GPVI. Within the last few years, major advances have been made in understanding platelet-collagen interactions including the molecular cloning of GPVI, the generation of mouse strains lacking individual collagen receptors, and the development of collagen receptor-specific antibodies and synthetic peptides. It is now recognized that platelet adhesion to collagen requires prior activation of integrins through "inside-out" signals generated by GPVI and reinforced by released second-wave mediators adenosine diphosphate (ADP) and thromboxane A2. These developments have led to revision of the original "2-site, 2-step" model, which now places GPVI in a central position in the complex processes of platelet tethering, activation, adhesion, aggregation, degranulation, and procoagulant activity on collagen. This review discusses these recent developments and proposes possible mechanisms for how GPVI acts in concert with other receptors and signaling pathways to initiate hemostasis and arterial thrombosis. Topics: Adenosine Diphosphate; Animals; Cloning, Molecular; Collagen; Hemostasis; Humans; Integrin alpha2beta1; Mice; Mice, Knockout; Models, Biological; Peptide Fragments; Platelet Activation; Platelet Adhesiveness; Platelet Glycoprotein GPIIb-IIIa Complex; Platelet Membrane Glycoproteins; Protein Isoforms; Receptors, Collagen; Signal Transduction; Thrombosis; Thromboxane A2 | 2003 |
Aspirin resistance: definition, mechanisms and clinical read-outs.
Topics: Anti-Inflammatory Agents, Non-Steroidal; Aspirin; Blood Platelets; Clinical Trials as Topic; Drug Interactions; Drug Resistance; Humans; Models, Biological; Risk; Thrombosis; Thromboxane A2; Thromboxane B2; Time Factors | 2003 |
COX inhibitors and thromboregulation.
Topics: Animals; Arthritis; Cyclooxygenase Inhibitors; Epoprostenol; Mice; Thrombosis; Thromboxane A2 | 2002 |
Cyclooxygenase inhibition and thrombogenicity.
Cyclooxygenase (COX)-1 and COX-2 catalyze the formation of prothrombotic and antithrombotic eicosanoids, respectively. Aspirin, conventional nonsteroidal anti-inflammatory drugs (NSAIDs), and COX-2-specific inhibitors exhibit different patterns of inhibition of COX-1-mediated thromboxane biosynthesis and COX-2-mediated prostacyclin biosynthesis. The relationship between the pharmacologic inhibition of these vasoactive eicosanoids and the thromboprophylaxis or thrombogenicity exhibited by different therapeutic agents is currently unclear. Future studies are needed to assess the antithrombotic properties of commonly used NSAIDs, the hypothetical thrombogenicity of COX-2-specific inhibitors in high-risk patients, the need for concomitant aspirin with selective versus nonselective COX inhibitors, and the antiplatelet and gastric toxicity of the aspirin/COX-2-specific inhibitor combination in comparison with the aspirin/conventional NSAID combination. Topics: Anti-Inflammatory Agents, Non-Steroidal; Cardiovascular Diseases; Cyclooxygenase 1; Cyclooxygenase 2; Cyclooxygenase 2 Inhibitors; Cyclooxygenase Inhibitors; Drug Interactions; Epoprostenol; Humans; Isoenzymes; Membrane Proteins; Prostaglandin-Endoperoxide Synthases; Risk Factors; Thrombosis; Thromboxane A2 | 2001 |
Angiotensin II AT(1) receptor antagonists and platelet activation.
Multiple factors are involved in thrombus formation and require complex and highly therapeutic strategies. Platelet activation plays a critical role in the genesis of acute coronary syndromes involving not only platelets but also endothelial cells, leucocytes and erythrocytes. Angiotensin II (Ang II) is a vasoconstrictor that could participate in the thrombotic process. Platelets also express Ang II AT1 type receptors on their surface. Losartan is a non-peptidic inhibitor of AT1 receptors. It has been demonstrated that losartan reduced platelet aggregation induced by the thromboxane A2 (TXA2) analogue U46619. This effect was not observed with the losartan metabolite EXP 3174. The effect of losartan was assessed in binding studies in which losartan competitively inhibited the binding of [3H]U46619 to platelets in a dose-dependent manner. Irbesartan also inhibits the TXA2 receptor in platelets, an effect that was not obtained with the active form of candesartan, CV11974, and with valsartan. These results suggest that the structural requirements necessary to antagonize the TXA2/PGH2 platelet receptor may be different from those involved in AT1 receptor antagonism. The in vivo relevance of the in vitro findings has been confirmed by the fact that in vivo administration of losartan decreases P-selectin expression in platelets obtained from stroke-prone spontaneously hypertensive rats. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Angiotensin Receptor Antagonists; Animals; Antihypertensive Agents; Benzimidazoles; Biphenyl Compounds; Coronary Disease; Coronary Thrombosis; Humans; Losartan; P-Selectin; Platelet Activation; Platelet Aggregation; Rats; Rats, Inbred SHR; Receptor, Angiotensin, Type 1; Receptor, Angiotensin, Type 2; Receptors, Thromboxane; Tetrazoles; Thrombosis; Thromboxane A2; Valine; Valsartan | 2001 |
Ifetroban sodium: an effective TxA2/PGH2 receptor antagonist.
This review presents a comprehensive discussion on the chemistry, pharmacokinetics, and pharmacodynamics of ifetroban sodium, a new thomboxane A2/prostaglandin H2 receptor antagonist. Thromboxane A2 is an arachidonic acid product, formed by the enzyme cyclooxygenase. In contrast to other cyclooxygenase products, thromboxane A2 has been shown to be involved in vascular contraction and has been implicated in platelet activation. In general, results of clinical studies and animal experiments indicate that hypertension is associated with hyperaggregability of platelets and increased thomboxane A2 levels in blood, urine, and tissues. The precursors to thromboxane A2, prostaglandin G2, and prostaglandin H2, also bind and activate the same receptors. Thus, a receptor antagonist was thought to be an improved strategy for reversing the actions of thromboxane A2/prostaglandin H2, rather than a thromboxane synthesis inhibitor. This review describes new methods for the synthesis and analysis of ifetroban, its tissue distribution, and its actions in a variety of animal models and disease states. We describe studies on the mechanisms of how ifetroban relaxes experimentally contracted isolated vascular tissue, and on the effects of ifetroban on myocardial ischemia, hypertension, stroke, thrombosis, and its effects on platelets. These experiments were conducted on several animal models, including dog, ferret, and rat, as well as on humans. Clinical studies are also described. These investigations show that ifetroban sodium is effective at reversing the effects of thromboxane A2- and prostaglandin H2-mediated processes. Topics: Animals; Bridged Bicyclo Compounds, Heterocyclic; Humans; Hypertension; Muscle Contraction; Muscle, Smooth, Vascular; Myocardial Ischemia; Oxazoles; Platelet Aggregation Inhibitors; Prostaglandin H2; Prostaglandins H; Randomized Controlled Trials as Topic; Receptors, Prostaglandin; Receptors, Thromboxane; Stroke; Thrombosis; Thromboxane A2 | 2001 |
Preoperative stop or continue? New antithrombotic and anticoagulants.
Topics: Anticoagulants; Coumarins; Fibrinolytic Agents; Hemorrhage; Heparin; Hirudins; Humans; Platelet Aggregation Inhibitors; Platelet Glycoprotein GPIIb-IIIa Complex; Preoperative Care; Purinergic P2 Receptor Antagonists; Risk Factors; Thrombosis; Thromboxane A2 | 2001 |
Roles of prostanoids revealed from studies using mice lacking specific prostanoid receptors.
The actions of prostanoids in various physiological and pathophysiological conditions have been being examined using mice lacking different prostanoid receptors. Prostaglandin (PG) I2 worked not only as a mediator of inflammation but also as an antithrombotic agent. PGF2alpha was found to be an essential inducer of labor. Several important actions of PGE2 are exerted via each of the four PGE2 receptor subtypes: EP1, EP2, EP3 and EP4. PGE2 participated in colon carcinogenesis via the EP1. PGE2 also participates in ovulation and fertilization and contributes to the control of blood pressure under high-salt intake via the EP2. PGE2 worked as a mediator of febrile responses to both endogenous and exogenous pyrogens and as a regulator of bicarbonate secretion induced by acid-stimulation in the duodenum via the EP3. It regulated the closure of ductus arteriosus and showed bone resorbing action via the EP4. PGD2 was found to be a mediator of allergic asthma. These studies have revealed important roles of prostanoids, some of which had not previously been known. Topics: Animals; Asthma; Bicarbonates; Colonic Neoplasms; Dinoprost; Dinoprostone; Female; Fever; Hypertension; Inflammation; Labor, Obstetric; Mice; Mice, Knockout; Pregnancy; Prostaglandins; Receptors, Prostaglandin; Reproduction; Thrombosis; Thromboxane A2 | 2000 |
[Effect of alcohol intake on blood vessels].
Topics: Alcohol Drinking; Blood Pressure; Cerebrovascular Circulation; Depression, Chemical; Endothelium, Vascular; Epoprostenol; Ethanol; Humans; Lipids; Platelet Aggregation; Thrombosis; Thromboxane A2 | 1997 |
Aspirin in essential thrombocythemia: status quo and quo vadis.
Aspirin has a well established role in the prevention of arterial thrombosis. Discussion on the efficacy and safety of aspirin in the treatment and prophylaxis of thrombosis in essential thrombocythemia (ET) has become an important issue. The rationale for its use in ET comes from the observation that arterial thrombosis and platelet-mediated microcirculatory disturbances are the major causes of morbidity and mortality in ET. Experimental data have shown persistently elevated levels of thromboxane A2 (TXA2) in ET patients probably reflecting an enhanced in vivo platelet activation. Increased TXA2 biosynthesis and platelet activation in vivo in ET are selectively suppressed by repeated low doses of aspirin. ET-related symptoms such as erythromelalgia, transient neurologic and ocular disturbances are sensitive to aspirin. However, the benefit of low-dose aspirin is still uncertain in the primary prevention of thrombosis in ET. Furthermore, aspirin may unmask a latent bleeding diathesis frequently present in ET which may result in severe hemorrhagic complications. Thus, aspirin is contraindicated in ET patients with a bleeding history or a very high platelet count (> 1500 x 10(9)/L) leading to the acquisition of von Willebrand factor deficiency. If indicated, aspirin is presently used in the widely accepted low-dose regimen of 100 mg daily. However, an optimal effective dose has not yet been established. To further evaluate the efficacy and safety of aspirin in ET, prospective clinical trials are needed. Topics: Abortion, Habitual; Aspirin; Cerebrovascular Disorders; Cohort Studies; Contraindications; Erythromelalgia; Female; Fibrinolytic Agents; Forecasting; Hemorrhage; Humans; Incidence; Middle Aged; Myeloproliferative Disorders; Pilot Projects; Platelet Activation; Platelet Aggregation Inhibitors; Pregnancy; Pregnancy Complications, Hematologic; Retrospective Studies; Safety; Thrombocythemia, Essential; Thrombophilia; Thrombosis; Thromboxane A2; Vision Disorders; von Willebrand Diseases | 1997 |
Mechanism of anti-proliferative action of eicosapentaenoic acid (EPA) in vascular cell growth: its effect on signal transduction system.
Topics: Administration, Oral; Animals; Anti-Inflammatory Agents, Non-Steroidal; Arachidonic Acid; Arteriosclerosis; Arteriosclerosis Obliterans; Aspirin; Cell Division; Cells, Cultured; Clinical Trials as Topic; Eicosanoids; Eicosapentaenoic Acid; Epoprostenol; Fibrinolytic Agents; Growth Inhibitors; Hemostasis; Humans; Hypolipidemic Agents; Muscle, Smooth, Vascular; Platelet Aggregation; Platelet Aggregation Inhibitors; Rats; Signal Transduction; Thrombophilia; Thrombosis; Thromboxane A2 | 1997 |
Injury models of the vascular endothelium: apoptosis and loss of thromboresistance induced by a viral protein.
Endothelial injury caused by viruses usually involves viral replication or transformation. We report a novel mechanism of endothelial damage by a toxic viral protein. We have isolated a new retrovirus from hemangiosarcomas which appeared among layer hens. The isolated avian hemangiosarcoma virus (AHV) is capable of inducing hemangiomas in hens in-vivo and causes a cytopathic effect (CPE) and loss of thromboresistance in cultured bovine aortic endothelial cells (BAEC). These effects do not require viral replication and can be induced by purified AHV envelop glycoprotein (gp85). AHV causes CPE in BAEC through a typical programmed cell death (apoptosis). Quiescent G0/G1-BAEC are much more sensitive to AHV induced apoptosis than actively dividing cells. These experiments demonstrate the capacity of viral proteins to affect the integrity and functionality of vascular endothelial cells. Topics: Animals; Aorta; Apoptosis; Avian Sarcoma Viruses; Cattle; Cell Cycle; Cells, Cultured; Chickens; Cytopathogenic Effect, Viral; Endothelium, Vascular; Extracellular Matrix; Hemangiosarcoma; Immunity, Innate; Poultry Diseases; Sarcoma, Avian; Thrombosis; Thromboxane A2; Viral Envelope Proteins | 1996 |
Platelet factors predisposing to arterial thrombosis.
Topics: Animals; Antibodies; Arteries; beta-Thromboglobulin; Blood Coagulation; Blood Coagulation Factors; Blood Platelets; Disease Susceptibility; Humans; Megakaryocytes; Platelet Activation; Platelet Aggregation; Platelet Factor 4; Stress, Mechanical; Thrombosis; Thromboxane A2; Vascular Diseases | 1994 |
Serotonin and thrombotic complications.
We believe that the abrupt conversion from chronic stable to unstable angina and the continuum to acute myocardial infarction may result from myocardial ischemia caused by progressive platelet aggregation and dynamic vasoconstriction themselves caused by local increases in thromboxane and serotonin at sites of coronary artery stenosis and endothelial injury. Platelet aggregation and dynamic coronary artery vasoconstriction probably result from the local accumulation of thromboxane and serotonin and also relative decreases in the local concentrations of endothelially derived vasodilators and inhibitors of platelet aggregation, such as endothelium-derived relaxing factor (EDRF) and prostacyclin. With severe reductions in coronary blood flow caused by these mechanisms, platelet aggregates may increase, and an occlusive thrombus composed of platelets and white and red blood cells in a fibrin mesh may develop. When coronary arteries are occluded or narrowed for a sufficient period of time by these mechanisms, myocardial necrosis, electrical instability, or sudden death may occur. We believe that unstable angina and acute myocardial infarction are a continuum in relation to the process of coronary artery thrombosis and vasoconstriction. When the period of platelet aggregation or dynamic vasoconstriction at sites of endothelial injury and coronary artery stenosis is brief, unstable angina or non-Q-wave infarction may occur. However, when the coronary artery obstruction by these mechanisms is prolonged for several hours. Q-wave myocardial infarction results. Chronic endothelial injury and coronary artery stenosis are probably associated with the accumulation of platelets, white and red blood cells, and a fibrin mesh at the site of stenosis and endothelial injury.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Coronary Disease; Humans; Nitric Oxide; Serotonin; Thrombosis; Thromboxane A2 | 1991 |
Mechanisms of platelet activation: thromboxane A2 as an amplifying signal for other agonists.
Thromboxane (Tx) A2 is a product of cyclooxygenase catalyzed metabolism of arachidonic acid. It is formed via prostaglandin (PG) endoperoxide intermediates (PGG2 and PGH2) by a specific synthase. PGH2 appears to exert the same biologic effects as TxA2. The cDNA for a TxA2 receptor has been cloned from a human placental library. Although pharmacologic and biochemical studies suggest the presence of multiple isoforms, this remains to be confirmed at the molecular level. A hydropathy plot of the deduced amino acid sequence of the available clone suggests that it has 7 transmembrane spanning domains, typical of a G protein linked receptor. Pharmacologic studies imply that Tx receptors in platelets are linked to phospholipase C activation via pertussis toxin insensitive G proteins. Candidates include the 42 kD Gq and the 60 kD Ge. TxA2 acts as an amplifying signal for platelet agonists and the response to this eicosanoid is tightly regulated. Mechanisms include rapid hydrolysis of the agonist to the inactive TxB2, autoinactivation of Tx synthase, rapid homologous TxA2 receptor desensitization due to receptor-G protein uncoupling, coincidental sensitization to counterregulatory Gs linked receptor systems and stimulation of prostacyclin formation by TxA2. Due to its role as an amplification signal in platelet activation, inhibition of Tx synthesis and action is an effective mechanism for preventing platelet-dependent vascular occlusion. Aspirin is of proven efficacy in this regard. Tx synthase inhibitors and antagonists are under clinical investigation. Topics: Humans; Platelet Activation; Thrombosis; Thromboxane A2 | 1991 |
The role of serotonin in thrombogenesis.
Serotonin, a weak activator on its own, elicits strong platelet reactions (aggregation, arachidonate metabolization, release reaction) through amplification. Such amplification operates via 5-HT2-serotonergic receptors which are linked to the turnover of polyphosphoinositides and increases of intracellular [Ca2+]i concentrations as signal transducing systems. In experimental animals, selective blockade of 5-HT2-serotonergic receptors eliminates arterial thrombus formation over sites of endothelial injury in stenosed canine coronaries; in combination with a manipulation of TXA2, it prevents coronary thrombotic reocclusion after fibrinolysis with rt-PA, and inhibits drastic platelet activation, elicited by injections of collagen in vivo. These observations suggest that serotonergic amplification substantially contributes to the aggregation of platelets in damaged vessels and offers perspectives for an improved antithrombotic approach. Topics: Animals; Blood Platelets; Calcium; Coronary Disease; Dogs; Fibrinolysis; Ketanserin; Phosphatidylinositols; Platelet Activation; Platelet Aggregation; Receptors, Serotonin; Serotonin; Serotonin Antagonists; Signal Transduction; Thrombosis; Thromboxane A2 | 1990 |
[New drugs for the inhibition of the pro-thrombotic effects of prostaglandins and thromboxane A2].
Topics: Humans; Platelet Aggregation; Platelet Aggregation Inhibitors; Prostaglandin Antagonists; Thrombosis; Thromboxane A2 | 1990 |
[Pharmacological correction of disorders of the prostacyclin- thromboxane balance in ischemic heart disease: problems and prospects].
Topics: Aspirin; Coronary Disease; Epoprostenol; Humans; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1990 |
Amplification mechanisms in platelet activation and arterial thrombosis.
Platelets react with different intensities to a large variety of agonists (e.g. 5-hydroxytryptamine, ADP, thrombin, thromboxane A2). In vitro, low concentrations of 'weak' agonists such as 5-hydroxytryptamine markedly enhance the platelet functional response in terms of aggregation, release of intragranular products and arachidonic acid metabolites to otherwise ineffective concentrations of other agonists. This type of amplification takes place at an early, postreceptor stage of the platelet activation cascade, with the turnover of inositol phospholipids and the increase in free cytoplasmic intracellular Ca2+ concentrations. Mutual amplification, rather than the single potency of each, also determines the contribution by platelet agonists to thrombogenesis in vivo. Indeed, in various animal models and, to some extent, in man, a pharmacological blockade of the platelet 5-HT2 serotonergic (e.g. ketanserin) or thromboxane A2/prostaglandin endoperoxide receptor (e.g. ridogrel) and manipulation of arachidonic acid metabolism (e.g. cyclo-oxygenase inhibition with aspirin or thromboxane A2 synthetase inhibition with ridogrel), when applied together, are more effective in preventing extensive platelet activation and arterial thrombotic occlusion and in improving the efficacy of thrombocytic agents (speed up of thrombolysis and prevention of thrombotic re-occlusion after lysis) than when the same compounds are applied separately. These observations suggest that an amplification between 5-hydroxytryptamine and thromboxane A2, for example, as platelet agonists is causally involved in arterial thrombus formation. Topics: Animals; Arachidonic Acid; Arachidonic Acids; Humans; Platelet Activation; Platelet Aggregation; Serotonin; Thrombosis; Thromboxane A2 | 1990 |
Evidence that combined thromboxane A2 and serotonin receptor blockade might prevent coronary artery thrombosis and the conversion from chronic to acute coronary heart disease syndromes.
Evidence that combined thromboxane A2 and serotonin blockade may prevent coronary artery thrombosis and the conversion from chronic to acute coronary heart disease syndromes is considered. Available data from clinical and experimental animal studies are consistent with the hypothesis that interference with thromboxane and serotonin's contributions to platelet aggregation and dynamic coronary artery constriction might prevent the conversion from chronic to acute disorders, including the development of unstable angina and myocardial infarction in some patients at risk. Substantial protection preventing the conversion from stable to unstable angina and myocardial infarction may very well require both thromboxane and serotonin receptor antagonists or the combination of a thromboxane synthesis inhibitor and receptor antagonist with a serotonin receptor antagonist. Future clinical studies should test this hypothesis directly. Topics: Animals; Coronary Disease; Disease Models, Animal; Humans; Serotonin Antagonists; Thrombosis; Thromboxane A2 | 1990 |
Aspirin as an antithrombotic drug: from the aggregometer to clinical trials.
Aspirin inhibits thromboxane A2 and prostaglandin formation in platelets and prostaglandin I2 (prostacyclin) in vascular cells. It prevents platelet aggregation by irreversible acetylation of cyclooxygenase, a key enzyme in the arachidonic acid metabolism. Oral aspirin can be extensively hydrolyzed to inactive salicylate in the stomach and the liver (first-pass) before it enters the systemic circulation. Presystemic acetylation of platelets thus occurs during aspirin absorption, with a concomitant sparing of peripheral vascular cyclo-oxygenase, mainly exposed to salicylate. On the basis of its antiplatelet effect, aspirin has been assessed during the past two decades in patients with a history of myocardial infarction, stroke, transient ischemic attack or unstable angina. A meta-analysis of randomized controlled trials of long term aspirin treatment for secondary prevention of vascular disease indicated that aspirin (300-1500 mg daily) significantly reduced fatal and non-fatal vascular events. More recently aspirin (160 mg daily) produced a significant reduction in hospital vascular mortality and in non-fatal events in patients with suspected acute myocardial infarction. Combination of aspirin with streptokinase was significantly better than either drug alone. On the other hand two primary prevention trials of aspirin in healthy doctors did not show any modification of vascular mortality despite an overall reduction of non-fatal myocardial infarction. Resolution of some problems related to the mechanism of action of aspirin and to selection of trial populations will possibly increase the benefit/risk ratio of aspirin treatment for prevention of vascular disease. Topics: Aspirin; Cerebrovascular Disorders; Humans; Meta-Analysis as Topic; Myocardial Infarction; Platelet Aggregation; Platelet Aggregation Inhibitors; Prostaglandin Antagonists; Randomized Controlled Trials as Topic; Thrombosis; Thromboxane A2 | 1990 |
Platelet aggregation: cellular regulation and physiologic role.
Topics: Blood Platelets; Cell Communication; Contractile Proteins; Diglycerides; Glycoproteins; Humans; Membrane Glycoproteins; Platelet Adhesiveness; Platelet Aggregation; Thrombosis; Thrombospondins; Thromboxane A2 | 1988 |
Overview of physiological and pathophysiological effects of thromboxane A2.
Thromboxane (Tx) A2 is a biologically potent and chemically unstable metabolite of prostaglandin endoperoxides. Recent developments in measurement techniques and the availability of both selective inhibitors of Tx synthetase and TxA2 receptor antagonists have facilitated the implication of TxA2 as a physiological modulator and as a mediator in thrombotic, vasospastic, and bronchospastic conditions. TxA2 is synthesized by platelets and contributes to platelet activation and irreversible platelet aggregation in physiological hemostasis and in thrombosis (e.g., unstable angina, stroke). TxA2 is also synthesized in intestinal, pulmonary, and renal tissues by cells other than platelets. Particularly in these tissues, TxA2 appears to act as a physiological modulator of changes in blood flow distribution and airway caliber. Strong stimuli for TxA2 release from these tissues may initiate ulcer, pulmonary hypertension, bronchoconstriction, and renal vasoconstriction. Evidence supports participation of TxA2 and/or TxA2 receptors in modulation of natural cytotoxic cell cytotoxicity, in tumor growth and metastasis, in complications of pregnancy (e.g., preeclampsia), and in the progression of ischemic injury after coronary artery occlusion. This evidence supports pivotal involvement of TxA2 in pathophysiology and provides a strong rationale for pursuing TxA2-blocking strategies in drug development. Topics: Animals; Blood Platelets; Bronchial Spasm; Cardiovascular Diseases; Hemostasis; Humans; Muscle Contraction; Muscle, Smooth; Platelet Aggregation; Prostaglandin Endoperoxides; Receptors, Prostaglandin; Receptors, Thromboxane; Thrombosis; Thromboxane A2; Thromboxane-A Synthase; Vasoconstriction | 1987 |
Eicosanoid biosynthesis in human cardiovascular disease.
Thromboxane A2, the predominant cyclooxygenase product in platelets, is a potent platelet agonist and vasoconstrictor in vitro. Prostacyclin, the major product of vascular endothelium, has opposite effects on platelet function and vascular tone. These properties prompted the hypothesis that a "balance" between these compounds regulated interactions between platelets and the vessel wall in vivo. Although this possibility has been addressed extensively through experiments in vitro, clinical investigations commonly have been confounded by problems with analytic methodology, by selection of inappropriate metabolic targets for analysis, and by artifacts of trial design. The most reliable forms of assessing biosynthesis that are currently available still do not provide definitive information as to the tissue of origin of the compound studied and are directed toward stable but biologically inactive metabolites rather than the evanescent primary compounds themselves. Despite these limitations, both biochemical evidence and clinical trials clearly implicate thromboxane A2 as an important mediator of vascular occlusive disease in humans. The role of prostacyclin is much more conjectural. It does not circulate in concentrations sufficient to exert a systemic effect, but it may play a local homeostatic role in the regulation of platelet-vascular interactions. Whether preservation of the capacity to form prostacyclin coincident with inhibition of thromboxane A2 is of functional importance can be addressed only by clinical trials comparing inhibitors of thromboxane synthesis inhibition that are selective with cyclooxygenase inhibitors that also block the biosynthesis of prostacyclin. The recognition that multiple factors have the potential to regulate both platelet and vascular function at their interface renders the concept of a thromboxane A2-prostacyclin "balance" somewhat unlikely. However, both eicosanoids may interact with other factors to determine the development or persistence of vascular occlusion. Inhibition of the synthesis or function of thromboxane A2 remains the predominant mechanism for achieving interference with platelet function in vivo. Accumulating evidence for the efficacy of aspirin in human syndromes of vascular occlusion suggests that the biologic role of these compounds in humans should be pursued. Topics: Cardiovascular Diseases; Eicosanoic Acids; Epoprostenol; Humans; Thrombosis; Thromboxane A2; Thromboxane B2 | 1987 |
[Physiopathology of thrombosis: plasma fatty acids].
Topics: Animals; Asian People; Diet; Eicosapentaenoic Acid; Epoprostenol; Fatty Acids; Fishes; Greenland; Humans; Inuit; Japan; Platelet Aggregation; Thrombosis; Thromboxane A2 | 1986 |
[Pathology of thrombosis].
Topics: Animals; Blood Coagulation; Coronary Disease; Coronary Vasospasm; Glycoproteins; Humans; Intracranial Embolism and Thrombosis; Muscle, Smooth, Vascular; Myocardial Infarction; Plasminogen Activators; Platelet Adhesiveness; Platelet Aggregation; Protein C; Thrombosis; Thromboxane A2 | 1986 |
[Physiopathology of thrombosis: prostaglandins].
Topics: Arachidonic Acids; Blood Platelets; Chemotaxis, Leukocyte; Diabetes Mellitus; Humans; Myeloproliferative Disorders; Neutrophils; Platelet Aggregation; Prostaglandins; Purpura, Thrombotic Thrombocytopenic; Thrombosis; Thromboxane A2; Vasoconstriction | 1986 |
Role of platelet activation and fibrin formation in thrombogenesis.
Further progress in the search for more effective but safe antithrombotic agents is coupled to an improved understanding of the factors involved in arterial and venous thrombogenesis. Although arterial thrombosis is initiated by formation of a layer of platelets on modified endothelium or subendothelial constituents and subsequent recruitment of passing-by platelets, this phenomenon is not sufficient to lead to a full thrombus. Further growth of such a platelet mass depends, to a large extent, on the presence of free thrombin. Thrombin is mainly generated by activation of factor XI on the platelet contact with collagen. In addition, thrombin leads to formation of fibrin, which maintains the stability of the arterial platelet thrombus and is the main component of the venous thrombus. The search for agents that inhibit platelet activation and thrombin formation is, therefore, a logical endeavor. Topics: Blood Coagulation; Blood Platelets; Calcium; Epoprostenol; Humans; Platelet Adhesiveness; Platelet Aggregation; Protein C; Thrombosis; Thromboxane A2 | 1986 |
[Role of prostacyclin in thromboresistance].
Topics: Animals; Dietary Fats; Eicosapentaenoic Acid; Epoprostenol; Humans; Platelet Aggregation; Thrombosis; Thromboxane A2 | 1985 |
Platelets, endothelium, and vessel injury.
Injury to the endothelial lining of arteries is an important mechanism in both the early and late stages of the development of atherosclerosis. Platelets can contribute to the early lesions by releasing factors that cause smooth muscle cell migration and proliferation. In the later stages, the formation of large platelet-fibrin thrombi that become organized into the vessel wall contributes to the development of focal atherosclerotic narrowing of arteries. Injury to the vessel wall can also be a factor in causing spasm of coronary arteries, particularly at sites of stenosis. The spasm may cause ischemia, anginal pain, and, in some individuals, ventricular fibrillation and death. In other individuals, the spasm may not cause death but may persist long enough for an occlusive thrombus to form and cause myocardial infarction. The events leading to thrombosis involve not only the release of arachidonic acid and the formation of TXA2, but other pathways that are independent of the arachidonate pathway. In some circumstances thrombin (which causes platelet aggregation and release that are largely independent of the arachidonate pathway and TXA2 formation) is the primary stimulus causing the initiation and growth of the thrombus. The role of products of the arachidonate pathway in causing spasm is not understood. PGI2 produced by the vessel wall could be important in preventing or minimizing coronary artery spasm. The best way to prevent the development of atherosclerosis and its clinical complications is to prevent or minimize injury of the endothelium. Topics: Animals; Arachidonic Acid; Arachidonic Acids; Arteriosclerosis; Blood Platelets; Cell Division; Coronary Vasospasm; Endothelium; Epoprostenol; Humans; Muscle, Smooth, Vascular; Platelet Aggregation; Thrombosis; Thromboxane A2; Vasoconstriction | 1985 |
Thromboxane in sudden death.
Thromboxane has characteristics that signify potential importance in cardiovascular disease states. In models developed for studying thrombotic sudden death, thromboxane appears to be an important mediator. Thus, in arachidonic acid-induced sudden death, agents that either inhibit thromboxane generation or block thromboxane receptor activation prevent the occurrence of thrombotic death. Thromboxane mimetics are also useful in modeling sudden death; when injected i.v., these compounds elicit effects similar to those obtained with arachidonic acid. In this case, however, pretreatment with cyclooxygenase or thromboxane synthetase inhibitors confers no protection, whereas the thromboxane receptor antagonist retains its efficacy. Other factors that affect susceptibility to experimental sudden death include gender, species, and endocrine status. Thrombotic sudden death models have now been used to test, in vivo, the in vitro antiplatelet aggregatory effect of calcium-channel blockers. The data suggest that dihydropyridine agents such as nifedipine and nisoldipine are protective against thrombosis, whereas verapamil may have little such activity. Furthermore, sudden death induced by a variety of thrombotic challenges is prevented by pretreatment with nifedipine. The thrombotic sudden death models currently employed are useful for the in vivo study of the thrombotic process and for the evaluation of agents with potentially thrombotic or antithrombotic properties. Topics: Angina Pectoris; Animals; Arachidonic Acid; Arachidonic Acids; Calcium Channel Blockers; Castration; Death; Estrogens; Fatty Acids, Monounsaturated; Fatty Acids, Unsaturated; Female; Glucocorticoids; Humans; Male; Methacrylates; Prostaglandin Endoperoxides, Synthetic; Sex Factors; Testosterone; Thrombosis; Thromboxane A2; Thromboxane B2; Thromboxanes | 1985 |
[Platelet aggregation, thrombosis formation and prostaglandins].
Topics: Animals; Arachidonic Acids; Blood Vessels; Collagen; Dogs; Epoprostenol; Fibrinogen; Glycoproteins; Humans; Membrane Proteins; Platelet Aggregation; Platelet Membrane Glycoproteins; Prostaglandins; Rabbits; Thrombosis; Thromboxane A2 | 1985 |
[Blood platelets, leukocytes and disseminated intravascular coagulation].
Topics: Adult; Aged; Blood Platelets; Disseminated Intravascular Coagulation; Female; Humans; Leukocytes; Male; Middle Aged; Platelet Aggregation; Prostaglandins; Thromboplastin; Thrombosis; Thromboxane A2 | 1985 |
[Advances in the laboratory diagnosis of disorders of hemostasis].
Topics: Antithrombin III; Blood Coagulation Tests; Chromogenic Compounds; Disseminated Intravascular Coagulation; Epoprostenol; Heparin; Heparin Antagonists; Humans; Thrombosis; Thromboxane A2 | 1985 |
[Pathogenetic importance of prostacyclin in atherosclerosis].
Topics: 6-Ketoprostaglandin F1 alpha; Animals; Arteriosclerosis; Blood Platelets; Blood Vessels; Diet, Atherogenic; Epoprostenol; Hemodynamics; Humans; Lipid Peroxides; Lipoproteins, LDL; Lipoproteins, VLDL; Muscle, Smooth, Vascular; Platelet Aggregation; Rabbits; Risk; Stress, Physiological; Thrombosis; Thromboxane A2; Time Factors | 1985 |
Rationale for the use of drugs inhibiting platelet function in claudicating patients with atherosclerotic leg arteries.
Topics: Anticoagulants; Arteriosclerosis; beta-Thromboglobulin; Blood Platelets; Clinical Trials as Topic; Humans; Intermittent Claudication; Leg; Platelet Aggregation; Platelet Factor 4; Suloctidil; Thiophenes; Thrombosis; Thromboxane A2; Ticlopidine | 1984 |
Clinical aspects of the arachidonic acid--thromboxane pathway.
Topics: Arachidonic Acid; Arachidonic Acids; Aspirin; Blood Coagulation; Cerebrovascular Disorders; Epoprostenol; Female; Humans; Male; Myocardial Infarction; Platelet Aggregation; Receptors, Prostaglandin; Receptors, Thromboxane; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1983 |
[Pharmacology of anti-platelet agents].
A new class of drugs has appeared alongside classical anti-thrombotic agents such as heparin and oral anticoagulants, characterised by their ability to modify the behaviour of platelets: "anti-platelet" agents. This article reviews the platelet actions, pharmacokinetics, conditions of use and side effects of the four chief agents available: acetylsalicylic acid (ASA), sulfinpyrazone, dipyridamole and ticlopidine. The mode of action of the first of them is that best studied. ASA opposes the conversion of arachidonic acid to prostaglandins and thromboxane, by the irreversible acetylation of cyclo-oxygenase. Nevertheless, major therapeutic trials involving ASA have yielded only poor results. There are at least two possible explanations for this state of affairs: --aggregation may occur even when thromboxane is blocked, in particular in response to thrombin; --ASA has been used at doses also capable of inhibiting the formation by the vascular wall of an anti-aggregant prostaglandin, PGI2. Current efforts by pharmacologists which should result in better adapted and hence more effective anti-thrombotic methods, are essentially concerned with the following points: --to understand why sulfinpyrazone, which in principle has the same mode of action as ASA, seems sometimes more active and sometimes less active than the latter according to whether coronary or cerebrovascular accidents are involved; --to propose a rational prescription programme for ASA, in such a way that it inhibits only little, and for as short a time as possible, the production of PGI2 (e.g. 200 mg every three days): --to perfect more active combinations; --synthesis of new substances, e.g. thromboxane synthetase inhibitors, or stable analogues of PGI2. The reasons which suggest that such substances could be used more beneficially in man are expanded. Topics: Animals; Anticoagulants; Aspirin; Blood Platelets; Cyclic AMP; Cyclooxygenase Inhibitors; Dipyridamole; Epoprostenol; Female; Fibrinolytic Agents; Humans; In Vitro Techniques; Intestinal Absorption; Kinetics; Male; Papio; Platelet Aggregation; Prostaglandin Antagonists; Rabbits; Sulfinpyrazone; Thiophenes; Thrombosis; Thromboxane A2; Ticlopidine | 1983 |
Introduction: thromboxane in biological systems and the possible impact of its inhibition.
Topics: Animals; Epoprostenol; Humans; Kidney; Neoplasms; Shock; Thrombosis; Thromboxane A2; Thromboxane-A Synthase; Thromboxanes; Vascular Resistance | 1983 |
Aspirin for prevention of stroke: a review.
Most neurologists concede that thromboembolism is the principal pathogenetic mechanism for ischemic cerebrovascular disease, including both transient ischemic attacks and cerebral infarction. Surgical removal of atherosclerotic lesions may eradicate the site of origin of emboli, but a safer and more rational approach may be found in using antithrombotic drugs. Aspirin has been shown in clinical trials to be an effective agent in treating transient ischemic attacks and preventing infarction. An apparent difference in response between men and women patients was found in the results of one large study but not substantiated by others. It has been suggested that a lower dose of aspirin than that used in these trials may be equally or more effective. This proposition still needs to be tested in a clinical trial. Topics: Aspirin; Cell Adhesion; Cerebrovascular Disorders; Humans; Ischemic Attack, Transient; Platelet Aggregation; Prostaglandins; Thrombosis; Thromboxane A2 | 1983 |
The prostacyclin--thromboxane A2 balance: pathophysiological and therapeutic implications.
Topics: Animals; Cytochrome P-450 Enzyme System; Diet; Epoprostenol; Hemorrhage; Humans; Intramolecular Oxidoreductases; Platelet Aggregation; Prostaglandin Endoperoxides; Thrombosis; Thromboxane A2; Thromboxane-A Synthase; Thromboxanes | 1983 |
Prostacyclin in physiology and pathophysiology.
Topics: Animals; Arachidonic Acids; Blood Platelets; Blood Vessels; Digestive System; Epoprostenol; Female; Hemostasis; Humans; Lipid Peroxides; Lung; Platelet Aggregation; Prostaglandin Endoperoxides; Prostaglandin-Endoperoxide Synthases; Prostaglandins; Thrombosis; Thromboxane A2; Uterus | 1982 |
Prostacyclin: a hormone with a therapeutic potential. The Sir Henry Dale Lecture for 1981.
Topics: Animals; Arachidonic Acids; Blood Platelets; Cardiopulmonary Bypass; Cyclooxygenase Inhibitors; Epoprostenol; Female; Hemostasis; Humans; Inflammation; Lung; Platelet Aggregation; Prostaglandin Endoperoxides; Prostaglandins; Thrombosis; Thromboxane A2; Uterus; Vasodilator Agents | 1982 |
Thromboxanes and ischaemic vascular disease.
Topics: Animals; Arteriosclerosis; Epoprostenol; Hemostasis; Humans; Platelet Aggregation; Thrombosis; Thromboxane A2; Thromboxanes | 1982 |
Drugs that modulate prostaglandins, prostacyclin, and thromboxane A2.
Topics: Animals; Cardiovascular Physiological Phenomena; Epoprostenol; Fatty Acids; Fibrinolytic Agents; Hemostasis; Humans; Prostaglandins; Thrombosis; Thromboxane A2; Thromboxanes | 1982 |
[The role of prostaglandins in hemostasis and thrombosis].
Topics: Animals; Arteriosclerosis; Chemical Phenomena; Chemistry; Epoprostenol; Hemostasis; Heparin; Humans; Prostaglandins; Prostaglandins G; Thrombosis; Thromboxane A2 | 1981 |
Prostacyclin.
Topics: Animals; Cell Communication; Cyclic AMP; Epoprostenol; Fatty Acids; Hemostasis; Intestinal Mucosa; Platelet Adhesiveness; Platelet Aggregation; Prostaglandin-Endoperoxide Synthases; Prostaglandins; Rabbits; Rats; Thrombosis; Thromboxane A2; Vasodilation | 1980 |
The prostanoids in hemostasis and thrombosis: a review.
Topics: Animals; Blood Platelets; Hemostasis; Humans; Muscle, Smooth; Phospholipases; Prostaglandin Endoperoxides; Prostaglandin Endoperoxides, Synthetic; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1980 |
Prostaglandins and platelets.
Release of arachidonate from cell membrane phospholipids by activation of phospholipase A1 is a key step in the formation of prostaglandins and thromboxanes. In platelets PGH2 and TXA2 are formed from arachidonate and can cause aggregation and the release of granule contents; in vascular tissue, PGI2 is formed instead and, by increasing platelet cAMP, inhibits platelet reactions. There is considerable interest in inhibitors of the enzymes in these pathways as drugs to modify thrombus formation. Results of the clinical trials, however, indicate that drugs which inhibit cyclo-oxygenase may not have a major effect on the thromboembolic complications of arterial disease. Topics: Arachidonic Acids; Aspirin; Blood Platelets; Epoprostenol; Fatty Acids; Humans; Oxygenases; Phospholipases A; Phospholipases A1; Platelet Adhesiveness; Platelet Aggregation; Prostaglandins; Sulfinpyrazone; Thrombosis; Thromboxane A2 | 1980 |
Thromboxane A2 in blood vessel walls and its physiological significance: relevance to thrombosis and hypertension.
It has been thought that blood vessels apart from the umbilical artery produce little or no thromboxane (TX) A2. However selective inhibitors of TXA2 biosynthesis have substantial effects on vessel physiology, suggesting that small amounts of TXA2 may be important in regulating function. This indirect evidence is now supported by direct measurements of TXB2 (the produce of TXA2 conversion) using both gas chromatography-mass spectrometry (GCMS) and radioimmunoassays. At least four independent laboratories have now demonstrated TXB2 production by various blood vessels. These studies suggest that vessel wall TXA2 is present in amounts more than adequate to exert biological actions on both vascular reactivity and on platelets. This may require re-evaluation and revision of present concepts of hypertension and thrombosis. Topics: Animals; Blood Vessels; Humans; Hypertension; Thrombosis; Thromboxane A2; Thromboxanes | 1980 |
The role of prostacyclin and thromboxane A2 in the regulation of platelet behaviour.
Topics: Aspirin; Dipyridamole; Epoprostenol; Hemorrhage; Humans; Platelet Aggregation; Prostaglandins; Thrombosis; Thromboxane A2; Thromboxanes | 1980 |
The role of prostacyclin in vascular tissue.
Prostacyclin (PGI2) generated by the vascular wall is a potent vasodilator, and the most potent endogenous inhibitor of platelet aggregation so far discovered. Prostacyclin inhibits platelet aggregation by increasing cyclic AMP levels. Prostacyclin is a circulating hormone continually released by the lungs into the arterial circulation. Circulating platelets are, therefore, subjected constantly to prostacyclin stimulation and it is via this mechanism that platelet aggregability in vivo is controlled. Moreover, phosphodiesterase inhibitors such as dipyridamole or theophylline exert their antithrombotic actions by potentiating circulating prostacyclin. The prostacyclin:thromboxane A2 ratio is important in the control of thrombus formation; manipulation of this ratio by small doses of aspirin (which will inhibit mainly platelet cyclooxygenase), a selective inhibitor of thromboxane formation, or the dietary use of a fatty acid like eicosapentaenoic acid (which would be the precursor for a delta17-prostacyclin (PGI3) but is transformed by the platelets into nonaggregating thromboxane A3) might have beneficial effects as antithrombotic therapies. Prostacyclin has interesting potential for clinical application in conditions where enhanced platelet aggregation is involved or to increase biocompatibility of extracorporeal circulation systems. Topics: Animals; Arachidonic Acids; Aspirin; Blood Coagulation; Blood Platelets; Blood Vessels; Cyclic AMP; Dipyridamole; Epoprostenol; Linolenic Acids; Platelet Aggregation; Prostaglandin-Endoperoxide Synthases; Prostaglandins; Thrombosis; Thromboxane A2; Vasodilation | 1979 |
Prostaglandins and thromboxanes: "middlemen" modulating platelet function in hemostasis and thrombosis.
Topics: Adenosine Diphosphate; Animals; Arachidonic Acids; Arteriosclerosis; Blood Coagulation; Blood Platelets; Calcium; Cyclic AMP; Hemostasis; Humans; Prostaglandins; Prostaglandins G; Prostaglandins H; Rats; Thrombosis; Thromboxane A2; Thromboxanes | 1978 |
[Thromboxane A2 antagonist, prostacyclin (PGI2)--its in vivo secretion and action mechanism].
Topics: Animals; Epoprostenol; Humans; Intracranial Arteriosclerosis; Microsomes; Platelet Aggregation; Prostaglandins; Rabbits; Rats; Thrombosis; Thromboxane A2; Thromboxanes | 1978 |
[Determination of TXA2 formed by blood platelets and PGX formed by blood vessels and formation of thrombi].
Topics: Blood Platelets; Blood Vessels; Epoprostenol; Humans; Platelet Aggregation; Prostaglandins; Thrombosis; Thromboxane A2; Thromboxanes | 1977 |
7 trial(s) available for thromboxane-a2 and Thrombosis
Article | Year |
---|---|
Thromboxane A(2) generation, in the absence of platelet COX-1 activity, in patients with and without atherothrombotic myocardial infarction.
Aspirin's therapeutic action is via inhibition of platelet cyclooxygenase 1 (COX-1) thromboxane A2 (TxA2) production. The aim of this study was to evaluate TxA2 production, in the absence of platelet COX-1 activity, in coronary atherosclerotic heart disease patients with and without atherothrombotic myocardial infarction (MI).. TxA2 production, in the absence of platelet COX-1 activity, was evaluated in 44 patients taking aspirin on 3 commercially available assays that detect metabolites of TxA2 in the urine. Two assays measure urine 11-dehydro-thromboxane B2 (TxB2) alone and 1 measures urine 11-dehydro-TxB2 plus 11-dehydro-2,3-dinor-TxB2. Platelet COX-1 inhibition was confirmed on <10% platelet aggregation in response to ≥1 mmol/L arachidonic acid. Median urine 11-dehydro-TxB2 was no different in those with and without a diagnosis of atherothrombotic MI (325 vs. 311 pg/mg creatinine, P=0.59 via polyclonal ELISA) and (312 vs. 244 pg/mg creatinine, P=0.11 via LC-MS/MS). Median urine 11-dehydro-TxB2 plus 11-dehydro-2,3-dinor-TxB2, however, was higher in those with vs. those without a diagnosis of atherothrombotic MI (1,035 vs. 606 pg/mg creatinine, P=0.03 via monoclonal ELISA).. Differences in TxA2 production, in the absence of platelet COX-1 activity, between those with vs. without atherothrombotic MI were not observed when TxA2 generation was assessed on 11-dehydro-TxB2 production alone (polyclonal ELISA or LC-MS/MS), but differences were observed when TxA2 generation was assessed using 11-dehydro-TxB2 plus 11-dehydro-2,3-dinor-TxB2 (monoclonal ELISA). These findings highlight important differences between different commercially available assays for TxA2 generation and suggest that 11-dehydro-2,3-dinor-TxB2 may be critical to the biology of atherothrombosis. Topics: Aged; Aspirin; Blood Platelets; Coronary Artery Disease; Creatinine; Cyclooxygenase 1; Cyclooxygenase Inhibitors; Female; Humans; Male; Middle Aged; Myocardial Infarction; Platelet Aggregation; Thrombosis; Thromboxane A2; Thromboxane B2 | 2013 |
Different therapy for different types of ulcerative colitis in China.
To study the different therapy for different types of ulcerative colitis (UC) in China.. Among 102 UC patients, 42 chronic relapse type UC patients were randomly divided into olsalazine sodium treatment group (n=21) and SASP group (n=21). Clinical effects and safety were observed in the 2 groups. Forty-two first episode type UC patients were randomly divided into Heartleaf houttuynia herb treatment group (n=21) and SASP group (n=21). Clinical effects were observed in the 2 groups while ultrastructure of colonic mucosa, ICAM-1 and the pressure of distant colon were studied in Heartleaf houttuynia herb group. Eighteen patients (8 males, 10 females) with refractory UC and unresponsive to high-dose prednisolone and sulfasalazine therapy more than one month were treated with Kangshuanling (7200 U/d). Prednisolone was gradually stopped and sulfasalazine was maintained. Stool frequency, rectal bleeding, colonoscopy, general well-being, histology were observed and CD62p, CD63, CD54, Pgp-170 (flow cytometry), TXA2 (RIA), blood platelet aggregation rate and thrombosis length in vitro were assessed.. In the 42 chronic relapse type UC patients, the overall clinical effects of olsalazine sodium group (complete remission in 16, improvement in 4, inefficiency in 1) were better than those of SASP group (complete remission in 10, improvement in 4, inefficiency in 7, P<0.05). Symptomatic remission of olsalazine sodium group (complete remission in 15, partial remission in 5, inefficiency in 1) was better than that of SASP group (complete remission in 10, partial remission in 5, inefficiency in 6, P<0.05). The colonoscopic remission of olsalazine sodium group(complete remission in 11, partial remission in 9, inefficiency in 1) was better than that of SASP group (complete remission in 7, partial remission in 8, inefficiency in 6, P<0.05). The histologic remission of olsalazine sodium group (complete remission in 13, partial remission in 7, inefficiency in in 1) was better than that of SASP group (complete remission in 6, partial remission in 10, inefficiency in 5, P<0.05). The side effects of gastrointestinal tract in olsalazine sodium group were less than those of SASP group except for frequency of watery diarrhea. No other side effects were observed in olsalazine sodium group while ALT increase, WBC decrease and skin eruption were observed in SASP group. Two patients relapsed in olsalazine sodium group while 8 cases relapsed in SASP group during the flow-up period (from six months to one year). In the 42 first episode type UC patients, the clinical effect of Heartleaf houttuynia herb group (complete remission in 20, 95.2%; improvement in 1, 4.8%) was better than that of SASP group (complete remission in 15, 72.4%, improvement in 5, 23.8%; inefficiency in 1, 3.8%, P<0.01). The time of stool frequency recovering to normal (5.6+/-3.3 d), and blood stool disappearance (6.7+/-3.8 d) and abdominal pain disappearance (6.1+/-3.5 d) in Heartleaf houttuynia herb group was all shorter than that in SASP group (9.5+/-4.9 d, 11.7+/-6.1 d, 10.6+/-5.3 d, P<0.01). Heartleaf houttuynia herb could inhibit the epithelial cell apoptosis of colonic mucous membrane and the expression of ICAM-1 (45.8+/-5.7% vs 30.7+/-4.1%, P<0.05). Compared with normal persons, the mean promotive speed of contraction wave stepped up (4.6+/-1.6 cm/min vs 3.2+/-1.8 cm/min, P<0.05) and the mean amplitude of the wave decreased (14.2+/-9.3 kPa vs 18.4+/-8.0 kPa, P<0.05) in active UC patients. After treatment with Heartleaf houttuynia herb, these 2 indexes improved significantly (17.3+/-8.3 kPa, 3.7+/-1.7. Based on the characteristics of UC cases in China, different therapy should be given to different types of UC with expected satisfactory results. Topics: Adult; Aminosalicylic Acids; Anti-Inflammatory Agents; Antigens, CD; ATP Binding Cassette Transporter, Subfamily B, Member 1; China; Colitis, Ulcerative; Drugs, Chinese Herbal; Epithelial Cells; Female; Houttuynia; Humans; Male; Platelet Activation; Prednisolone; Sulfasalazine; Thrombosis; Thromboxane A2 | 2004 |
Continued thromboxane A2 formation despite administration of a platelet glycoprotein IIb/IIIa antagonist in patients undergoing coronary angioplasty.
Experimental data suggest that formation of thromboxane A2 may be suppressed during administration of a glycoprotein IIb/IIIa antagonist. We determined the dose of one such compound, fradafiban, required to provide > 80% occupancy of the platelet glycoprotein IIb/IIIa and examined its effects on thromboxane A2 formation in patients undergoing PTCA. The dose response to fradafiban and additional effects of aspirin were explored initially in patients with stable coronary artery disease. Fradafiban induced a dose-dependent inhibition of platelet aggregation that correlated with fibrinogen receptor occupancy and plasma drug concentration. Addition of aspirin 300 mg had no effect on these parameters. At the highest dose, mean fibrinogen receptor occupancy was 89.7 +/- 1.2% (n = 3) at 4 hours and platelet aggregation had decreased by 93.4 +/- 2.7%. Eighteen patients undergoing coronary angioplasty were randomized to receive either aspirin 330 mg or that dose of fradafiban producing > 80% fibrinogen receptor occupancy. Platelet aggregation was suppressed throughout the infusion of fradafiban to a greater extent than with aspirin. However, there was a marked increase in urinary excretion of 11-dehydrothromboxane B2 in patients treated with fradafiban: from 1973 +/- 889 to a peak of 9760 +/- 3509 pg/mg creatinine (P = .0046). Despite this evidence of continued platelet activation in vivo, there were no cases of coronary thrombosis. In conclusion, fradafiban suppresses platelet aggregation and may be a useful alternative to aspirin in the prevention of thrombotic events in patients undergoing PTCA. However, there is continued formation of thromboxane A2, which may continue to exert its effects as a potent vasoconstrictor and vascular smooth muscle mitogen. Topics: Administration, Oral; Angioplasty, Balloon, Coronary; Anticoagulants; Aspirin; Biphenyl Compounds; Bleeding Time; Cell Division; Coronary Disease; Dose-Response Relationship, Drug; Double-Blind Method; Drug Therapy, Combination; Female; Fibrinogen; Heparin; Humans; Infusions, Intravenous; Male; Middle Aged; Muscle, Smooth, Vascular; P-Selectin; Pilot Projects; Platelet Aggregation; Platelet Aggregation Inhibitors; Platelet Glycoprotein GPIIb-IIIa Complex; Pyrrolidines; Thrombosis; Thromboxane A2; Thromboxane B2; Vasoconstriction | 1997 |
The mechanism of anti-thrombotic, thrombolytic and fibrinolytic actions of camonagrel--a new thromboxane synthase inhibitor.
So far pharmacological consequences of inhibition of thromboxane A2 (TXA2) synthase by imidazole derivatives (e.g., camonagrel or dazoxiben) were linked to suppression of platelet activity. Here we report that in patients with peripheral atherosclerosis or in cats with extracorporeal thrombogenesis treatment with camonagrel is associated with activation of fibrinolysis or thrombolysis. These phenomena seem to be related to the camonagrel-induced shift in metabolism of prostaglandin endoperoxides from TXA2 to prostacyclin (PGI2), although in an in vitro model the involvement of the L-arginine/nitric oxide pathway cannot be excluded. In cats camonagrel (10 mg/kg i.v.) produced not only a fall in TXB2 but also a rise in 6-keto-PGF1 alpha and no change in cyclic-GMP plasma levels. This points to PGI2 rather than to nitric oxide as an in vivo mediator of camonagrel-induced thrombolysis. The crucial role of endogenous PGI2 in the thrombolytic response to camonagrel in cats was evidenced by its blockade following pretreatment of animals with a megadose of aspirin (50 mg/kg i.v.) and lack of any effect on pretreatment with L-NAME (100 micrograms/kg/min, i.v.). Obviously TXA2 synthase inhibitors (e.g., camonagrel) and cyclo-oxygenase inhibitors (e.g., aspirin) antagonize each other in their anti-thrombotic actions and must not be administered at the same time. Furthermore, in patients camonagrel (800 mg orally) suppressed TXA2 generation by 99.5% and doubled the plasma level of 6-keto-PGF1 alpha.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Arteriosclerosis; Blood Coagulation Tests; Cats; Dose-Response Relationship, Drug; Double-Blind Method; Endothelium, Vascular; Female; Fibrinolysis; Humans; Imidazoles; Male; Middle Aged; Platelet Aggregation; Thrombosis; Thromboxane A2; Thromboxane-A Synthase; Ultrasonography, Doppler | 1995 |
A dose-ranging study of the antiplatelet effect of enteric coated aspirin in man.
Enteric coated aspirin was given to eight human volunteers in escalating doses (20, 40, 60, 80, 100 mg daily), each dose being given over two weeks. In addition, to measure the maximum effect of aspirin, each volunteer was given two single doses of 600 mg of soluble aspirin. At the end of each dosing interval we measured platelet aggregation and thromboxane formation in response to four aggregating agents and to whole blood coagulation. The doses of aspirin required to inhibit platelet aggregation in response to various stimuli were: for collagen 60-80 mg, for adenosine diphosphate and adrenaline 60 mg, and for arachidonate 40 mg. For maximum inhibition of thromboxane formation the doses were: for collagen greater than 100 mg, for adenosine diphosphate and adrenaline 60 mg, for arachidonate 80 mg, and for whole blood coagulation 100 mg. Different aspirin doses are required to inhibit the responses to different stimuli. Furthermore, for some stimuli, inhibition of thromboxane generation may require more aspirin than is required for inhibition of aggregation. The clinical implications of these findings are uncertain since we do not know which stimuli are important in arterial thrombosis in man. Topics: Adult; Aspirin; Dose-Response Relationship, Drug; Female; Humans; Male; Platelet Aggregation; Thrombosis; Thromboxane A2; Thromboxane B2 | 1987 |
Inhibition of prostacyclin and thromboxane A2 generation by low-dose aspirin at the site of plug formation in man in vivo.
In a double-blind placebo-controlled crossover study, we investigated in seven healthy male volunteers the effect of a low-dose aspirin regimen (35 mg acetylsalicylate per day for 7 days) on the formation of thromboxane A2 (TxA2) and prostacyclin (PGI2) in blood emerging from a standardized injury of the microvasculature made to determine skin bleeding time. When subjects were treated with placebo, there was rapid and substantial generation of TxA2 and PGI2 at the site of platelet-vessel wall interaction within the first 2 min after vascular injury. This was reflected by a greater than 100-fold and greater than 10-fold increase in thromboxane B2 (TxB2) and 6-keto-prostaglandin F1 alpha (6-keto-PGF1 alpha) in blood obtained from incisions made to determine bleeding time as compared with the corresponding plasma values. Low-dose aspirin caused a significant inhibition of both TxA2 and PGI2 generation in blood sampled from the skin incisions, represented by a 85% and 92% and 81% and 84% inhibition of TxB2 and 6-keto-PGF1 alpha, respectively, as compared with controls. We therefore conclude that rapid activation of both platelet prostaglandin metabolism and vascular PGI2 biosynthesis occurs at the site of platelet-vessel wall interaction, and low-dose aspirin results in a significant inhibition of both platelet and vascular cyclooxygenase activity. Thus, our data fail to confirm the concept of a differential effect of low-dose aspirin on platelet and vascular prostaglandin synthesis in man in vivo. Topics: 6-Ketoprostaglandin F1 alpha; Adult; Aspirin; Bleeding Time; Blood Platelets; Blood Vessels; Double-Blind Method; Epoprostenol; Hemostasis; Humans; Male; Thrombosis; Thromboxane A2; Thromboxane B2 | 1987 |
Rationale for the use of drugs inhibiting platelet function in claudicating patients with atherosclerotic leg arteries.
Topics: Anticoagulants; Arteriosclerosis; beta-Thromboglobulin; Blood Platelets; Clinical Trials as Topic; Humans; Intermittent Claudication; Leg; Platelet Aggregation; Platelet Factor 4; Suloctidil; Thiophenes; Thrombosis; Thromboxane A2; Ticlopidine | 1984 |
127 other study(ies) available for thromboxane-a2 and Thrombosis
Article | Year |
---|---|
Exploring the mechanism of the antithrombotic effects of Pueraria lobata and Pueraria lobata var. thomsonii based on network pharmacology.
Pueraria lobata (Willd.) Ohwi and Pueraria lobata var. thomsonii (Benth.) Maesen are nutritious medicine food homology plants that are widely used in the food and health products industry and are excellent natural materials for the development of new health foods, with great potential for domestic and foreign markets. Clinically, P. lobata and P. thomsonii are used to treat coronary heart disease, atherosclerosis, cerebral infarction and other cardiovascular diseases, and antithrombotic actions may be their core effect in the treatment of thrombotic diseases. However, the underlying mechanisms of the antithrombotic properties of P. lobata and P. thomsonii have not been clarified.. First, P. lobata and P. thomsonii were identified by high-performance liquid chromatography (HPLC). An arteriovenous bypass thrombosis rat model was established. Thrombus dry‒wet weight, platelet accumulation rate and the four coagulation indices, including activated partial thromboplastin time (APTT), prothrombin time (PT), thrombin time (TT) and fibrinogen (FIB), were detected in plasma to manifest the P. lobata and P. thomsonii antithrombotic function. Network pharmacology and molecular docking methods were used to obtain key targets and verify reliability. David 6.8 was used for GO and KEGG analyses to explore pathways and potential targets for P. lobata and P. thomsonii antithrombotic functions. Prostaglandin I2 (PGI2), thromboxane A2 (TXA2), cyclooxygenase 2 (COX-2), myeloperoxidase (MPO) and endothelial nitric oxide synthase (eNOS) were tested by enzyme-linked immunosorbent assay (ELISA).. The results indicated that P. lobata and P. thomsonii can reduce thrombus dry‒wet weight and platelet accumulation in rats and inhibit TT, APTT, FIB, and PT. A comprehensive network pharmacology approach successfully identified 9 active ingredients in P. lobata and P. thomsonii. The main active ingredients include polyphenols, amino acids and flavonoids. A total of 15 antithrombotic function targets were obtained, including 3 key targets (PTGS2, NOS3, MPO). Pathway analysis showed 10 significant related pathways and 29 biological processes. P. lobata and P. thomsonii inhibited platelet aggregation by upregulating PGI2 and downregulating TXA2, inhibited PTGS2 to reduce inflammation, and increased the level of eNOS to promote vasodilation. In addition, P. lobata and P. thomsonii alleviated oxidative stress by increasing SOD levels and significantly decreasing MDA contents.. The results of the study further clarify the antithrombotic mechanism of action of P. lobata and P. thomsonii, which provides a scientific basis for the development of new drugs for thrombogenic diseases and lays the foundation for the development of P. lobata and P. thomsonii herbal resources and P. lobata and P. thomsonii health products. Topics: Amino Acids; Animals; Cyclooxygenase 2; Epoprostenol; Fibrinogen; Fibrinolytic Agents; Flavonoids; Molecular Docking Simulation; Network Pharmacology; Nitric Oxide Synthase Type III; Peroxidase; Pueraria; Rats; Reproducibility of Results; Superoxide Dismutase; Thrombosis; Thromboxane A2 | 2023 |
Resveratrol Inhibits Metabolism and Affects Blood Platelet Function in Type 2 Diabetes.
Chronic hyperglycemia contributes to vascular complications in diabetes. Resveratrol exerts anti-diabetic and anti-platelet action. This study aimed to evaluate the effects of resveratrol on metabolism and the function of blood platelets under static and in in vitro flow conditions in patients with type 2 diabetes. Blood obtained from 8 healthy volunteers and 10 patients with type 2 diabetes was incubated with resveratrol and perfused over collagen-coated capillaries. Isolated blood platelets were incubated with resveratrol and activated by collagen to assess platelet function, metabolism, ATP release, TXA Topics: Blood Platelets; Cardiovascular Diseases; Collagen; Diabetes Mellitus, Type 2; Humans; Platelet Aggregation; Resveratrol; Thrombosis; Thromboxane A2 | 2022 |
Correlation of platelet-derived growth factor and thromboxane A2 expression with platelet parameters and coagulation indices in chronic altitude sickness patients.
What is the central question of this study? Is the expression of platelet-derived growth factor (PDGF) and thromboxane A2 (TXA2) elevated in chronic altitude patients, and are they related to thrombosis in chronic mountain sickness? What is the main finding and its importance? The expression of PDGF and TXA2 in both the bone marrow and the peripheral blood of patients with chronic mountain sickness is elevated, and they are considered to be correlated in the mechanism of thrombosis in the chronic mountain sickness.. The purpose of this study was to evaluate the expression of platelet-derived growth factor (PDGF) and thromboxane A2 (TXA2) along with platelet parameters and coagulation indices in chronic mountain sickness (CMS) patients and healthy individuals on the Qinghai-Tibet Plateau. The levels of PDGF and TXA2 were examined in 22 CMS patients (age, 52.77 ± 9.92 years, haemoglobin, 219 ± 13 g/l) and 25 healthy individuals (age, 47.80 ± 9.78 years, haemoglobin, 146 ± 18 g/l), and the association between platelet parameters and coagulation indices was investigated. Mean platelet volume and fibrinogen degradation product were higher in the CMS compared to the control group (10.58 ± 0.83 vs. 8.92 ± 1.61, 7.50 ± 2.15 vs. 4.40 ± 2.51), platelet count and plateletcrit were lower in the CMS compared to the control group (0.13 (0.80, 0.16) vs. 0.23 (0.18, 0.24), 109 ± 46 vs. 204 ± 86). The levels of PDGF and TXA2 in the bone marrow and peripheral blood of CMS patients were higher (P < 0.01) in comparison to the control group. The two factors had no statistically significant relationship with platelet parameters or coagulation indices (P > 0.159). According to the current findings, platelets in CMS patients were activated, resulting in aberrant coagulation and PDGF and TXA2 expression, which could be due to physiological adjustments to the plateau's high altitude. To summarize, PDGF and TXA2 levels in CMS patients were not correlated with coagulation or platelet parameters, implying that the mechanism behind their increased expression warrants additional investigation. Topics: Adult; Altitude; Altitude Sickness; Chronic Disease; Hemoglobins; Humans; Middle Aged; Platelet-Derived Growth Factor; Thrombosis; Thromboxane A2 | 2022 |
Redirecting thromboxane A
Topics: Animals; Catalysis; Epoprostenol; Fibrinolytic Agents; Humans; Models, Animal; Prostaglandin-Endoperoxide Synthases; Recombinant Proteins; Thrombosis; Thromboxane A2 | 2021 |
Aspirin blocks formation of metastatic intravascular niches by inhibiting platelet-derived COX-1/thromboxane A2.
Because metastasis is associated with the majority of cancer-related deaths, its prevention is a clinical aspiration. Prostanoids are a large family of bioactive lipids derived from the activity of cyclooxygenase-1 (COX-1) and COX-2. Aspirin impairs the biosynthesis of all prostanoids through the irreversible inhibition of both COX isoforms. Long-term administration of aspirin leads to reduced distant metastases in murine models and clinical trials, but the COX isoform, downstream prostanoid, and cell compartment responsible for this effect are yet to be determined. Here, we have shown that aspirin dramatically reduced lung metastasis through inhibition of COX-1 while the cancer cells remained intravascular and that inhibition of platelet COX-1 alone was sufficient to impair metastasis. Thromboxane A2 (TXA2) was the prostanoid product of COX-1 responsible for this antimetastatic effect. Inhibition of the COX-1/TXA2 pathway in platelets decreased aggregation of platelets on tumor cells, endothelial activation, tumor cell adhesion to the endothelium, and recruitment of metastasis-promoting monocytes/macrophages, and diminished the formation of a premetastatic niche. Thus, platelet-derived TXA2 orchestrates the generation of a favorable intravascular metastatic niche that promotes tumor cell seeding and identifies COX-1/TXA2 signaling as a target for the prevention of metastasis. Topics: Animals; Anti-Inflammatory Agents, Non-Steroidal; Aspirin; Blood Platelets; Cell Line, Tumor; Cyclooxygenase Inhibitors; Female; Humans; Lung Neoplasms; Macrophages; Melanoma, Experimental; Mice; Mice, Inbred BALB C; Mice, Inbred C57BL; Monocytes; Neoplasm Metastasis; Neoplasm Transplantation; Platelet Aggregation; Platelet Aggregation Inhibitors; Prostaglandins; Protein Isoforms; Thrombosis; Thromboxane A2 | 2019 |
Phosphoinositide 3-kinase p110α negatively regulates thrombopoietin-mediated platelet activation and thrombus formation.
Phosphoinositide 3-kinase (PI3K) plays an important role in platelet function and contributes to platelet hyperreactivity induced by elevated levels of circulating peptide hormones, including thrombopoietin (TPO). Previous work established an important role for the PI3K isoform; p110β in platelet function, however the role of p110α is still largely unexplored. Here we sought to investigate the role of p110α in TPO-mediated hyperactivity by using a conditional p110α knockout (KO) murine model in conjunction with platelet functional assays. We found that TPO-mediated enhancement of collagen-related peptide (CRP-XL)-induced platelet aggregation and adenosine triphosphate (ATP) secretion were significantly increased in p110α KO platelets. Furthermore, TPO-mediated enhancement of thrombus formation by p110α KO platelets was elevated over wild-type (WT) platelets, suggesting that p110α negatively regulates TPO-mediated priming of platelet function. The enhancements were not due to increased flow through the PI3K pathway as phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P Topics: Animals; Blood Platelets; Mice; Mice, Knockout; p38 Mitogen-Activated Protein Kinases; Phosphatidylinositol 3-Kinase; Phosphorylation; Platelet Activation; Platelet Aggregation; Protein Kinase Inhibitors; Signal Transduction; Thrombopoietin; Thrombosis; Thromboxane A2 | 2018 |
TxA
Topics: Humans; Receptors, Thromboxane A2, Prostaglandin H2; Thrombosis; Thromboxane A2; Vaccination; Vaccines | 2018 |
Receptor-interacting protein kinase 3 promotes platelet activation and thrombosis.
Previous studies have shown that receptor-interacting protein kinase 3 (RIP3) is involved in many important biological processes, including necroptosis, apoptosis, and inflammation. Here we show that RIP3 plays a critical role in regulating platelet functions and in vivo thrombosis and hemostasis. Tail bleeding times were significantly longer in Topics: Adenosine Diphosphate; Animals; Blood Platelets; Disease Models, Animal; Gene Expression; Hemostasis; Humans; Mice; Mice, Knockout; Phosphatidylserines; Phosphorylation; Platelet Activation; Platelet Aggregation; Proto-Oncogene Proteins c-akt; Receptor-Interacting Protein Serine-Threonine Kinases; Signal Transduction; Thrombin; Thrombosis; Thromboxane A2 | 2017 |
Apelin: an antithrombotic factor that inhibits platelet function.
Apelin peptide and its receptor APJ are directly implicated in various physiological processes ranging from cardiovascular homeostasis to immune signaling. Here, we show that apelin is a key player in hemostasis with an ability to inhibit thrombin- and collagen-mediated platelet activation. Mice lacking apelin displayed a shorter bleeding time and a prothrombotic profile. Their platelets exhibited increased adhesion and a reduced occlusion time in venules, and displayed a higher aggregation rate after their activation by thrombin compared with wild-type platelets. Consequently, human and mouse platelets express apelin and its receptor APJ. Apelin directly interferes with thrombin-mediated signaling pathways and platelet activation, secretion, and aggregation, but not with ADP and thromboxane A2-mediated pathways. IV apelin administration induced excessive bleeding and prevented thrombosis in mice. Taken together, these findings suggest that apelin and/or APJ agonists could potentially be useful adducts in antiplatelet therapies and may provide a promising perspective for patients who continue to display adverse thrombotic events with current antiplatelet therapies. Topics: Adipokines; Animals; Apelin; Apelin Receptors; Blood Platelets; Hemorrhage; Humans; Intercellular Signaling Peptides and Proteins; Mice; Mice, Knockout; Platelet Adhesiveness; Receptors, G-Protein-Coupled; Signal Transduction; Thrombin; Thrombosis; Thromboxane A2 | 2016 |
ATP-binding cassette transporter 1 (ABCA1) deficiency decreases platelet reactivity and reduces thromboxane A2 production independently of hematopoietic ABCA1.
ESSENTIALS: The role of ATP-binding cassette transporter 1 (ABCA1) in platelet functions is poorly characterized. We studied the impact of ABCA1 deficiency on platelet responses in a mouse model and two Tangier patients. ABCA1-deficient platelets exhibit reduced positive feedback loop mechanisms. This reduced reactivity is dependent on external environment and independent of hematopoietic ABCA1.. The ATP-binding cassette transporter ABCA1 is required for the conversion of apolipoprotein A-1 to high-density lipoprotein (HDL), and its defect causes Tangier disease, a rare disorder characterized by an absence of HDL and accumulation of cholesterol in peripheral tissues. The role of ABCA1 in platelet functions remains poorly characterized.. To determine the role of ABCA1 in platelet functions and to clarify controversies concerning its implication in processes as fundamental as platelet phosphatidylserine exposure and control of platelet membrane lipid composition.. We studied the impact of ABCA1 deficiency on platelet responses in a mouse model and in two Tangier patients. We show that platelets in ABCA1-deficient mice are slightly larger in size and exhibit aggregation and secretion defects in response to low concentrations of thrombin and collagen. These platelets have normal cholesterol and major phospholipid composition, granule morphology, or calcium-induced phosphatidylserine exposure. Interestingly, ABCA1-deficient platelets display a reduction in positive feedback loop mechanisms, particularly in thromboxane A2 (TXA2) production. Hematopoietic chimera mice demonstrated that defective eicosanoids production, particularly TXA2, was primarily dependent on external environment and not on the hematopoietic ABCA1. Decreased aggregation and production of TXA2 and eicosanoids were also observed in platelets from Tangier patients.. Absence of ABCA1 and low HDL level induce reduction of platelet reactivity by decreasing positive feedback loops, particularly TXA2 production through a hematopoietic ABCA1-independent mechanism. Topics: Animals; ATP Binding Cassette Transporter 1; Blood Platelets; Cell Size; Disease Models, Animal; Feedback, Physiological; Female; Genetic Predisposition to Disease; Hematopoietic Stem Cell Transplantation; Hematopoietic Stem Cells; Hemostasis; Humans; Lipoproteins, HDL; Male; Mice, Inbred DBA; Mice, Knockout; Middle Aged; Phenotype; Platelet Aggregation; Tangier Disease; Thrombosis; Thromboxane A2; Time Factors | 2016 |
A novel thromboxane receptor antagonist, nstpbp5185, inhibits platelet aggregation and thrombus formation in animal models.
A novel benzimidazole derivative, nstpbp5185, was discovered through in vitro and in vivo evaluations for antiplatelet activity. Thromaboxane receptor (TP) is important in vascular physiology, haemostasis and pathophysiological thrombosis. Nstpbp5185 concentration-dependently inhibited human platelet aggregation caused by collagen, arachidonic acid and U46619. Nstpbp5185 caused a right-shift of the concentration-response curve of U46619 and competitively inhibited the binding of 3H-SQ-29548 to TP receptor expressed on HEK-293 cells, with an IC50 of 0.1 µM, indicating that nstpbp5185 is a TP antagonist. In murine thrombosis models, nstpbp5185 significantly prolonged the latent period in triggering platelet plug formation in mesenteric and FeCl3-induced thrombi formation, and increased the survival rate in pulmonary embolism model with less bleeding than aspirin. This study suggests nstpbp5185, an orally selective anti-thrombotic agent, acting through blockade of TXA2 receptor, may be efficacious for prevention or treatment of pathologic thrombosis. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Animals; Arachidonic Acid; Aspirin; Benzimidazoles; Blood Platelets; Calcium; Collagen; Humans; In Vitro Techniques; Male; Mice; Mice, Inbred ICR; Microvessels; P-Selectin; Platelet Aggregation; Pulmonary Embolism; Receptors, Thromboxane A2, Prostaglandin H2; Thrombosis; Thromboxane A2 | 2016 |
Evaluation on antithrombotic effect of aspirin eugenol ester from the view of platelet aggregation, hemorheology, TXB2/6-keto-PGF1α and blood biochemistry in rat model.
Based on the prodrug principle, aspirin and eugenol, as starting precursors, were esterified to synthesize aspirin eugenol ester (AEE). The aim of the present study was to evaluate the antithrombotic effect of AEE in an animal disease model. In order to compare the therapeutic effects of AEE and its precursors, aspirin, eugenol and a combination of aspirin and eugenol were designed at the same molar quantities as the AEE medium dose in the control group.. After oral administration of AEE (dosed at 18, 36 and 72 mg/kg) for seven days, rats were treated with k-carrageenan to induce tail thrombosis. Following the same method, aspirin (20 mg/kg), eugenol (18 mg/kg) and 0.5 % CMC-Na (30 mg/kg) were administered as control drug. Different drug effects on platelet aggregation, hemorheology, TXB2/6-keto-PGF1α ratio and blood biochemistry were studied.. AEE significantly inhibited ADP and AA-induced platelet aggregation in vivo. AEE also significantly reduced blood and plasma viscosity. Moreover, AEE down-regulated TXB2 and up-regulated 6-keto-PGF1α, normalizing the TXB2/6-keto-PGF1α ratio and blood biochemical profile. In comparison with aspirin and eugenol, AEE produced more positive therapeutic effects than its precursors under the same molar quantity.. It may be concluded that AEE was a good candidate for new antithrombotic and antiplatelet medicine. Additionally, this study may help to understand how AEE works on antithrombosis in different ways. Topics: 6-Ketoprostaglandin F1 alpha; Administration, Oral; Animals; Aspirin; Blood Chemical Analysis; Eugenol; Fibrinolytic Agents; Hemorheology; Male; Platelet Aggregation; Rats; Rats, Wistar; Thrombosis; Thromboxane A2 | 2016 |
SDF-1α is a novel autocrine activator of platelets operating through its receptor CXCR4.
Platelets store and secrete the chemokine stromal cell-derived factor (SDF)-1α upon platelet activation, but the ability of platelet-derived SDF-1α to signal in an autocrine/paracrine manner mediating functional platelet responses relevant to thrombosis and haemostasis is unknown. We sought to explore the role of platelet-derived SDF-1α and its receptors, CXCR4 and CXCR7 in facilitating platelet activation and determine the mechanism facilitating SDF-1α-mediated regulation of platelet function. Using human washed platelets, CXCR4 inhibition, but not CXCR7 blockade significantly abrogated collagen-mediated platelet aggregation, dense granule secretion and thromboxane (Tx) A2 production. Time-dependent release of SDF-1α from collagen-activated platelets supports a functional role for SDF-1α in this regard. Using an in vitro whole blood perfusion assay, collagen-induced thrombus formation was substantially reduced with CXCR4 inhibition. In washed platelets, recombinant SDF-1α in the range of 20-100 ng/mL(-1) could significantly enhance platelet aggregation responses to a threshold concentration of collagen. These enhancements were completely dependent on CXCR4, but not CXCR7, which triggered TxA2 production and dense granule secretion. Rises in cAMP were significantly blunted by SDF-1α, which could also enhance collagen-mediated Ca2+ mobilisation, both of which were mediated by CXCR4. This potentiating effect of SDF-1α primarily required TxA2 signalling acting upstream of dense granule secretion, whereas blockade of ADP signalling could only partially attenuate SDF-1α-induced platelet activation. Therefore, this study supports a potentially novel autocrine/paracrine role for platelet-derived SDF-1α during thrombosis and haemostasis, through a predominantly TxA2-dependent and ADP-independent pathway. Topics: Adenosine Diphosphate; Animals; Autocrine Communication; Blood Platelets; Calcium Signaling; Chemokine CXCL12; Collagen; Cyclic AMP; Horses; Humans; Platelet Activation; Platelet Aggregation; Receptors, CXCR4; Thrombosis; Thromboxane A2 | 2015 |
Salivary Thromboxane A2-Binding Proteins from Triatomine Vectors of Chagas Disease Inhibit Platelet-Mediated Neutrophil Extracellular Traps (NETs) Formation and Arterial Thrombosis.
The saliva of blood-feeding arthropods contains a notable diversity of molecules that target the hemostatic and immune systems of the host. Dipetalodipin and triplatin are triatomine salivary proteins that exhibit high affinity binding to prostanoids, such as TXA2, thus resulting in potent inhibitory effect on platelet aggregation in vitro. It was recently demonstrated that platelet-derived TXA2 mediates the formation of neutrophil extracellular traps (NETs), a newly recognized link between inflammation and thrombosis that promote thrombus growth and stability.. This study evaluated the ability of dipetalodipin and triplatin to block NETs formation in vitro. We also investigated the in vivo antithrombotic activity of TXA2 binding proteins by employing two murine models of experimental thrombosis. Remarkably, we observed that both inhibitors abolished the platelet-mediated formation of NETs in vitro. Dipetalodipin and triplatin significantly increased carotid artery occlusion time in a FeCl3-induced injury model. Treatment with TXA2-binding proteins also protected mice from lethal pulmonary thromboembolism evoked by the intravenous injection of collagen and epinephrine. Effective antithrombotic doses of dipetalodipin and triplatin did not increase blood loss, which was estimated using the tail transection method.. Salivary TXA2-binding proteins, dipetalodipin and triplatin, are capable to prevent platelet-mediated NETs formation in vitro. This ability may contribute to the antithrombotic effects in vivo. Notably, both molecules inhibit arterial thrombosis without promoting excessive bleeding. Our results provide new insight into the antihemostatic effects of TXA2-binding proteins and may have important significance in elucidating the mechanisms of saliva to avoid host's hemostatic responses and innate immune system. Topics: Animals; Arthropod Proteins; Blood Platelets; Chagas Disease; Extracellular Traps; Female; Humans; Insect Vectors; Male; Mice; Mice, Inbred BALB C; Neutrophils; Platelet Aggregation; Platelet Aggregation Inhibitors; Protein Binding; Recombinant Proteins; Salivary Proteins and Peptides; Thrombosis; Thromboxane A2; Triatominae | 2015 |
Sphingosine 1-Phosphate Produced by Sphingosine Kinase 2 Intrinsically Controls Platelet Aggregation In Vitro and In Vivo.
Platelets are known to play a crucial role in hemostasis. Sphingosine kinases (Sphk) 1 and 2 catalyze the conversion of sphingosine to the bioactive metabolite sphingosine 1-phosphate (S1P). Although platelets are able to secrete S1P on activation, little is known about a potential intrinsic effect of S1P on platelet function.. To investigate the role of Sphk1- and Sphk2-derived S1P in the regulation of platelet function.. We found a 100-fold reduction in intracellular S1P levels in platelets derived from Sphk2(-/-) mutants compared with Sphk1(-/-) or wild-type mice, as analyzed by mass spectrometry. Sphk2(-/-) platelets also failed to secrete S1P on stimulation. Blood from Sphk2-deficient mice showed decreased aggregation after protease-activated receptor 4-peptide and adenosine diphosphate stimulation in vitro, as assessed by whole blood impedance aggregometry. We revealed that S1P controls platelet aggregation via the sphingosine 1-phosphate receptor 1 through modulation of protease-activated receptor 4-peptide and adenosine diphosphate-induced platelet activation. Finally, we show by intravital microscopy that defective platelet aggregation in Sphk2-deficient mice translates into reduced arterial thrombus stability in vivo.. We demonstrate that Sphk2 is the major Sphk isoform responsible for the generation of S1P in platelets and plays a pivotal intrinsic role in the control of platelet activation. Correspondingly, Sphk2-deficient mice are protected from arterial thrombosis after vascular injury, but have normal bleeding times. Targeting this pathway could therefore present a new therapeutic strategy to prevent thrombosis. Topics: Animals; Arachidonic Acid; Blood Coagulation; Blood Coagulation Tests; Blood Platelets; Carotid Artery Injuries; Disease Models, Animal; Erythrocytes; Lysophospholipids; Mice, Inbred BALB C; Mice, Inbred C57BL; Mice, Knockout; Phosphotransferases (Alcohol Group Acceptor); Platelet Adhesiveness; Platelet Aggregation; Platelet Function Tests; Receptors, Lysosphingolipid; Signal Transduction; Sphingosine; Sphingosine-1-Phosphate Receptors; Thrombosis; Thromboxane A2; Vascular System Injuries | 2015 |
Acetic acid in aged vinegar affects molecular targets for thrombus disease management.
To elucidate the mechanism underlying the action of dietary vinegar on antithrombotic activity, acetic acid, the main acidic component of dietary vinegar, was used to determine antiplatelet and fibrinolytic activity. The results revealed that acetic acid significantly inhibits adenosine diphosphate (ADP)-, collagen-, thrombin-, and arachidonic acid (AA)-induced platelet aggregation. Acetic acid (2.00 mM) reduced AA-induced platelet aggregation to approximately 36.82 ± 1.31%, and vinegar (0.12 mL L(-1)) reduced the platelet aggregation induced by AA to 30.25 ± 1.34%. Further studies revealed that acetic acid exerts its effects by inhibiting cyclooxygenase-1 and the formation of thromboxane-A2. Organic acids including acetic acid, formic acid, lactic acid, citric acid, and malic acid also showed fibrinolytic activity; specifically, the fibrinolytic activity of acetic acid amounted to 1.866 IU urokinase per mL. Acetic acid exerted its fibrinolytic activity by activating plasminogen during fibrin crossing, thus leading to crosslinked fibrin degradation by the activated plasmin. These results suggest that organic acids in dietary vinegar play important roles in the prevention and cure of cardiovascular diseases. Topics: Acetic Acid; Adult; Animals; Blood Platelets; Cyclooxygenase 1; Disease Management; Female; Humans; Male; Middle Aged; Platelet Aggregation; Platelet Aggregation Inhibitors; Rabbits; Thrombin; Thrombosis; Thromboxane A2; Young Adult | 2015 |
The antithrombotic effect of RSNK in blood-stasis model rats.
Reduction of Sheng-Nao-Kang decoction (RSNK), composed of Salvia miltiorrhiza Bge., Ligusticum chuanxiong Hort., Astragalus membranaceus (Fisch.) Bunge., Pueraria lobata (Willd.) Ohwi., Paeonia lactiflora Pall. and Panax notoginseng (Burk.) F. H. Chen., is a modified traditional Chinese medicinal formula of Sheng-Nao-Kang pill preparation, which has been investigated its protective effect on focal cerebral ischemia-reperfusion injury in rat in our previous report.. To evaluate the antithrombotic effect of RSNK in blood stasis model rats and explore the potential mechanisms.. Subcutaneous injection of norepinephrine and bovine serum albumin combined with ice water bath was used to establish the acute blood stasis rat model. The anticoagulant activities were investigated by measuring activated partial thromboplastin time (APTT), thrombin time (TT), prothrombin time (PT), and the content of fibrinogen (FIB). Meanwhile, the levels of thromboxane A2 (TXA2), prostaglandins I2 (PGI2), endothelial nitric oxide synthase (eNOS) and endothelin (ET) were detected.. The treatment of RSNK was able to prolong APTT, TT and PT, and decrease FIB content obviously. Furthermore, it markedly suppressed TXB2 level and up-regulated 6-keto-PGF1α level of the blood-stasis model rats, accompanied with the decrease of T/K. The level of ET and TXA2 in plasma was down-regulated and the levels of eNOS in plasma and PGI2 in serum was up-regulated in RSNK-treated rats compared with model rats (P<0.05).. The present study suggested that RSNK possessed remarkable antithrombotic property in blood stasis model rats induced by ice water bath and subcutaneous injection of norepinephrine and bovine serum albumin. This property could be associated with its anticoagulation activity, the regulation of active substances in vascular endothelium and maintaining the balance of TXA2 and PGI2. Topics: 6-Ketoprostaglandin F1 alpha; Abietanes; Animals; Anticoagulants; Blood Coagulation; Blood Coagulation Tests; Caffeic Acids; Carotid Arteries; Catechols; Cold Temperature; Disease Models, Animal; Drugs, Chinese Herbal; Endothelins; Male; Nitric Oxide Synthase Type III; Norepinephrine; Phytotherapy; Rats, Sprague-Dawley; Serum Albumin, Bovine; Thrombosis; Thromboxane A2 | 2015 |
N2 extenuates experimental ischemic stroke through platelet aggregation inhibition.
Thromboxane A2 (TXA2) can induce the platelet aggregation and lead to thrombosis. This will cause the low-reflow phenomenon after ischemic stroke and aggravate the damage of brain issues. Therefore, it is potential to develop the drugs inhibiting TXA2 pathway to treat cerebral ischemia.. This study aims to prove the protective effect of N2 (4-(2-(1H-imidazol-1-yl) ethoxy)-3-methoxybenzoic acid) on focal cerebral ischemia and reperfusion injury through platelet aggregation inhibition.. Middle cerebral artery occlusion/reperfusion (MCAO/R) is used as the animal model. Neurological deficit score, Morris water maze, postural reflex test, Limb-use asymmetry test, infarct volume, and water content were performed to evaluate the protective effect of N2 in MCAO/R rats. 9, 11-dieoxy-11α, 9α-methanoepoxyprostaglandin F2α (U46619) or adenosine diphosphate (ADP) was used as the inducer of platelet aggregation.. N2 can improve the motor function, learning and memory ability in MCAO/R rats while reducing the infarct volume. N2 can inhibit TXA2 formation but promote PGI2, and can inhibit platelet aggregation induced by U46619 and ADP. Further, N2 inhibits thrombosis with a minor adverse effect of bleeding than Clopidogrel. In conclusion, N2 can produce the protective effect on MCAO/R brain injury through inhibiting TXA2 formation, platelet aggregation and thrombosis. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Adenosine Diphosphate; Animals; Arteriovenous Shunt, Surgical; Blood Coagulation; Brain; Brain Ischemia; Edema; Enzyme-Linked Immunosorbent Assay; Epoprostenol; Female; Imidazoles; Male; Maze Learning; Platelet Aggregation; Rats; Rats, Sprague-Dawley; Stroke; Thrombosis; Thromboxane A2; Vanillic Acid | 2015 |
The role of Nox1 and Nox2 in GPVI-dependent platelet activation and thrombus formation.
Activation of the platelet-specific collagen receptor, glycoprotein (GP) VI, induces intracellular reactive oxygen species (ROS) production; however the relevance of ROS to GPVI-mediated platelet responses remains unclear.. The objective of this study was to explore the role of the ROS-producing NADPH oxidase (Nox)1 and 2 complexes in GPVI-dependent platelet activation and collagen-induced thrombus formation.. ROS production was measured by quantitating changes in the oxidation-sensitive dye, H2DCF-DA, following platelet activation with the GPVI-specific agonist, collagen related peptide (CRP). Using a pharmacological inhibitor specific for Nox1, 2-acetylphenothiazine (ML171), and Nox2 deficient mice, we show that Nox1 is the key Nox homolog regulating GPVI-dependent ROS production. Nox1, but not Nox2, was essential for CRP-dependent thromboxane (Tx)A2 production, which was mediated in part through p38 MAPK signaling; while neither Nox1 nor Nox2 was significantly involved in regulating CRP-induced platelet aggregation/integrin αIIbβ3 activation, platelet spreading, or dense granule and α-granule release (ATP release and P-selectin surface expression, respectively). Ex-vivo perfusion analysis of mouse whole blood revealed that both Nox1 and Nox2 were involved in collagen-mediated thrombus formation at arterial shear.. Together these results demonstrate a novel role for Nox1 in regulating GPVI-induced ROS production, which is essential for optimal p38 activation and subsequent TxA2 production, providing an explanation for reduced thrombus formation following Nox1 inhibition. Topics: Animals; Arteries; Carrier Proteins; Fluoresceins; MAP Kinase Signaling System; Membrane Glycoproteins; Mice; NADH, NADPH Oxidoreductases; NADPH Oxidase 1; NADPH Oxidase 2; NADPH Oxidases; Peptides; Phenothiazines; Platelet Activation; Platelet Membrane Glycoproteins; Reactive Oxygen Species; Thrombosis; Thromboxane A2 | 2014 |
Group VIB calcium-independent phospholipase A2 (iPLA2γ) regulates platelet activation, hemostasis and thrombosis in mice.
In platelets, group IVA cytosolic phospholipase A2 (cPLA2α) has been implicated as a key regulator in the hydrolysis of platelet membrane phospholipids, leading to pro-thrombotic thromboxane A2 and anti-thrombotic 12-(S)-hydroxyeicosatetranoic acid production. However, studies using cPLA2α-deficient mice have indicated that other PLA2(s) may also be involved in the hydrolysis of platelet glycerophospholipids. In this study, we found that group VIB Ca2+-independent PLA2 (iPLA2γ)-deficient platelets showed decreases in adenosine diphosphate (ADP)-dependent aggregation and ADP- or collagen-dependent thromboxane A2 production. Electrospray ionization mass spectrometry analysis of platelet phospholipids revealed that fatty acyl compositions of ethanolamine plasmalogen and phosphatidylglycerol were altered in platelets from iPLA2γ-null mice. Furthermore, mice lacking iPLA2γ displayed prolonged bleeding times and were protected against pulmonary thromboembolism. These results suggest that iPLA2γ is an additional, long-sought-after PLA2 that hydrolyzes platelet membranes and facilitates platelet aggregation in response to ADP. Topics: Adenosine Diphosphate; Animals; Blood Platelets; Calcium; Collagen; Disease Susceptibility; Group VI Phospholipases A2; Mice; Mice, Inbred C57BL; Mice, Knockout; Phospholipids; Platelet Activation; Platelet Aggregation; Receptors, Purinergic P2Y; Serotonin; Signal Transduction; Spectrometry, Mass, Electrospray Ionization; Thrombosis; Thromboxane A2 | 2014 |
LIM kinase-1 selectively promotes glycoprotein Ib-IX-mediated TXA2 synthesis, platelet activation, and thrombosis.
Current antithrombotic drugs have an adverse effect on bleeding, highlighting the need for new molecular targets for developing antithrombotic drugs that minimally affect hemostasis. Here we show that LIMK1(-/-) mice have defective arterial thrombosis in vivo but do not differ from wild-type mice with respect to bleeding time. LIMK1(-/-) mice show a selective defect in platelet activation induced through the von Willebrand Factor (VWF) receptor, the glycoprotein Ib-IX-V complex (GPIb-IX), but not by GPIb-IX-independent platelet agonists. In fact, LIMK1(-/-) platelets show an enhanced reaction to certain GPIb-IX-independent agonists. The defect of LIMK1(-/-) platelets in GPIb-IX-mediated platelet activation is attributed to a selective inhibition in VWF/GPIb-IX-induced phosphorylation of cytosolic phospholipase A2 (cPLA2) and consequent thromboxane A2 (TXA2) production. Supplementing a TXA2 analog, U46619, corrected the defect of LIMK1(-/-) platelets in VWF-induced stable platelet adhesion. Although LIMK1(-/-) platelets also showed reduced actin polymerization after GPIb-IX-mediated platelet aggregation, actin polymerization inhibitors did not reduce TXA2 generation, but rather accelerated platelet aggregation, suggesting that the role of LIMK1 in GPIb-mediated platelet activation is independent of actin polymerization. Thus, LIMK1 plays a novel role in selectively mediating GPIb-IX-dependent TXA2 synthesis and thrombosis and represents a potential target for developing antithrombotic drugs with minimal bleeding side effect. Topics: Actin Depolymerizing Factors; Actins; Animals; Blood Platelets; Cell Adhesion; Drug Design; Fibrinolytic Agents; Hemorrhage; Humans; Lim Kinases; Mice; Mice, Inbred C57BL; Mice, Knockout; Platelet Activation; Platelet Glycoprotein GPIb-IX Complex; Stress, Mechanical; Thrombosis; Thromboxane A2; von Willebrand Factor | 2013 |
Tumor vascular disrupting agent 5,6-dimethylxanthenone-4-acetic acid inhibits platelet activation and thrombosis via inhibition of thromboxane A2 signaling and phosphodiesterase.
5,6-Dimethylxanthenone-4-acetic acid (DMXAA) is a tumor vascular disrupting agent under clinical trials as an adjacent antitumor agent. DMXAA is structurally similar to flavone-8-acetic acid (FAA), an old tumor vascular disrupting agent with antiplatelet and antithrombotic effects. In contrast to FAA, which causes bleeding in tumor patients, no bleeding has been reported in patients receiving DMXAA. Whether DMXAA also affects platelet function is not clear.. To determine the effects of DMXAA on platelet function and explore the underlying mechanisms.. DMXAA concentration-dependently inhibited human platelet aggregation and ATP release induced by U46619, arachidonic acid, ADP, collagen, or ristocetin. Furthermore, DMXAA inhibited phosphorylation of Erk1/2 and Akt downstream of thromboxane A2 signaling inhibition. DMXAA also inhibited human platelet phosphodiesterase. The antiplatelet effects were further confirmed using mice administered DMXAA intravenously. DMXAA dramatically inhibited thrombus formation in FeCl3 -injured mouse mesenteric arterial thrombus model and laser-injured mouse cremaster arteriole thrombus model. Notably, at a dose exhibiting antithrombotic effects similar to those of clopidogrel in mice, DMXAA did not significantly increase bleeding.. For the first time, we found that tumor vascular disrupting agent DMXAA has potent antiplatelet and antithrombotic effects without any bleeding diathesis. As DMXAA inhibits platelet activity with safe profile, DMXAA could be used as an efficacious and safe antiplatelet drug. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Adenosine Triphosphate; Animals; Antineoplastic Agents; Humans; MAP Kinase Signaling System; Mice; Phosphodiesterase Inhibitors; Phosphorylation; Platelet Activation; Platelet Aggregation Inhibitors; Proto-Oncogene Proteins c-akt; Signal Transduction; Thrombosis; Thromboxane A2; Thromboxane-A Synthase; Xanthones | 2013 |
Impaired thrombin-induced platelet activation and thrombus formation in mice lacking the Ca(2+)-dependent tyrosine kinase Pyk2.
In the present study, we used a knockout murine model to analyze the contribution of the Ca(2+)-dependent focal adhesion kinase Pyk2 in platelet activation and thrombus formation in vivo. We found that Pyk2-knockout mice had a tail bleeding time that was slightly increased compared with their wild-type littermates. Moreover, in an in vivo model of femoral artery thrombosis, the time to arterial occlusion was significantly prolonged in mice lacking Pyk2. Pyk2-deficient mice were also significantly protected from collagen plus epinephrine-induced pulmonary thromboembolism. Ex vivo aggregation of Pyk2-deficient platelets was normal on stimulation of glycoprotein VI, but was significantly reduced in response to PAR4-activating peptide, low doses of thrombin, or U46619. Defective platelet aggregation was accompanied by impaired inside-out activation of integrin α(IIb)β(3) and fibrinogen binding. Granule secretion was only slightly reduced in the absence of Pyk2, whereas a marked inhibition of thrombin-induced thromboxane A(2) production was observed, which was found to be responsible for the defective aggregation. Moreover, we have demonstrated that Pyk2 is implicated in the signaling pathway for cPLA(2) phosphorylation through p38 MAPK. The results of the present study show the importance of the focal adhesion kinase Pyk2 downstream of G-protein-coupled receptors in supporting platelet aggregation and thrombus formation. Topics: Animals; Blood Platelets; Calcium; Focal Adhesion Kinase 2; Group II Phospholipases A2; Mice; Mice, Knockout; p38 Mitogen-Activated Protein Kinases; Phosphorylation; Platelet Activation; Platelet Aggregation; Signal Transduction; Thrombin; Thrombosis; Thromboxane A2 | 2013 |
Ginsenoside-Rp1 inhibits platelet activation and thrombus formation via impaired glycoprotein VI signalling pathway, tyrosine phosphorylation and MAPK activation.
Ginsenosides are the main constituents for the pharmacological effects of Panax ginseng. Such effects of ginsenosides including cardioprotective and anti-platelet activities have shown stability and bioavailability limitations. However, information on the anti-platelet activity of ginsenoside-Rp1 (G-Rp1), a stable derivative of ginsenoside-Rg3, is scarce. We examined the ability of G-Rp1 to modulate agonist-induced platelet activation.. G-Rp1 in vitro and ex vivo effects on agonist-induced platelet-aggregation, granule-secretion, [Ca(2+) ](i) mobilization, integrin-α(IIb) β(3) activation were examined. Vasodilator-stimulated phosphoprotein (VASP) and MAPK expressions and levels of tyrosine phosphorylation of the glycoprotein VI (GPVI) signalling pathway components were also studied. G-Rp1 effects on arteriovenous shunt thrombus formation in rats or tail bleeding time and ex vivo coagulation time in mice were determined. KEY RESULT: G-Rp1 markedly inhibited platelet aggregation induced by collagen, thrombin or ADP. While G-Rp1 elevated cAMP levels, it dose-dependently suppressed collagen-induced ATP-release, thromboxane secretion, p-selectin expression, [Ca(2+) ](i) mobilization and α(IIb) β(3) activation and attenuated p38(MAPK) and ERK2 activation. Furthermore, G-Rp1 inhibited tyrosine phosphorylation of multiple components (Fyn, Lyn, Syk, LAT, PI3K and PLCγ2) of the GPVI signalling pathway. G-Rp1 inhibited in vivo thrombus formation and ex vivo platelet aggregation and ATP secretion without affecting tail bleeding time and coagulation time, respectively.. G-Rp1 inhibits collagen-induced platelet activation and thrombus formation through modulation of early GPVI signalling events, and this effect involves VASP stimulation, and ERK2 and p38(-MAPK) inhibition. These data suggest that G-Rp1 may have therapeutic potential for the treatment of cardiovascular diseases involving aberrant platelet activation. Topics: Adenosine Triphosphate; Animals; Blood Coagulation; Calcium; Cell Adhesion Molecules; Collagen; Cyclic AMP; Cyclic GMP; Ginsenosides; Male; Mice; Mice, Inbred C57BL; Microfilament Proteins; Mitogen-Activated Protein Kinases; P-Selectin; Phosphoproteins; Phosphorylation; Platelet Aggregation; Platelet Aggregation Inhibitors; Platelet Membrane Glycoproteins; Rats; Rats, Sprague-Dawley; Thrombosis; Thromboxane A2; Tyrosine | 2012 |
Blood clots are rapidly assembled hemodynamic sensors: flow arrest triggers intraluminal thrombus contraction.
Blood clots form under flow during intravascular thrombosis or vessel leakage. Prevailing hemodynamics influence thrombus structure and may regulate contraction processes. A microfluidic device capable of flowing human blood over a side channel plugged with collagen (± tissue factor) was used to measure thrombus permeability (κ) and contraction at controlled transthrombus pressure drops.. The collagen (κ(collagen)=1.98 × 10(-11) cm(2)) supported formation of a 20-µm thick platelet layer, which unexpectedly underwent massive platelet retraction on flow arrest. This contraction resulted in a 5.34-fold increase in permeability because of collagen restructuring. Without stopping flow, platelet deposits (no fibrin) had a permeability of κ(platelet)=5.45 × 10(-14) cm(2) and platelet-fibrin thrombi had κ(thrombus)=2.71 × 10(-14) cm(2) for ΔP=20.7 to 23.4 mm Hg, the first ever measurements for clots formed under arterial flow (1130 s(-1) wall shear rate). Platelet sensing of flow cessation triggered a 4.6- to 6.5-fold (n=3, P<0.05) increase in contraction rate, which was also observed in a rigid, impermeable parallel-plate microfluidic device. This triggered contraction was blocked by the myosin IIA inhibitor blebbistatin and by inhibitors of thromboxane A2 (TXA(2)) and ADP signaling. In addition, flow arrest triggered platelet intracellular calcium mobilization, which was blocked by TXA(2)/ADP inhibitors. As clots become occlusive or blood pools following vessel leakage, the flow diminishes, consequently allowing full platelet retraction.. Flow dilution of ADP and thromboxane regulates platelet contractility with prevailing hemodynamics, a newly defined flow-sensing mechanism to regulate clot function. Topics: Adenosine Diphosphate; Blood Coagulation; Blood Platelets; Calcium; Cardiovascular Physiological Phenomena; Collagen; Equipment Design; Hemodynamics; Heterocyclic Compounds, 4 or More Rings; Humans; In Vitro Techniques; Microfluidic Analytical Techniques; Nonmuscle Myosin Type IIA; Regional Blood Flow; Signal Transduction; Thrombosis; Thromboxane A2 | 2012 |
[Dynamics of morphological structures of platelets in patients with ischemic heart disease in dependence on blood levels of fatty acids].
Levels of fatty acids in platelets of ischemic heart disease (IHD) patients (n = 39) has been compared with those in healthy subjects (n = 12). Increased content of arachidonic acid and thromboxane A2 in platelets of IHD patients forms thrombogenic picture of IHD. High level of fibrinogen and decrease in heparin and antithrombin III in IHD patients facilitates formation of blood clots. Morphological examination of platelets in IHD patients has demonstrated an increase of levels of discocytes and spherocytes as well as appearance of small and large platelet-erythrocyte aggregates promoting blood slot formation. Topics: Antithrombins; Arachidonic Acid; Blood Coagulation Factors; Blood Platelets; Erythrocyte Aggregation; Erythrocyte Deformability; Humans; Myocardial Ischemia; Platelet Adhesiveness; Platelet Aggregation; Thrombosis; Thromboxane A2 | 2011 |
An important role for Akt3 in platelet activation and thrombosis.
The Akt family of serine/threonine kinases includes Akt1, Akt2, and Akt3 isoforms. Prior studies have reported that Akt1 and Akt2, but not Akt3, are expressed in platelets. Here, we show that Akt3 is expressed in substantial amounts in platelets. Akt3(-/-) mouse platelets selectively exhibit impaired platelet aggregation and secretion in response to low concentrations of thrombin receptor agonists and thromboxane A₂ (TXA₂), but not collagen or VWF. In contrast, platelets from Akt1(-/-) or Akt2(-/-) mice are defective in platelet activation induced by thrombin, TXA₂, and VWF, but only Akt1(-/-) platelets show significant defects in response to collagen, indicating differences among Akt isoforms. Akt3(-/-) platelets exhibit a significant reduction in thrombin-induced phosphorylation of glycogen synthase kinase 3β (GSK-3β) at Ser9, which is known to inhibit GSK-3β function. Thus, Akt3 is important in inhibiting GSK-3β. Accordingly, treatment of Akt3(-/-) platelets with a GSK-3β inhibitor rescued the defect of Akt3(-/-) platelets in thrombin-induced aggregation, suggesting that negatively regulating GSK-3β may be a mechanism by which Akt3 promotes platelet activation. Importantly, Akt3(-/-) mice showed retardation in FeCl₃-induced carotid artery thrombosis in vivo. Thus, Akt3 plays an important and distinct role in platelet activation and in thrombosis. Topics: Animals; Blood Platelets; Glycogen Synthase Kinase 3; Glycogen Synthase Kinase 3 beta; Humans; Mice; Mice, Knockout; Phosphorylation; Platelet Aggregation; Proto-Oncogene Proteins c-akt; Thrombin; Thrombosis; Thromboxane A2; von Willebrand Factor | 2011 |
[The study of relationship between platelet function and thrombus in patients with essential thrombocythaemia].
To observe the influence of the plasma thromboxane B2 (TXB2), 6-keto-PGF1alpha, CD62P and PAC-1 and Thrombus in patients with primary thrombocytosis (ET). To observe the effect of sodium ozagrel to prevent and treat thrombosis in patients with ET.. The subjects including 48 patients with ET. All patients were measured the plasma TXB2, 6-keto-PGF1alpha, CD62P and PAC-1 before and after treatment with or without sodium ozagrel.. The plasma levels of CD62P, PAC-1, TXB2, 6-keto-PGF1alpha and TXA2/PGI2 in the patients with ET were significantly higher than the normal people (P < 0.01). The levels of CD62P, PAC-1, TXB2, TXB2/6-keto-PGF1alpha in patients with treatment of sodium ozagrel were higher than patients without treatment of sodium ozagrel (P < 0.01). The plasma levels of CD62P, PAC-1 and TXA2/PGI2 in patients with treatment of sodium ozagrel and that in normal people had no significant distinction (P < 0.01). All the index of conventional therapy group were higher than normal people (P < 0.01) but had no significant distinction with the patients before conventional treating. The incidence of thrombus in patients treated with sodium ozagrel was lower than patients treated without sodium ozagrel (P < 0.05).. With the treatment of sodium ozagrel in patients with ET, the CD62P, PAC-1, TXB2 and TXA2/PGI2 of plasma could be decreased. And the incidence of thrombus was decreased. Topics: 6-Ketoprostaglandin F1 alpha; Adult; Antibodies, Monoclonal; Blood Platelets; Female; Humans; Male; Methacrylates; Middle Aged; P-Selectin; Receptors, Fibrinogen; Receptors, Pituitary Adenylate Cyclase-Activating Polypeptide, Type I; Thrombocythemia, Essential; Thrombosis; Thromboxane A2; Thromboxane B2 | 2010 |
Characterization of human platelet binding of recombinant T cell receptor ligand.
Recombinant T cell receptor ligands (RTLs) are bio-engineered molecules that may serve as novel therapeutic agents for the treatment of neuroinflammatory conditions such as multiple sclerosis (MS). RTLs contain membrane distal α1 plus β1 domains of class II major histocompatibility complex linked covalently to specific peptides that can be used to regulate T cell responses and inhibit experimental autoimmune encephalomyelitis (EAE). The mechanisms by which RTLs impede local recruitment and retention of inflammatory cells in the CNS, however, are not completely understood.. We have recently shown that RTLs bind strongly to B cells, macrophages, and dendritic cells, but not to T cells, in an antigenic-independent manner, raising the question whether peripheral blood cells express a distinct RTL-receptor. Our study was designed to characterize the molecular mechanisms by which RTLs bind human blood platelets, and the ability of RTL to modulate platelet function.. Our data demonstrate that human blood platelets support binding of RTL. Immobilized RTL initiated platelet intracellular calcium mobilization and lamellipodia formation through a pathway dependent upon Src and PI3 kinases signaling. The presence of RTL in solution reduced platelet aggregation by collagen, while treatment of whole blood with RTL prolonged occlusive thrombus formation on collagen.. Platelets, well-known regulators of hemostasis and thrombosis, have been implicated in playing a major role in inflammation and immunity. This study provides the first evidence that blood platelets express a functional RTL-receptor with a putative role in modulating pathways of neuroinflammation. Topics: Animals; Blood Platelets; Calcium; Chelating Agents; Cytoskeleton; Egtazic Acid; Enzyme Inhibitors; Humans; Ligands; Mice; Platelet Adhesiveness; Platelet Glycoprotein GPIIb-IIIa Complex; Protein Binding; Receptors, Antigen, T-Cell; Recombinant Fusion Proteins; Signal Transduction; Thrombosis; Thromboxane A2 | 2010 |
Aspirin and the in vitro linear relationship between thromboxane A2-mediated platelet aggregation and platelet production of thromboxane A2.
Currently, 'aspirin resistance', the anti-platelet effects of non-steroid anti-inflammatory drugs (NSAIDs) and NSAID-aspirin interactions are hot topics of debate. It is often held in this debate that the relationship between platelet activation and thromboxane (TX) A(2) formation is non-linear and TXA(2) generation must be inhibited by at least 95% to inhibit TXA(2)-dependent aggregation. This relationship, however, has never been rigorously tested.. To characterize, in vitro and ex vivo, the concentration-dependent relationships between TXA(2) generation and platelet activity.. Platelet aggregation, thrombi adhesion and TXA(2) production in response to arachidonic acid (0.03-1 mmol L(-1)), collagen (0.1-30 microg mL(-1)), epinephrine (0.001-100 micromol L(-1)), ADP, TRAP-6 amide and U46619 (all 0.1-30 micromol L(-1)), in the presence of aspirin or vehicle, were determined in 96-well plates using blood taken from naïve individuals or those that had taken aspirin (75 mg, o.d.) for 7 days.. Platelet aggregation, adhesion and TXA(2) production induced by either arachidonic acid or collagen were inhibited in concentration-dependent manners by aspirin, with logIC(50) values that did not differ. A linear relationship existed between aggregation and TXA(2) production for all combinations of arachidonic acid or collagen and aspirin (P < 0.01; R(2) 0.92; n = 224). The same relationships were seen in combinations of aspirin-treated and naïve platelets, and in blood from individuals taking an anti-thrombotic dose of aspirin.. These studies demonstrate a linear relationship between inhibition of platelet TXA(2) generation and TXA(2)-mediated aggregation. This finding is important for our understanding of the anti-platelet effects of aspirin and NSAIDs, NSAID-aspirin interactions and 'aspirin resistance'. Topics: Anti-Inflammatory Agents, Non-Steroidal; Arachidonic Acid; Aspirin; Blood Platelets; Collagen; Drug Resistance; Humans; Platelet Aggregation; Thrombosis; Thromboxane A2 | 2008 |
Preliminary study of pulsed-electromagnetic fields effects on endothelial (HUVEC) cell secretions--modulation of the thrombo-hemorrhagic balance.
We investigated the role of low-amplitude magnetic pulse with low repetition frequency superimposed on the environmental electromagnetic field (EMF) on the secretion of anti-aggregant (Prostacyclin or PGI(2)) and pro-aggregant (Thromboxane A(2)) agents in endothelial cells of the human umbilical cord vein (HUVEC). We established that magnetic pulse exposure modulates both PGI(2) and TXA(2). These modulations depend on the frequency, width of the pulse, and intensity of the magnetic field. Moreover, we corroborated previous results obtained with an endothelial cell line (EaHy-926), concerning the increased thrombo-embolic risk for the 1 Hz frequency. Topics: Animals; Cell Line; Electromagnetic Fields; Endothelial Cells; Epoprostenol; Hemorrhage; Humans; Platelet Activating Factor; Platelet Aggregation Inhibitors; Thrombosis; Thromboxane A2; Time Factors | 2008 |
Additive anti-atherogenic effect of thromboxane receptor antagonism with 12/15lipoxygenase gene disruption in apolipoprotein E-deficient mice.
Previous studies in mouse models showed that 12/15lipoxygenase (12/15LO) gene disruption diminishes atherosclerosis. Pharmacologic suppression of thromboxane (Tx) A(2) biosynthesis or blockade of its receptor also reduces the development of the disease in the same models. We tested the hypothesis that simultaneous genetic absence of 12/15LO with TxA(2) receptor blockade might result in an additive anti-atherogenic effect. Apolipoprotein E (apoE)-deficient mice and apoE-deficient mice lacking 12/15LO were maintained on normal chow diet, or chow supplemented with BM-573, a selective TxA(2) receptor antagonist, for 12 weeks. Urinary TxA(2) and prostacyclin metabolites, isoprostaneF(2*)-III and atherosclerotic aortic lesions were assessed. 12/15LO gene disruption resulted in significantly reduced atherosclerotic lesion areas and decreased urinary isoprostaneF(2*)-III in apoE-deficient mice. TxA(2) receptor antagonism alone also afforded a significant reduction in atherosclerosis in apoE-deficient mice. However, thromboxane receptor blockade resulted in an additive and more potent anti-inflammatory and anti-atherogenic effect when administered to apoE-deficient mice lacking 12/15LO. These results suggest that the 12/15LO- and TxA(2) receptor-mediated pro-atherogenic effects are two distinct pathways and represent two separate therapeutic targets for a better anti-atherogenic strategy. Topics: Animals; Antioxidants; Apolipoproteins E; Arachidonate 12-Lipoxygenase; Arachidonate 15-Lipoxygenase; Atherosclerosis; Gene Expression Regulation; Inflammation; Mice; Mice, Inbred C57BL; Mice, Knockout; Receptors, Thromboxane; Thrombosis; Thromboxane A2; Vitamin E | 2008 |
Experimental arterial thrombosis regulated by androgen and its receptor via modulation of platelet activation.
The aim of our study is to elucidate whether experimental arterial thrombosis is regulated by physiological doses of androgen and its receptor via modulation of platelet activation.. Surgical castration was performed in male rats and ferric chloride (FeCl(3)), as a stimulator, induced the experimental arterial thrombosis. Testosterone was measured directly by chemiluminescent immunoassay on the Bayer ADVIA Centaur analyzer. Dihydrotestosterone (DHT) was determined by ELISA using a commercially available kit. A platelet aggregometer was used to assess aggregation, and a platelet adherometer was used to measure adhesion. The contents of TXB(2) and 6-Keto-PGF(1alpha) were assayed by radio-immunoassay using commercially available kits.. Our data showed that DHT replaced restored circulating DHT of castrated rats to physiological levels, without being altered by treatment with flutamide. Castration caused significant increases in the thrombus area and weight in castrated rats as compared with control group. In PRP diluted with autologous PPP, ADP-induced platelet aggregation rate was only 9.10%. However, in PRP diluted with Tyrode's buffer, 1 microM ADP-induced platelet aggregation rate rose to 63.65%. In PRP diluted with Tyrode's buffer, and pretreated with DHT (1 nM, 2 nM), ADP-induced platelet aggregation was significantly lowered again. Platelet aggregation in PRP diluted with autologous PPP was enhanced in castrated rats as compared with sham-operated rats, and DHT (2 nM) replacement suppressed platelet aggregation in castrated PRP to the level similar to that of sham-operated rats. However, presence of flutamide (3 microM) significantly increased platelet aggregation in PRP diluted with autologous PPP or Tyrode's buffer. DHT (2 nM) replacement significantly inhibited the ADP-induced platelet adhesion. However, presence of flutamide (3 microM) increased ADP-induced platelet adhesion again. DHT replacement obviously reduced the ratio of TXB(2) to 6-keto-PGF(1alpha) in castrated rats. However, administration of flutamide and DHT to castrated rats caused an increase in the ratio of TxB(2) to 6-keto-PGF1alpha.. Inhibition of experimental arterial thrombosis by androgen at physiological doses and its receptor is mediated via modulation of platelet activation. Topics: 6-Ketoprostaglandin F1 alpha; Androgens; Animals; Arteries; Castration; Chlorides; Dihydrotestosterone; Ferric Compounds; Male; Platelet Activation; Platelet Adhesiveness; Platelet Aggregation; Rats; Receptors, Androgen; Testosterone; Thrombosis; Thromboxane A2 | 2007 |
Role of murine integrin alpha2beta1 in thrombus stabilization and embolization: contribution of thromboxane A2.
Platelets stably interact with collagen via glycoprotein (GP)VI and alpha2beta1integrin. With alpha2-null mice, we investigated the role of alpha2beta1 in thrombus formation and stability in vivo and in vitro. Using a FeCl(3)-induced thrombosis model, in arteries from alpha2-null mice smaller thrombi were formed with more embolization compared to vessels from wild-type mice. Aspirin treatment of wild-type mice causes similar effects, while the thromboxane A(2) analogue U46619 was borderline effective in suppressing the embolisation in alpha2-null mice. In vitro, perfusion of alpha2-null blood over collagen resulted in formation of thrombi that were smaller and looser in appearance, regardless of the presence or absence of coagulation. Aspirin treatment or blockage of thromboxane receptors provoked embolus formation in wildtype blood, while U46619 normalized thrombus formation in blood from alpha2-null mice. We conclude that integrin alpha2beta1 plays a role in stabilizing murine thrombi, likely by enhancing GPVI activation and thromboxane A(2) release. The increased embolization in alpha2-null mice may argue against the use of alpha2beta1 integrin inhibitors for antithrombotic therapy. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Animals; Aspirin; Chlorides; Collagen; Ferric Compounds; Integrin alpha2beta1; Mice; Mice, Knockout; Thromboembolism; Thrombosis; Thromboxane A2 | 2007 |
Bruton tyrosine kinase is essential for botrocetin/VWF-induced signaling and GPIb-dependent thrombus formation in vivo.
Botrocetin (bt)-facilitated binding of von Willebrand factor (VWF) to the platelet membrane glycoprotein (GP) Ib-IX-V complex on platelets in suspension initiates a signaling cascade that causes alphaIIbbeta3 activation and platelet aggregation. Previous work has demonstrated that bt/VWF-mediated agglutination activates alphaIIbbeta3 and elicits ATP secretion in a thromboxane A2 (TxA2)-dependent manner. The signaling that results in TxA2 production was shown to be initiated by Lyn, enhanced by Src, and propagated through Syk, SLP-76, PI3K, PLCgamma2, and PKC. Here, we demonstrate that the signaling elicited by GPIb-mediated agglutination that results in TxA2 production is dependent on Bruton tyrosine kinase (Btk). The results demonstrate that Btk is downstream of Lyn, Syk, SLP-76, and PI3K; upstream of ERK1/2, PLCgamma2, and PKC; and greatly enhances Akt phosphorylation. The relationship(s), if any, between ERK1/2, PLCgamma2, and PKC were not elucidated. The requirement for Btk and TxA2 receptor function in GPIb-dependent arterial thrombosis was confirmed in vivo by characterizing blood flow in ferric chloride-treated mouse carotid arteries. These results demonstrate that the Btk family kinase, Tec, cannot provide the function(s) missing because of the absence of Btk and that Btk is essential for both bt/VWF-mediated agglutination-induced TxA2 production and GPIb-dependent stable arterial thrombus formation in vivo. Topics: Agammaglobulinaemia Tyrosine Kinase; Animals; Carotid Artery Thrombosis; Crotalid Venoms; Male; MAP Kinase Signaling System; Mice; Mice, Inbred CBA; Mice, Knockout; Mice, Mutant Strains; Phospholipase C gamma; Phosphorylation; Platelet Aggregation; Platelet Glycoprotein GPIb-IX Complex; Protein-Tyrosine Kinases; Signal Transduction; Thrombosis; Thromboxane A2; von Willebrand Factor | 2006 |
COX-2 inhibitors and the thrombotic risk.
Topics: Animals; Clinical Trials as Topic; Cyclooxygenase 2 Inhibitors; Dinoprostone; Epoprostenol; Humans; Myocardial Infarction; Risk Assessment; Thrombosis; Thromboxane A2 | 2006 |
Stronger inhibition by nonsteroid anti-inflammatory drugs of cyclooxygenase-1 in endothelial cells than platelets offers an explanation for increased risk of thrombotic events.
Recent data have suggested that regular consumption of nonsteroid anti-inflammatory drugs (NSAIDs), particularly selective inhibitors of cyclo-oxygenase-2 (COX-2), is associated with an increased risk of thrombotic events. It has been suggested that this is due to NSAIDs reducing the release from the endothelium of the antithrombotic mediator prostaglandin I2 as a result of inhibition of endothelial COX-2. Here, however, we show that despite normal human vessels and endothelial cells containing cyclo-oxygenase-1 (COX-1) without any detectable COX-2, COX-1 in vessels or endothelial cells is more readily inhibited by NSAIDs and COX-2-selective drugs than COX-1 in platelets (e.g., log IC50+/-SEM values for endothelial cells vs. platelets: naproxen -5.59+/-0.07 vs. -4.81+/-0.04; rofecoxib -4.93+/-0.04 vs. -3.75+/-0.03; n=7). In broken cell preparations, the selectivities of the tested drugs toward endothelial cell over platelet COX-1 were lost. These observations suggest that variations in cellular conditions, such as endogenous peroxide tone and substrate supply, and not the isoform of cyclo-oxygenase present, dictate the effects of NSAIDs on endothelial cells vs. platelets. This may well be because the platelet is not a good representative of COX-1 activity within the body as it produces prostanoids in an explosive burst that does not reflect tonic release from other cells. The results reported here can offer an explanation for the apparent ability of NSAIDs and COX-2-selective inhibitors to increase the risk of myocardial infarction and stroke. Topics: Anti-Inflammatory Agents, Non-Steroidal; Aorta; Blood Platelets; Cyclooxygenase 1; Cyclooxygenase 2; Cyclooxygenase Inhibitors; Endothelial Cells; Epoprostenol; Gene Expression Regulation, Enzymologic; Humans; Thrombosis; Thromboxane A2; Veins | 2006 |
Role of p38 mitogen-activated protein kinase in thrombus formation.
The present study was designed to elucidate the role of p38 mitogen-activated protein kinase (p38) in thrombus formation. We used p38alpha heterozygous (p38alpha+/-) mice and used ferric chloride (FeCl3)-induced carotid artery injury as a model of thrombus formation. The time to thrombotic occlusion induced by FeCl3 in p38alpha+/- mice was prolonged compared to that in wild-type (WT) mice. Platelets prepared from p38alpha+/- mice showed impairment of the aggregatory response to a low concentration of U46619, a thromboxane A2 analogue. Furthermore, platelets prepared from p38alpha+/- mice and activated by U46619 were poorly bound to fibrinogen compared with those from WT mice. Both the expression and activity of tissue factor induced by FeCl3 in WT mice were higher than those in p38alpha+/- mice. These results suggest that p38 plays an important role in thrombus formation by regulating platelet function and tissue factor activity. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Alleles; Animals; Blood Coagulation; Blood Platelets; Chlorides; Ferric Compounds; Fibrinogen; Heterozygote; Homozygote; Male; Mice; Mice, Inbred C57BL; Mice, Transgenic; p38 Mitogen-Activated Protein Kinases; Placenta; Platelet Adhesiveness; Protein Binding; Protein Isoforms; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Thromboplastin; Thrombosis; Thromboxane A2; Time Factors; Vasoconstrictor Agents | 2004 |
Complementary roles of glycoprotein VI and alpha2beta1 integrin in collagen-induced thrombus formation in flowing whole blood ex vivo.
Platelets interact vigorously with subendothelial collagens that are exposed by injury or pathological damage of a vessel wall. The collagen-bound platelets trap other platelets to form aggregates, and they expose phosphatidylserine (PS) required for coagulation. Both processes are implicated in the formation of vaso-occlusive thrombi. We previously demonstrated that the immunoglobulin receptor glycoprotein VI (GPVI), but not integrin alpha2beta1, is essential in priming platelet-collagen interaction and subsequent aggregation. Here, we report that these receptors have yet a complementary function in ex vivo thrombus formation during perfusion of whole blood over collagen. With mice deficient in GPVI or blocking antibodies, we found that GPVI was indispensable for collagen-dependent Ca2+ mobilization, exposure of PS, and aggregation of platelets. Deficiency of integrin beta1 reduces the GPVI-evoked responses but still allows the formation of loose platelet aggregates. By using mice deficient in G(alpha)q or specific thromboxane A2 and ADP antagonists, we show that these autocrine agents mediated aggregation but not collagen-induced Ca2+ mobilization or PS exposure. Collectively, these data indicate that integrin alpha2beta1 facilitates the central function of GPVI in the platelet activation processes that lead to thrombus formation, whereas the autocrine thromboxane A2 and ADP serve mainly to trigger aggregate formation. Topics: Adenosine Diphosphate; Animals; Blood Platelets; Calcium; Collagen; GTP-Binding Protein alpha Subunits, Gq-G11; Heterotrimeric GTP-Binding Proteins; Integrin alpha2beta1; Mice; Mice, Knockout; Platelet Adhesiveness; Platelet Aggregation; Platelet Membrane Glycoproteins; Receptors, IgG; Thrombosis; Thromboxane A2 | 2003 |
Role for thromboxane A2 from glomerular thrombi in nephropathy with type 2 diabetic rats.
We used rats (the Otsuka Long-Evans Tokushima Fatty strain) as a model of type 2 diabetes to find whether thromboxane (TX) A2 is involved in diabetic nephropathy, and if so, to identify where it is synthesized. We measured urinary excretion of TXB2 and 2,3-dinor-TXB2 in rats up to 60 weeks of age as markers of renal and platelet synthesis of TXA2, respectively. Some diabetic rats were given daily oral doses of OKY-046 (100 mg/kg), a TXA2 synthase inhibitor, starting when they were 10 weeks of age. Healthy Long-Evans Tokushima Otsuka rats served as the controls. Urinary excretion of protein was greater in diabetic rats at 26 weeks than in controls, and the difference increased with age. Urinary excretion of TXB2 by diabetic rats was about 150% that of controls at 14 weeks, and remained at that level. In diabetic rats, urinary excretion of 2,3-dinor-TXB2 increased with age in parallel to increases in proteinuria, but in controls, excretion of these metabolites did not change with age. In diabetic rats, OKY-046 prevented the increase in urinary excretion of both metabolites, and decreased the proteinuria. Histologic examination at 60 weeks showed intraglomerular thrombi in diabetic rats but not in controls. OKY-046 reduced intraglomerular thrombi formation and the score for glomerulosclerosis. When platelet aggregation began, more TXA2 than before was released from the thrombi that formed, and the TXA2 contributed to the progress of nephropathy in this rat model of type 2 diabetes. Topics: 6-Ketoprostaglandin F1 alpha; Aging; Animals; Blood Glucose; Blood Pressure; Body Weight; Diabetes Mellitus, Type 2; Diabetic Nephropathies; Disease Progression; Enzyme Inhibitors; Glomerular Mesangium; Male; Methacrylates; Prostaglandins; Proteinuria; Rats; Rats, Inbred OLETF; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 2003 |
Enhancement of platelet aggregation and thrombus formation by arsenic in drinking water: a contributing factor to cardiovascular disease.
Arsenic in drinking water is a worldwide health problem that is associated with cardiovascular disease, but the cause is currently unknown. Arsenic effects on platelets, which are important in development of cardiovascular disease, were examined in vitro and in a drinking water study using a rat animal model. Trivalent inorganic arsenic (arsenite) induced in vitro aggregation when platelets were exposed to subthreshold challenge by thrombin and several other agonists in a concentration-dependent manner, with arsenite being the most potent form tested. Arsenite also induced significant increases in serotonin secretion, thromboxane A(2) formation, and adhesion protein expression in platelets. Consistent with the in vitro studies, 4-week ingestion of arsenite-contaminated drinking water resulted in enhanced arterial thrombosis. Human platelets showed similar responses, suggesting that the effects seen in animal experiments are applicable to humans. These results will provide new insights into the mechanism of arsenic-induced cardiovascular disease. They will also allow regulatory agencies to estimate risk from arsenic-induced cardiovascular disease and to determine if drinking water regulatory levels based on human cancer studies will protect against noncancer effects associated with cardiovascular disease. Topics: Animals; Arsenic; Blood Platelets; Cardiovascular Diseases; Humans; In Vitro Techniques; P-Selectin; Platelet Aggregation; Poisons; Rats; Rats, Sprague-Dawley; Serotonin; Thrombin; Thrombosis; Thromboxane A2; Water Supply | 2002 |
Modulation of rat platelet activation by vessel wall-derived prostaglandin and platelet-derived thromboxane: effects of dietary fish oil on thromboxane-prostaglandin balance.
By dietary manipulation of rats with n-3 polyunsaturated fatty acids (PUFAs), platelets and endothelium-containing aortic tissue were obtained with decreased levels of arachidonate and increased levels of eicosapentaenoate and docosahexaenoate. These diet-induced changes were accompanied by a reduced formation of thromboxane A(2) (TXA(2)) and prostaglandin I(2) (PGI(2)) in platelets and aortic tissue, respectively. When platelets were incubated with autologous, aorta-derived PGI(2), the dietary modulation of PGI(2) generation had a stronger effect on the activation process than the dietary effect on TXA(2) generation. The platelet-inhibiting effect of PGI(2) was independent of the type of agonist and involved both TXA(2)-dependent and -independent activation responses. PGI(2) also inhibited the agonist-induced formation of TXA(2). In addition, the platelet-inhibitory effect of PGI(2) was more prolonged in time than the brief, stimulatory effect of TXA(2). We conclude that, in the thromboxane-prostaglandin balance of platelet activation, PGI(2) plays a more prominent role than TXA(2). Furthermore, dietary n-3 PUFAs appear to influence platelet activation more by reducing formation of endothelial PGI(2) than by decreasing autocrine-produced TXA(2). Thus, in rats, the proposed antithrombotic effect of fish oil is unlikely to be caused by an altered thromboxane-prostaglandin balance. Topics: Animals; Aorta, Abdominal; Arachidonic Acid; Blood Platelets; Calcium; Epoprostenol; Fatty Acids, Omega-3; Fatty Acids, Unsaturated; Fish Oils; Hypolipidemic Agents; Male; Platelet Activation; Rats; Rats, Wistar; Thrombosis; Thromboxane A2; Triglycerides | 2001 |
Relationships between thrombohemorrhagic complications and platelet function in patients with essential thrombocythaemia.
Three subgroups have been distinguished in essential thrombocythaemia (ET) patients, on the basis of clinical and laboratory findings. ET patients with bleeding incidents had smaller platelet volume, lower concentrations of beta-thromboglobulin and platelet factor 4 in their plasma, 10%, 26%, and 26% lower compared to patients without complications, respectively. ATP secretion from platelets of bleeders, clotters, and "no-complications" ET patients was found to be 75%, 36%, and 45%, respectively, lower than in healthy people. Spontaneous platelet aggregation appeared to be normal in about 90% of ET patients with no complications and in all bleeders but only in 35% patients with clotting incidents. All bleeders had abnormal agonist-evoked aggregation assays. Among remaining ET patients 30%-60% displayed normal values of different evoked aggregation tests. Thus, clinically distinguished group of bleeding ET patients may be differentiated from other subgroups on the basis of laboratory findings. Topics: Adenosine Triphosphate; Adult; Aged; Blood Platelets; Female; Hemorrhage; Humans; Male; Middle Aged; Platelet Aggregation; Reference Values; Thrombocytosis; Thrombosis; Thromboxane A2 | 2001 |
Increased synthesis of thromboxane A(2) and expression of procoagulant activity by monocytes in response to arachidonic acid in diabetes mellitus.
Thromboxane A(2) (TXA(2)) synthesis and expression of procoagulant activity (PCA) were investigated in mononuclear cells and monocytes prepared from a control and a Type 2 diabetic group. Monocytes from the diabetic group produced 2.10+/-0.81 ng of TXB(2)/5 x 10(5) monocytes compared to 1.26+/-0.43 ng/5 x 10(5) monocytes by the control group (P<0.01, n=11) when incubated in autologous plasma containing arachidonic acid (200 microg/ml). When monocytes were incubated in buffer containing arachidonic acid (20 microg/ml), cells from the diabetic group produced 1.65+/-0.68 ng of TXB(2)/5 x 10(5) monocytes compared to 1.07+/-0.31 ng/5 x 10(5) monocytes by the control group (P<0.02, n=12). Expression of PCA was examined in mononuclear cell preparations. Basal and maximally stimulated PCA with lipopolysaccharide (4.2 microg/ml) were not different between control and diabetic groups. However, arachidonic acid induced a four-fold (P<0.001) increase in PCA in the diabetic group. This activity was characterized as tissue factor. Increased synthesis of TXA(2) and expression of PCA may potentiate thrombosis and increase fibrin deposition, events that play primary roles in the development of vascular disease. Topics: Arachidonic Acid; Arteriosclerosis; Blood Coagulation Factors; Cells, Cultured; Culture Media, Serum-Free; Diabetes Complications; Diabetes Mellitus; Diabetic Angiopathies; Female; Humans; Kinetics; Leukocytes, Mononuclear; Lipopolysaccharides; Male; Middle Aged; Monocytes; Thrombosis; Thromboxane A2 | 2001 |
[The significance of thrombosis after experimental pulmonary thromboembolism].
To study the significance of thrombosis after experimental pulmonary thromboembolism (PTE).. Acute PTE models of rabbits were established with injection of autologous blood clots (0.04 g/kg) stabilized in a temperature-controlled (70 degrees C) of distilled water for 10 minutes through the femoral vein, then the regulation of thrombosis was explored at dissection and upon microscopic examination after PTE. Moreover, the coagulability of blood and the plasma level of thromboxane A2(TXA2) and endothelin (ET) were examined.. Thrombotic propensity was found at 1 h, and fresh thrombosis started to form at 24 h following clots infusion. Emboli were completely or partly dissolved at 5 d and organized at 10 and 14 d after clots infused. Prothrombin time was significantly lower [(7.15 +/- 0.06)s], and fibrinogen was higher [(5.86 +/- 1.50) g/L] at 24 h post-clots, compared with pre-clots [(7.34 +/- 0.19)s, (3.37 +/- 1.02) g/L] (P < 0.05). Venous plasma level of TXA2 began to increase at 5 min [(2.5 +/- 0.7) micrograms/L] and continued to rise to its maximum at 15 min [(2.5 +/- 0.6) micrograms/L], then declined at 60 min after clots infusion. The level of ET in both arterial and venous blood increased at 5 d post-clots [(0.84 +/- 0.15) micrograms/L and (0.23 +/- 0.05) micrograms/L] separately, while most of emboli resolved.. There is thrombus formation after autologous-blood-clots-induced PTE. Furthermore, thrombus formation, fibrinolysis and organization may always interact on each other consistently, and control the pathogenesis of PTE. Abnormalities of ET metabolism occur after PTE and the major mediator of TXA2 plays an important role in the early phase of PTE. Topics: Animals; Disease Models, Animal; Endothelins; Female; Fibrinogen; Lung; Male; Prothrombin Time; Pulmonary Embolism; Rabbits; Thrombosis; Thromboxane A2 | 2001 |
Results of the BRAT study--a pilot study investigating the possible significance of ASA nonresponsiveness on the benefits and risks of ASA on thrombosis in patients undergoing coronary artery bypass surgery.
Several studies suggest that acetylsalicylic acid (ASA) is less effective in preventing thrombotic events in ASA nonresponder patients. If so, the thrombotic event rate in ASA nonresponders should be higher than in ASA responders.. To conduct a prospective, multicentre observational pilot study to determine the thrombotic event rates in ASA responders and nonresponders.. Patients undergoing nonurgent coronary artery bypass grafting (CABG) who were prescribed 325 mg ASA/day were recruited. Patients were classified as an ASA responder or nonresponder based on the ASA effect (or lack thereof) on their bleeding times. All thrombotic events that occurred in the two years following CABG were recorded. These data were stored in a blinded fashion until the last patient follow-up, and then adjudicated by a validation committee.. A total of 289 patients recruited at three sites completed the two-year follow-up. Of these patients, 45.3% were classified as ASA responders and 54.7% were classified as ASA nonresponders. Of ASA responders, 6.9% had thrombotic events compared with 9.5% of the ASA nonresponders, but this difference was not significant (P=0.526).. While ASA responder or nonresponder status did not appear to affect the thrombotic event rate in patients undergoing nonurgent CABG, the possibility that ASA responder or nonresponder status affects the thrombotic event rate in more acutely ill CABG patients cannot be excluded. Topics: Aspirin; Bleeding Time; Coronary Artery Bypass; Female; Follow-Up Studies; Humans; Male; Middle Aged; Pilot Projects; Platelet Aggregation Inhibitors; Prospective Studies; Risk Factors; Thrombosis; Thromboxane A2 | 2000 |
Effects of green tea catechin on phospholipase A2 activity and antithrombus in streptozotocin diabetic rats.
The purpose of this study was to investigate the effects of dietary green tea catechin on phospholipase A2 (PLA2) activity and the antithrombotic reaction of platelets in streptozotocin (STZ)-diabetic rats. Sprague-Dawley male rats weighing 100 +/- 10 g were randomly divided into one normal and three STZ-diabetic groups, which were subdivided into catechin-free group (DM-0C), 0.5% catechin group (DM-0.5C) and 1% catechin group (DM-1C). The activity level of platelet phospholipase A2 was higher in the diabetic groups than in the normal group, while it was lower in DM-0.5C and DM-1C than in DM-0C. The activity of platelet cyclooxygenase in DM-0C was 1.1-fold as high as in the normal group, but was significantly reduced by catechin supplementation. The platelet thromboxane A2 (TXA2) formation became higher in DM-0C as compared to the normal group, but not in DM-0.5C and DM-1C. The synthesis of aortic prostacyclin (PGI2) was lower in DM-0C and DM-0.5C than in the normal group. The PGI2/TXA2 ratio was decreased to 55% in DM-0C, but was restored by catechin supplementation. These results indicate that STZ-diabetic rats are sensitive to platelet aggregation and thrombosis, and that the abnormality can be improved by dietary catechin. Topics: Animals; Aorta; Blood Platelets; Catechin; Diabetes Mellitus, Experimental; Epoprostenol; Male; Phospholipases A; Phospholipases A2; Platelet Aggregation; Prostaglandin-Endoperoxide Synthases; Rats; Rats, Sprague-Dawley; Tea; Thiobarbituric Acid Reactive Substances; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1999 |
A new thromboxane receptor antagonist, Z-335, ameliorates experimental thrombosis without prolonging the rat tail bleeding time.
We investigated the antithrombotic activity of Z-335, a new thromboxane A2 receptor antagonist, using experimental thrombosis models, and also tested its effect on the rat tail bleeding time. Z-335 (0.1, 0.3, and 1 mg/kg, p.o.) dose-dependently prevented the occurrence of arachidonic acid-induced pulmonary thromboembolism in mice. During photochemically induced thrombosis in the femoral artery of guinea pigs, Z-335 (0.3, 1, and 3 mg/kg, i.v.) dose-dependently prolonged the time required to form thrombi. Moreover, Z-335 (10 mg/kg/day, p.o.) strongly suppressed lauric acid-induced hind limb injury in rats. Z-335 (0.3, 3, 30, and 300 mg/kg, p.o.) did not prolong the tail bleeding time in rats. These results strongly suggest that Z-335 ameliorates experimental thrombosis without prolonging the rat tail bleeding time, and may therefore be a useful antithrombotic drug. Topics: Animals; Bleeding Time; Fibrinolytic Agents; Indans; Male; Mice; Mice, Inbred ICR; Rats; Rats, Sprague-Dawley; Receptors, Thromboxane; Thrombosis; Thromboxane A2 | 1998 |
A ferret model of electrical-induction of arterial thrombosis that is sensitive to aspirin.
An experimental model of acute thrombosis was developed in pentobarbital- anesthetized ferrets. A 10-min anodal electrical stimulation of 1 mA was delivered to the external surface of the carotid artery while measuring carotid blood flow (CBF). This produced an occlusive thrombus in all vehicle-treated ferrets within 41 +/- 3 min with an average weight of 8 +/- 1 mg (n = 7). These thrombi were enriched in both platelets and fibrin and were adherent at the site of transmural vascular injury as determined by light and electron microscopy. To determine the model's sensitivity to antiplatelet drugs, aspirin or a thromboxane (TxA2) receptor antagonist (ifetroban) were administered 15 min before electrical stimulation. Thrombus weight was reduced 58% by aspirin (10 mg/kg, i.v.) and 74% by ifetroban (1 mg/kg + 1 mg/kg per hr, i.v.). Both drugs also improved CBF and decreased vascular occlusion. Ferrets were more sensitive than rats to aspirin's inhibition of collagen-induced platelet aggregation as determined ex vivo in whole blood. Separate in vitro platelet aggregation studies revealed species differences in reactivity to U-46619 (TxA2 receptor agonist) and collagen in the order of human > ferret > rat, with relatively lesser variations in ADP responses. These studies identify the ferret as a useful species for evaluating antithrombotic drugs in a model in which aspirin is efficacious. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Animals; Aspirin; Blood Platelets; Bridged Bicyclo Compounds, Heterocyclic; Carotid Arteries; Carotid Artery Injuries; Collagen; Disease Models, Animal; Electric Stimulation; Ferrets; Fibrin; Humans; In Vitro Techniques; Male; Microscopy, Electron; Microscopy, Electron, Scanning; Oxazoles; Platelet Aggregation; Platelet Aggregation Inhibitors; Prostaglandin Endoperoxides, Synthetic; Prothrombin Time; Rats; Rats, Sprague-Dawley; Regional Blood Flow; Species Specificity; Thrombosis; Thromboxane A2; Vasoconstrictor Agents | 1996 |
Induction mechanism of small intestinal lesions caused by intravenous injection of endotoxin in rats.
The pathogenesis of intestinal damage caused by bolus intravenous injection of endotoxin (ETX; 3 mg/kg) was investigated. Administration of ETX to rats induced reddish discoloration suggestive of bleeding, increased hemoglobin amounts, and leakage of plasma protein in the intestine. However, light microscopic examination of the intestine demonstrated blood congestion of the microvessels. Plasma accumulation was partially inhibited by combined pretreatment with a histamine H1 antagonist and a serotonin (5-HT) antagonist. Neither a 5-lipoxygenase inhibitor, a soybean trypsin inhibitor, nor atropine was observed to inhibit plasma accumulation. Both the intestinal leakage of plasma and the accumulation of hemoglobin were completely inhibited by indomethacin, a selective thromboxane A synthetase inhibitor (OKY 1581), and a stable PGI2 analogue (beraprost). Intravital microscopic observation of the microvessels of the small intestinal villi demonstrated microthrombus formation within several minutes after the injection of ETX, and pretreatment with OKY 1581 attenuated the formation of microthrombus. Platelet counts decreased significantly 10 min after ETX administration, and the decrease was not inhibited by pretreatment with either OKY 1581 or beraprost. Prothrombin time (PT) and activated partial thromboplastin time (APTT) were not prolonged. These observations thus suggest that microcirculatory disturbances by platelet thrombus, which are mediated by thromboxane A2 at least in part, play an important role in ETX-induced intestinal damage. Topics: Animals; Capillary Leak Syndrome; Cyclooxygenase Inhibitors; Endotoxins; Enzyme Inhibitors; Epoprostenol; Escherichia coli; Gastrointestinal Hemorrhage; Indomethacin; Intestine, Small; Male; Methacrylates; Microcirculation; Platelet Activation; Rats; Rats, Sprague-Dawley; Shock, Septic; Specific Pathogen-Free Organisms; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1996 |
Isolation and purification of anticardiolipin antibody from plasma of a patient with antiphospholipid syndrome: induced generation of platelet thromboxane A2 synthesis.
Antiphospholipid antibodies, particularly anticardiolipin antibodies (aCL) are autoantibodies frequently detected in the serum of patients with systemic lupus erythematosus (SLE) and the primary antiphospholipid antibody syndrome (PAPS). These patients commonly suffer from thrombosis, recurrent fetal loss and thrombocytopenia. Since platelet aggregation is pivotal in the genesis of thrombosis, we tested the hypothesis that perturbation of platelet membrane by aCL/beta 2-glycoprotein (aCL/beta 2GP) complex could trigger the biosynthesis of TXA2, a proaggregatory metabolite of AA. The preincubation of 14C-arachidonic acid (14C-AA)-labeled platelet pellets (14C-PP) from normal individuals with aCL alone followed by incubation with thrombin, resulted in a moderate increase in platelet thromboxane B2 (14C-TXB2) biosynthesis when compared to controls (without aCL). Similar incubations with beta 2GP-I alone resulted in negligible 14C-TXB2 biosynthesis. In contrast, the preincubations of normal 14C-PP with aCL/beta 2GP-I complex resulted in marked thrombin-induced TXB2 biosynthesis, underscoring the requirement of beta 2GP-I in aCL-induced platelet TXB2 biosynthesis. Taken together, these results are consistent with the view that aCL/beta 2GP-I platelet interactions do play a role, at least in part, in platelet hyperactivity and thrombosis in antiphospholipid antibody syndrome. Topics: Adult; Antibodies, Anticardiolipin; Antiphospholipid Syndrome; Aorta; beta 2-Glycoprotein I; Blood Platelets; Chromatography, Affinity; Chromatography, Ion Exchange; Dose-Response Relationship, Drug; Drug Synergism; Female; Glycoproteins; Humans; Nerve Tissue Proteins; Thrombin; Thrombosis; Thromboxane A2; Thromboxane B2 | 1996 |
Prostacyclin and thromboxane A2 synthesis are increased in acute lower limb ischaemia.
Prostacyclin (PGI2) and thromboxane A2 (TXA2) play an important role in the pathophysiology of various cardiovascular diseases. The balance between PGI2 and TXA2 regulates the interaction between platelets and the vessel wall in vivo. In this study we measured PGI2 and TXA2 synthesis by analysing their urinary index metabolites 2,3-dinor-6-keto-PGF1 alpha and 11-dehydro-TXB2, respectively, in acute (10 patients) and chronic (10 patients) lower limb ischaemia. Both PGI2 and TXA2 synthesis were increased about two-fold in patients with acute lower limb ischaemia compared to chronic lower limb ischaemia. However, the PGI2/TXA2 ratio was more or less the same in acute and chronic lower limb ischaemia. In patients with acute lower limb ischaemia caused by thrombotic occlusion, PGI2 and TXA2 formation were about two times higher than in patients with acute lower limb ischaemia caused by embolic occlusion. Elevation of PGI2 and TXA2 synthesis in acute lower limb ischaemia may reflect increased platelet-vascular wall interactions without changing the PGI2/TXA2 ratio. Topics: 6-Ketoprostaglandin F1 alpha; Aged; Aged, 80 and over; Chronic Disease; Creatinine; Epoprostenol; Extremities; Female; Humans; Ischemia; Male; Middle Aged; Thrombosis; Thromboxane A2; Thromboxane B2 | 1996 |
Comparison of antithrombotic effects of GPIIb-IIIa receptor antagonist and TXA2 receptor antagonist in the guinea-pig thrombosis model: possible role of TXA2 in reocclusion after thrombolysis.
The effects of Ro 44-9883, a new specific antagonist of platelet glycoprotein IIb-IIIa receptor, on thrombus formation and reocclusion after thrombolysis induced by tissue-type plasminogen activator (t-PA) were compared with those of vapiprost, a thromboxane (TX) A2 receptor antagonist, using a photochemically-induced thrombosis model in the guinea-pig femoral artery. Pretreatment with Ro 44-9883 (5, 10 and 20 micrograms/kg/min, i.v.) prolonged the time required to occlude the artery in a dose-dependent manner. Ro 44-9883 at 10 and 20 micrograms/kg/min significantly inhibited ex vivo platelet aggregation in whole blood induced by collagen, ADP or U46619. Vapiprost 0.3 mg/kg inhibited thrombus formation and platelet aggregation induced by collagen or U46619, to the same extent as Ro 44-9883 at the higher doses. In the thrombolysis study, Ro 44-9883 at the higher doses given as comedication with t-PA reduced the time to achieve reperfusion and increased the vascular patency after successful reperfusion. Vapiprost also significantly reduced the time to reperfusion and prevented reocclusion. However, the vascular patency after thrombolysis by t-PA with vapiprost was significantly increased compared with Ro 44-9883. Ro 44-9883 inhibited platelet aggregation, but did not prevent TXA2 formation in platelets. Thus, vascular contraction mediated by platelet-derived TXA2 may be responsible for lower efficacy of Ro 44-9883 against reocclusion compared with vapiprost. These results indicate that not only platelet aggregation but also vasoconstriction may contribute to reocclusion after t-PA-induced thrombolysis in the guinea-pig. Topics: Acetates; Animals; Biphenyl Compounds; Femoral Artery; Guinea Pigs; Heptanoic Acids; Platelet Aggregation Inhibitors; Platelet Membrane Glycoproteins; Receptors, Thromboxane; Thrombosis; Thromboxane A2; Tyrosine | 1995 |
Vitamin E deficiency increases serum thromboxane A2, platelet arachidonate and lipid peroxidation in male Sprague-Dawley rats.
This study was designed to determine whether dietary linoleate and all-rac-alpha-tocopheryl acetate (vitamin E) interact to affect serum thromboxane A2 (TXA2) and prostacyclin (PGI2) status and therefore, thrombogenic potential. 6 groups of 12 weanling male Sprague-Dawley rats were fed semipurified diets containing 11 or 18% of energy from linoleate and 0, 100 or 5000 mg vitamin E/kg diet for 10 weeks. Platelet and serum alpha-tocopherol concentrations increased logarithmically with increasing dietary vitamin E. Serum TXA2, measured as TXB2, platelet arachidonate and thiobarbituric acid reactive substances were significantly greater in the vitamin E deficient groups than in groups receiving vitamin E (p < 0.05). Serum PGI2 levels, determined as 6-keto-PGF1 alpha, were not affected by diets. No interaction was found between dietary linoleate and vitamin E. However, vitamin E supplementation produced significantly less serum TXB2 than did vitamin E deficient diets (p < 0.05). Vitamin E deficiency may be prothrombogenic by increasing platelet arachidonate, lipid peroxidation and serum TXA2 levels while vitamin E supplementation at levels used in this study may decrease such effects. Topics: 6-Ketoprostaglandin F1 alpha; Animals; Arachidonic Acid; Blood Platelets; Epoprostenol; Linoleic Acid; Linoleic Acids; Lipid Peroxidation; Male; Rats; Rats, Sprague-Dawley; Thiobarbituric Acid Reactive Substances; Thrombosis; Thromboxane A2; Thromboxane B2; Vitamin E; Vitamin E Deficiency | 1994 |
Antithrombotic effects of KW-3635, a thromboxane A2-receptor antagonist, in guinea pigs.
Antithrombotic effects of KW-3635, a newly synthesized thromboxane (TX) A2-receptor antagonist, were studied in guinea pigs. In the extracorporeal circulation thrombosis model, the shunt was filled with thrombi, and reduction of platelet count and increase in plasma TXA2 concentration were observed. KW-3635 (30 and 100 mg/kg, p.o.) inhibited the thrombus formation in the shunt and prevented the decrease in platelet count in the circulating blood without affecting the red blood cell count. BM13,505 (30, 100 mg/kg, p.o.), another TXA2-receptor antagonist, and ticlopidine (300 mg/kg, p.o.), an antiplatelet drug, also inhibited the thrombus formation, while aspirin (10, 300 mg/kg, p.o.) did not. Peripheral arterial occlusive disease was induced by injection of sodium laurate into the femoral artery in guinea pigs. Daily oral administration of KW-3635 (3-30 mg/kg) significantly prevented the progression of vascular lesions. BM13,505 (3-30 mg/kg, p.o.) and ticlopidine (100 mg/kg, p.o.) also ameliorated the vascular lesions, whereas aspirin (10, 100 mg/kg, p.o.) did not. KW-3635 at concentrations up to 10(-4) M did not affect coagulation parameters in vitro. These results suggest that TXA2 is involved in the pathogenesis of arterial thrombotic and ischemic disorders. KW-3635 may be useful for the treatment of thrombotic disease and peripheral arterial occlusive diseases. Topics: Animals; Arterial Occlusive Diseases; Aspirin; Benzimidazoles; Benzoxepins; Blood Coagulation; Blood Platelets; Disease Models, Animal; Erythrocyte Count; Femoral Artery; Guinea Pigs; Lauric Acids; Male; Phenylacetates; Platelet Count; Sulfonamides; Thrombosis; Thromboxane A2; Thromboxanes; Ticlopidine | 1994 |
Enhancement of thrombotic arterial occlusion following cholesterol feeding in the guinea-pig: a role for thromboxane A2.
We have developed a photochemical model to induce thrombotic occlusion of the guinea-pig femoral artery. Using this model, we investigated the effect of cholesterol feeding on arterial occlusion time in the guinea-pig. Animals were divided into two groups, one on standard diet and the other on standard diet containing 0.5% cholesterol for 3 weeks. The time for femoral artery occlusion was significantly shorter (p < 0.05) in cholesterol fed animals as compared to the control group. In vitro collagen-, U-46619- (a thromboxane A2 adenosine diphosphate analogue) and (ADP)-induced platelet aggregation responses in whole blood in cholesterol-fed animals were increased 13-, 10- and 4-fold, respectively. U-46619- and collagen-induced washed platelet aggregation responses were also significantly enhanced by cholesterol feeding (p < 0.01). Further, TXA2 generation by collagen-stimulated washed platelets in cholesterol-fed animals increased similar to the platelet aggregation responses. However, platelet-activating factor (PAF)-induced platelet aggregation in whole blood was relatively unaffected by cholesterol feeding. 11-dehydro TXB2 levels in plasma were increased significantly by cholesterol feeding. Our observations suggest that increased plasma TXA2 level and platelet aggregation response to TXA2 and stimulated TXA2 synthesis in platelets play a role in enhanced arterial occlusion in cholesterol fed guinea-pigs. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Adenosine Diphosphate; Animals; Arterial Occlusive Diseases; Cholesterol, Dietary; Collagen; Femoral Artery; Guinea Pigs; Male; Photochemistry; Platelet Activating Factor; Platelet Aggregation; Prostaglandin Endoperoxides, Synthetic; Thrombosis; Thromboxane A2; Time Factors | 1994 |
Inhibitory effect of KW-3635, a new thromboxane A2-receptor antagonist, on arterial thrombosis in guinea pigs.
The antithrombotic effects of the thromboxane (TX) A2-receptor antagonist and aspirin were determined using a photochemically-induced arterial thrombosis model in the femoral arteries of guinea pigs. KW-3635 (sodium (E)-11-[2-(5,6-dimethyl-1-benzimidazolyl)ethylidene]-6,11- dihydrodibenz[b,e]-oxepine-2-carboxylate monohydrate) and BM-13505, both of which are TXA2-receptor antagonists, and aspirin inhibited the thrombus formation at the doses that inhibited the ex vivo platelet aggregation induced by sodium arachidonate (100 microM) or collagen (3 micrograms/ml). These results support the notion that TXA2 is an important mediator in the present model of arterial thrombogenesis. Topics: Animals; Arachidonic Acid; Aspirin; Benzimidazoles; Benzoxepins; Collagen; Disease Models, Animal; Femoral Artery; Guinea Pigs; Male; Phenylacetates; Platelet Aggregation; Sulfonamides; Thrombosis; Thromboxane A2 | 1993 |
Thromboxane and prostacyclin synthesis in experimental pancreas transplantation. Changes in parenchymal and vascular prostanoids.
The principal causes of failure of a pancreas transplant are rejection and vascular thrombosis. There is an unusually high attrition rate for pancreas transplants, but study models have been difficult to develop. In a rat model that allows study of acute rejection to the exclusion of nonspecific effects of transplant surgery on the pancreas, in vitro synthesis of prostacyclin (PGI2) and thromboxane A2 (TXA2) by transplanted pancreas and the blood vessels transplanted with it was measured using an RIA for their stable hydrolysis products 6-keto-prostaglandin F1 alpha and thromboxane B2 (TXB2). TXB2 synthesis was significantly greater in allotransplanted pancreas than isotransplanted pancreas from the 5th day after transplantation. Rejection was complete in the allografted group 7-9 days after transplantation. 6-Keto-prostaglandin F1 alpha synthesis was similar in the pancreas for both allografts and isografts. Similar changes were seen in aorta, celiac artery, superior mesenteric artery, and portal vein transplanted with the pancreas. In the transplanted aorta, TXB2 was significantly greater in the allograft group from the third posttransplant day. A group of CsA-treated allografts sampled after 9 days had transplanted pancreatic parenchymal and vascular prostanoid synthesis in the isograft range. The changes in PGI2 and TXA2 synthesis that accompany cellular rejection may mediate vascular failure in rejecting pancreas transplants, and changes in PGI2 and TXA2 synthesis in blood vessels transplanted with the pancreas could promote early vascular thrombosis. Topics: 6-Ketoprostaglandin F1 alpha; Acute Disease; Animals; Aorta; Celiac Artery; Cyclosporine; Epoprostenol; Graft Rejection; Male; Mesenteric Artery, Superior; Pancreas; Pancreas Transplantation; Rats; Rats, Inbred Strains; Thrombosis; Thromboxane A2; Thromboxane B2; Transplantation, Homologous; Transplantation, Isogeneic; Vena Cava, Inferior | 1993 |
Comparison of the inhibitory effects of the TXA2 receptor antagonist, vapiprost, and other antiplatelet drugs on arterial thrombosis in rats: possible role of TXA2.
The antithrombotic effect of the thromboxane A2 receptor antagonist, vapiprost, was compared with those of other antiplatelet drugs using an arterial thrombosis model which utilized photochemical reaction in the rat femoral artery. Vapiprost prolonged the time required to occlude the artery with thrombus and inhibited collagen-induced rat platelet aggregation in whole blood ex vivo, in a dose-dependent manner. The potency ranking of antithrombotic effect was vapiprost > ketanserin (serotonin 5-HT2 receptor antagonist) >> ticlopidine (inhibitor of ADP-induced platelet aggregation) = dipyridamole (adenosine uptake inhibitor) > aspirin (cyclooxygenase inhibitor). On the other hand, the ranking of antiplatelet effect was ticlopidine > or = vapiprost > or = aspirin. Ketanserin and dipyridamole were ineffective. Relative to their antiplatelet effect, vapiprost and ketanserin had powerful antithrombotic effects. It is possible that the potent antithrombotic effects of vapiprost and ketanserin in vivo reflect the ability of these drugs to inhibit mediator-induced vascular contractions in addition to platelet aggregation. The results of the present study also suggest that TXA2 may play an important role in thrombogenesis in rats. Topics: Animals; Arteries; Biphenyl Compounds; Fibrinolytic Agents; Heptanoic Acids; Male; Photochemistry; Platelet Aggregation Inhibitors; Rats; Rats, Wistar; Receptors, Thromboxane; Thrombosis; Thromboxane A2 | 1992 |
A new bioactive molecule for improving vascular graft patency: exploratory trials in dogs.
Myxalin is a new bioactive molecule that we have isolated from the culture medium of Myxococcus xanthus, a non-pathogenic Gram negative bacterium. This glycopeptide possesses an antithrombotic effect in vivo and has been shown to promote human endothelial cell growth in vitro. With the object of exploring its ability to improve vascular graft healing and patency, myxalin was immobilized on 6 mm diameter knitted polyester prostheses using gelatin as a carrier, and the prosthesis was then implanted as an infrarenal abdominal arterial substitute in dogs for a period of 2 weeks. Two additional series of implantations were conducted for control purposes: one with gelatin-coated prostheses without myxalin, the other following normal preclotting of the polyester grafts. In order to select adequate sterilization conditions which can preserve the biological activity of myxalin, the prostheses were sterilized according to 3 different sterilization processes (gamma radiation and ethylene oxide either at 63 degrees C or 37 degrees C). At the sacrifice, all grafts were patent. The myxalin treated prostheses exhibited improved blood compatibility in terms of fewer thrombotic deposits and significant inhibition of platelet and fibrinogen uptake on their luminal surfaces. In addition, the development of a thin collagenous internal capsule with endothelial cells secreting high levels of prostacyclin was observed at both anastomoses of the myxalin-treated grafts sterilized by gamma radiation. Topics: 6-Ketoprostaglandin F1 alpha; Animals; Bacterial Proteins; Blood Platelets; Blood Vessel Prosthesis; Dogs; Fibrinogen; Glycopeptides; Microscopy, Electron, Scanning; Myxococcus xanthus; Thrombosis; Thromboxane A2; Vascular Patency | 1992 |
Evidence for a role in thrombus stabilization for thromboxane A2 in human platelet deposition on collagen.
The role of thromboxane A2 (TxA2) in platelet deposition onto collagen was studied in flowing whole heparinized human blood in vitro by using a cyclooxygenase inhibitor, aspirin, and a TxA2 receptor antagonist, GR32191B. Previous studies have demonstrated a role for TxA2 in platelet aggregation in citrated plasma, and for platelet deposition in flowing citrated human and rabbit blood, but not in flowing heparinized rabbit blood. In contrast with the literature regarding rabbit blood, aspirin was demonstrated to be effective in reducing platelet accumulation in heparinized human blood, as was GR32191B. The temporal pattern of the platelet deposition, which reached an asymptote with TxA2 inhibition at 2.0 minutes but did not do so without inhibition, suggested that TxA2 plays a role in thrombus stabilization. The reduction in platelet deposition (which included some aggregation) seen with aspirin and GR32191B in the first 1.5 minutes of perfusion indicated that some inhibition of platelet recruitment occurred. Scanning electron microscopy revealed that less fibrin was present in thrombi derived from GR32191B-treated heparinized blood than in thrombi derived from control heparinized blood. No fibrin formation was observed in citrated blood with or without TxA2 inhibition. It is proposed that, in addition to its role as a mediator of platelet recruitment, TxA2 is involved in the stabilization of platelet-platelet interactions in the thrombus, perhaps by enhancing local fibrin formation or binding. Topics: Aspirin; Biphenyl Compounds; Blood Platelets; Citrates; Collagen; Heparin; Heptanoic Acids; Humans; In Vitro Techniques; Kinetics; Microscopy, Electron, Scanning; Platelet Adhesiveness; Receptors, Prostaglandin; Receptors, Thromboxane; Thrombosis; Thromboxane A2 | 1992 |
[Anti-thrombotic and anti-atherogenic action of eicosapentaenoic acid].
Epidemiological studies performed in Greenland Eskimos and Japanese indicated that ingestion of fish meat rich in omega-3 polyunsaturated fatty acids, such as eicosapentaenoic acid (EPA), reduces the incidence of cerebro- and cardiovascular thrombotic diseases. Therefore, the effect of administration of purified EPA, derived from sardine oil, on hemorrheological properties and serum lipids in patients with thrombotic disorders was studied. Dietary supplementation of EPA resulted in decrease in platelet aggregability with reduction of TXA2 production, prolongation of bleeding time, decrease of platelet adhesiveness, increase of red cell deformability, and improvement of serum lipid concentration. Decrease in platelet TXA2 production may be ascribed to the decrease in arachidonate (AA) content of platelet membrane, inhibition of AA release from platelet membrane and competitive inhibition of AA metabolism at the level of cyclo-oxygenase, by EPA. Administration of EPA increased PGI2 production in the rat thoracic aorta and co-culture of rat aorta smooth muscle cell with EPA also increased PGI2 production, but suppressed smooth muscle cell proliferation. This may be explained by a EPA-derived peroxide stimulation of cyclo-oxygenase activity. Administration of EPA decreased platelet-derived growth factor (PDGF) production in rat peritoneal macrophages. In EPA-rich peritoneal macrophages from rat given EPA, incorporation of acetyl LDL and accumulation of cholesterol in macrophages decreased. This may indicate that dietary supplementation of EPA suppress foam cell formation. An interesting finding is that EPA-rich LDL obtained from rabbits given EPA is less susceptible to Cu(2+)-catalyzed oxidative modification. This seems to indicate that EPA may also suppress lipid peroxidation of lipoprotein.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Arachidonic Acid; Arteriosclerosis; Eicosapentaenoic Acid; Epoprostenol; Fibrinolytic Agents; Humans; Lipid Peroxidation; Lipids; Platelet Aggregation Inhibitors; Thrombosis; Thromboxane A2 | 1992 |
Enhancement of tissue-type plasminogen activator-induced thrombolysis and prevention of reocclusion by sodium (E)-11-[2-(5,6-dimethyl-1-benzimidazolyl)- ethylidene]-6,11-dihydrodibenz[b,e]oxepine-2-carboxylate monohydrate in a canine model of femoral thro
The purpose of this study was to examine whether the blockade of thromboxane A2 (TxA2)/prostaglandin H2 (PGH2) receptor by the selective TxA2/PGH2 receptor antagonist KW-3635 (sodium (E)-11-[2-(5,6-dimethyl-1-benzimidazolyl)ethylidene]-6,11- dihydrodibenz[b,e]oxepine-2-carboxylate monohydrate, CAS 127166-41-0) is effective in enhancing tissue-type plasminogen activator (tPA)-induced thrombolysis and preventing reocclusion in a model of femoral artery thrombosis in anesthetized dogs. The thrombus was formed by inserting a copper coil into the femoral artery. Sodium heparin (100 U/kg i.v.) was administered shortly after the formation of thrombus. All dogs received i.v. tPA at a dose of 20 micrograms/kg/min starting 60 min after the formation of the occlusive thrombus for up to 60 min if necessary, to achieve reperfusion. After 30 min of thrombotic occlusion, the animals received vehicle (Group I, controls, n = 9) or KW-3635 (Group II, 0.3 mg/kg bolus i.v. + 0.3 mg/kg/h infusion, n = 9; Group III, 1 mg/kg bolus i.v. + 1 mg/kg/h infusion, n = 9) and the infusion of either vehicle or KW-3635 was continued thereafter throughout the experiment. The time to reperfusion in Group I was 37.3 +/- 5.2 min, while those in Group II and Group III were 25.3 +/- 6.2 min (p greater than 0.05) and 17.3 +/- 3.1 min (p less than 0.05), respectively. Reocclusion occurred within 4 h in 100% of Group I, whereas the incidence of reocclusion was reduced to 67% in Group II and to 0% in Group III. These data suggest that endogenous TxA2 generation is involved in lysis and rethrombosis during thrombolytic therapy by tPA.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Animals; Arterial Occlusive Diseases; Benzimidazoles; Benzoxepins; Dogs; Female; Femoral Artery; Fibrinolytic Agents; Male; Platelet Aggregation; Prostaglandin Endoperoxides, Synthetic; Recurrence; Thrombosis; Thromboxane A2; Tissue Plasminogen Activator | 1991 |
Effect of preservation on blood flow and production of prostacyclin and thromboxane A2 in canine segmental pancreatic autografts.
Topics: 6-Ketoprostaglandin F1 alpha; Animals; Dogs; Epoprostenol; Organ Preservation; Pancreas; Pancreas Transplantation; Regional Blood Flow; Splenic Artery; Thrombosis; Thromboxane A2; Thromboxane B2 | 1991 |
[Experimental study of the pathological mechanism of thromboxane A2 in alkaline ocular burn].
The authors investigated the pathological mechanism of thromboxane A2 in alkaline burn of the eye. Thromboxane B2 (metabolic product of thromboxane A2) in aqueous humor of the burned eye significantly increased in 30 minutes after the burn. The pathological change was the formation of thrombosis 2 hours after the alkaline burn, especially in the anterior segment. The role of thromboxane A2 in alkaline burn of the eye was ascertained. Topics: Alkalies; Animals; Anterior Eye Segment; Aqueous Humor; Burns, Chemical; Eye Burns; Male; Rabbits; Thrombosis; Thromboxane A2; Thromboxane B2 | 1990 |
Increased thromboxane formation in patients with antiphospholipid syndrome.
Thirty-one patients with IgG antibodies to cardiolipin (ACLA) were studied to determine their in vivo formation of the platelet aggregating and vasoconstricting substance thromboxane A2 (TxA2) and the platelet inhibiting and vasodilating substance prostacyclin (PGI2). This was done by measurements in urine of their enzymatically formed metabolites 2,3-dinor-TxB2 and 2,3-dinor-6-keto-PGF1 alpha, respectively, using gas chromatography-mass spectrometry. It is demonstrated that patients with IgG ACLA have a highly significant increase in the biosynthesis of TxA2 compared with age-matched healthy controls (807 +/- 163 [SEM] vs. 230 +/- 15 pg mg-1 creatinine, P = 0.0000005). A significant increment of the formation of PGI2 was also found (189 +/- 23 (SEM) vs. 125 +/- 11 pg mg-1 creatinine, P = 0.03), although this was much less pronounced than that for TxA2. We conclude that the highly increased formation of TxA2, reflecting platelet activation, in patients with IgG ACLA is of pathophysiologic relevance for their tendency to arterial and venous thrombosis and hence that they should be considered for prophylactic treatment with inhibitors of TxA2 formation, like aspirin. Topics: Adult; Autoimmune Diseases; Cardiolipins; Epoprostenol; Female; Humans; Male; Middle Aged; Phospholipids; Platelet Activation; Syndrome; Thrombosis; Thromboxane A2 | 1990 |
Influence of transluminal angioplasty on the prostanoid release from the arterial wall.
Vasospasm and thrombosis complicate percutaneous transluminal angioplasty (PTA). To study if the release of the prostanoids PGI2 and TxA2 are affected by PTA, the following experiment was undertaken: In ten rabbits, the upper or lower half of the aorta was randomised either to transluminal angioplasty or control segment. After excision the segments were simultaneously but separately perfused ex vivo with Hank's balanced salt solution for five consecutive 15 min periods. Arachidonic acid was added to the perfusate for the last 15 min period. PGI2 and TxA2 were measured by radioimmunoassay in the perfusate as the stable degradation products 6-keto-PGF1 alpha and TxB2. After perfusion, the two aortic segments were prepared for scanning electron microscopy (SEM). Angioplasty decreased the basic release of PGI2 as well as the response to arachidonic acid. This is likely to be due to endothelial denudation as seen by SEM. The release of TxA2 from the vessel wall was very low and was not increased by dilatation. The influence of angioplasty on the prostanoid system may be of importance in the complications of vasospasm and thrombosis. Topics: Angioplasty, Balloon; Animals; Aorta; Endothelium, Vascular; Epoprostenol; Female; Male; Microscopy, Electron, Scanning; Rabbits; Thrombosis; Thromboxane A2 | 1990 |
Activation of platelets by eosinophil granule proteins.
Two of the four principal cationic proteins of the eosinophil granule, major basic protein (MBP) and eosinophil peroxidase (EPO), were shown to be platelet agonists. Both MBP and EPO evoked a dose-dependent nonlytic secretion of platelet 5-hydroxytryptamine in unstirred platelet suspensions even in the presence of 10 microM indomethacin. MBP also evoked secretion of platelet alpha granule and lysosome components. Secretion by MBP and EPO was inhibited by 1 microM PGE1, but the nature of the inhibition differed from that observed with thrombin. Thus, MBP and EPO can be classified as strong platelet agonists with a distinct mechanism of activation. Topics: Alprostadil; Blood Proteins; Eosinophil Granule Proteins; Eosinophils; Humans; In Vitro Techniques; Peroxidases; Platelet Activation; Ribonucleases; Serotonin; Thrombosis; Thromboxane A2 | 1990 |
Prevention of thrombosis and rethrombosis. New approaches.
Topics: Animals; Antithrombins; Cardiology; Fibrinogen; Humans; Platelet Aggregation Inhibitors; Protein C; Recombinant Proteins; Recurrence; Thrombosis; Thromboxane A2 | 1990 |
Effect of thromboxane antagonism on recanalization during streptokinase-induced thrombolysis in anesthetized monkeys.
The effect of a selective thromboxane A2 (TxA2) receptor antagonist, SQ 30,741, on streptokinase-induced thrombolysis was examined in pentobarbital-anesthetized cynomolgus monkeys. The intimal surface of a stenosed carotid artery was stimulated with 100 microA anodal current to produce an occlusive thrombus. After 45 min of zero blood flow, a 1-h intraarterial (i.a.) infusion of streptokinase (680 U/min) was injected proximal to the thrombus. Five minutes before streptokinase (SK) intravenous (i.v.) infusions of heparin (200 U/kg + 120 U/h) and either SQ 30,741 (2 mg/kg + 2 mg/kg/min, n = 8) or vehicle (1 ml/h saline, n = 7) were started and maintained for 3 h. In four monkeys not given streptokinase or heparin, no recanalization was detected and occlusive thrombi were observed after 3 h. All animals receiving streptokinase were recanalized. SQ 30,741 had no effect on return of flow during streptokinase infusion, but increased average reflows during the second (60%, p less than 0.05) and third hours (159%, p less than 0.01). Average blood flows were decreased from the second to third hours with vehicle (p less than 0.001) and remained stable with SQ 30,741. Thromboxane antagonism also increased minimal blood flows during the third hour (438%, p less than 0.01) and decreased the total time reoccluded by 73% (p less than 0.05). However, SQ 30,741 had no effect on the time to recanalization, the maximum reflow, and both number of animals reoccluded and average number of reocclusions. Fibrinogen levels were equivalently diminished (8%, p less than 0.05), and platelet counts were unaffected in both treatment groups.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Carotid Artery Diseases; Female; Macaca fascicularis; Male; Regional Blood Flow; Streptokinase; Thrombosis; Thromboxane A2 | 1989 |
The short-term blood biocompatibility of poly(hydroxyethyl methacrylate-co-methyl methacrylate) in an in vitro flow system measured by digital videomicroscopy.
An in vitro flow system for short-term blood biocompatibility testing of solution-castable polymeric biomaterials was developed. This system was relatively free of artefacts resulting from blood contact with materials other than the test material itself. In conjunction with epifluorescence videomicroscopy and digital image processing, this method provided a high resolution, quantitative, continuous analysis of platelet adhesion, aggregation, thrombus formation, and embolization on the biomaterial surface. This system was well suited for performing biochemical assays on post-contact blood for assessment of platelet activation and release as additional measures of the thrombogenicity of the test material. This method for biomaterials evaluation in vitro was demonstrated by a detailed examination of copolymers of hydroxyethyl methacrylate (HEMA) and methyl methacrylate (MMA). Videomicroscopic analysis of fluorescently labelled platelets adhering per unit area of the polymer surface after 5 min of flow at a wall shear rate of 500 s-1 showed a dramatic decrease with increasing HEMA fraction in the polymer. The release of serotonin and thromboxane A2 by platelets decreased with increasing HEMA fraction. Reflection interference contrast microscopy was used to examine focal contacts of platelets on the copolymer surfaces as a qualitative measure of the platelet-surface interaction. A polymer-dependent gradation in contact extent and morphology was observed, ranging from large contacts on P(MMA) to none on P(HEMA). Topics: Biocompatible Materials; Embolism; Image Processing, Computer-Assisted; In Vitro Techniques; Methacrylates; Microscopy, Fluorescence; Platelet Activation; Polyhydroxyethyl Methacrylate; Rheology; Serotonin; Thrombosis; Thromboxane A2; Videotape Recording | 1989 |
Effect of selective endoperoxide/thromboxane A2 receptor antagonism with sulotroban on tPA-induced thrombolysis in a rabbit model of femoral arterial thrombosis.
The thrombolytic efficacy of recombinant tissue-type plasminogen activator (tPA) in the presence and absence of the selective endoperoxide/thromboxane A2 (TXA2) receptor antagonist, sulotroban (BM 13.177, SK&F 95587) was studied in a model of femoral artery thrombosis in the anesthetized rabbit. The thrombus was formed by injection of thrombin, CaCl2 and whole blood into an isolated segment of the femoral artery. After 30 min of stable thrombotic occlusion of the femoral artery, tPA was infused IV for 90 min at doses of 5.0, 7.5 and 10.0 micrograms/kg/min. In other experiments, sulotroban was administered as a bolus dose of 1 mg/kg/IV, followed by a constant infusion of 1 mg/kg/hr concurrent with tPA infusion. Sulotroban had no effect on the incidence of tPA-induced reperfusion at any dose studied or on residual clot weight. However, at a tPA dose of 10 micrograms/kg/min, IV lysis time was reduced in sulotroban treated animals from 65 min to 29 min (P less than 0.05), and the magnitude of femoral artery blood flow achieved as a result of tPA-induced reperfusion was greater in sulotroban-treated animals. These data suggest that adjunctive therapy with a selective endoperoxide/TXA2 antagonist improves the response to tPA when tPA is administered at a maximal or near maximally effective pharmacological dose. Topics: Animals; Femoral Artery; Fibrinolytic Agents; In Vitro Techniques; Male; Platelet Aggregation; Prostaglandin Endoperoxides; Rabbits; Receptors, Prostaglandin; Receptors, Thromboxane; Recombinant Proteins; Regional Blood Flow; Sulfonamides; Thrombosis; Thromboxane A2; Tissue Plasminogen Activator | 1989 |
Mathematical analysis of mural thrombogenesis. Concentration profiles of platelet-activating agents and effects of viscous shear flow.
The concentration profiles of adenosine diphosphate (ADP), thromboxane A2 (TxA2), thrombin, and von Willebrand factor (vWF) released extracellularly from the platelet granules or produced metabolically on the platelet membrane during thrombus growth, were estimated using finite element simulation of blood flow over model thrombi of various shapes and dimensions. The wall fluxes of these platelet-activating agents were estimated for each model thrombus at three different wall shear rates (100 s-1, 800 s-1, and 1,500 s-1), employing experimental data on thrombus growth rates and sizes. For that purpose, whole human blood was perfused in a parallel-plate flow chamber coated with type l fibrillar human collagen, and the kinetic data collected and analyzed by an EPl-fluorescence video microscopy system and a digital image processor. It was found that thrombin concentrations were large enough to cause irreversible platelet aggregation. Although heparin significantly accelerated thrombin inhibition by antithrombin lll, the remaining thrombin levels were still significantly above the minimum threshold required for irreversible platelet aggregation. While ADP concentrations were large enough to cause irreversible platelet aggregation at low shear rates and for small aggregate sizes, TxA2 concentrations were only sufficient to induce platelet shape change over the entire range of wall shear rates and thrombi dimensions studied. Our results also indicated that the local concentration of vWF multimers released from the platelet alpha-granules could be sufficient to modulate platelet aggregation at low and intermediate wall shear rates (less than 1,000 s-1). The sizes of standing vortices formed adjacent to a growing aggregate and the embolizing stresses and the torque, acting at the aggregate surface, were also estimated in this simulation. It was found that standing vortices developed on both sides of the thrombus even at low wall shear rates. Their sizes increased with thrombus size and wall shear rate, and were largely dependent upon thrombus geometry. The experimental observation that platelet aggregation occurred predominantly in the spaces between adjacent thrombi, confirmed the numerical prediction that those standing vortices are regions of reduced fluid velocities and high concentrations of platelet-activating substances, capable of trapping and stimulating platelets for aggregation. The average shear stress and normal stress, as well as the torque, acting t Topics: Adenosine Diphosphate; Antithrombin III; Blood Platelets; Collagen; Computer Simulation; Heparin; Humans; Kinetics; Mathematics; Microscopy, Fluorescence; Models, Biological; Platelet Activation; Regional Blood Flow; Thrombin; Thrombosis; Thromboxane A2; Video Recording; Viscosity; von Willebrand Factor | 1989 |
Fate of polyester arterial prostheses implanted as thoraco-abdominal by-passes in dogs: haematology, pathology, and biochemistry.
This study was designed to evaluate the haematological, pathological and biochemical characteristics of the flow surface in polyester arterial prostheses. Their implantations as thoraco-abdominal by-passes in dogs were scheduled to remain in place for periods ranging from four hours to six months. The haematologic parameters of the hosts were evaluated before implanting the prostheses and before harvesting the graft. To quantify the thrombogenicity of the flow surface, the platelet deposition and fibrinogen uptake were measured by means of an isotopic technique involving Indium-III labelled platelets and iodine-125 labelled fibrinogen. Synthesis of prostacyclin (PGI2) and thromboxane (TXA2) by the internal capsulae was determined by radioimmunoassay. The pathology was assessed according to previously described protocols. The haematologic parameters of the hosts did not alter substantially in the presence of the prostheses. The observed changes, though slight, were not pathologically significant 48 hours after implantation. Platelet and fibrin deposition was highest on the luminal surface of the prostheses 24 hours post-implantation. These decreased and became negligible after one month when the flow surface became smooth and glistening. The PGI2/TXA2 ratio of the luminal surface was much lower than that observed in native aortic tissue. These results show that the large calibre Dacron vascular prosthesis has a flow surface with a low degree of blood compatibility. The concentration of PGI2 on the newly-formed luminal surface remained low. This characteristic and the incomplete endothelialization of the luminal surface might be among the main factors that determine the high failure rate of synthetic prostheses when used as small diameter blood conduits. Topics: 6-Ketoprostaglandin F1 alpha; Animals; Blood Platelets; Blood Vessel Prosthesis; Dogs; Epoprostenol; Female; Fibrin; Fibrinogen; Hematologic Tests; Microscopy, Electron, Scanning; Polyethylene Terephthalates; Thrombosis; Thromboxane A2; Time Factors | 1988 |
Thrombosis associated with antiphospholipid antibodies cannot be explained by effects on endothelial and platelet prostanoid synthesis.
The effect of 23 antiphospholipid antibody positive SLE sera, 4 antiphospholipid antibody negative SLE sera and 17 control sera on endothelial prostacyclin and platelet thromboxane A2 production was studied. Endothelial cells and platelets were stimulated with different agonists. Depending on the stimulus used, 4-19% of the SLE sera inhibited the prostacyclin release, whereas 4-28% enhanced prostacyclin production. Our data suggest that the pathophysiological mechanisms underlying decreased prostacyclin production are heterogeneous. Follow-up of two patients showed that prostacyclin inhibitory activity was variable in time. Platelet thromboxane production was normal or increased, but never decreased in the presence of the SLE sera. An imbalance in thromboxane A2/prostacyclin ratio was present in some patients, but did not correlate with a history of thrombosis. We conclude that, in general, interference of antiphospholipid antibodies with endothelial or platelet prostanoid synthesis does not explain the occurrence of thromboembolic manifestations in antiphospholipid antibody positive SLE patients. Topics: Adult; Autoantibodies; Blood Coagulation Factors; Blood Platelets; Endothelium; Epoprostenol; Fatty Acids; Female; Humans; Lupus Coagulation Inhibitor; Lupus Erythematosus, Systemic; Male; Middle Aged; Prostanoic Acids; Thrombosis; Thromboxane A2 | 1988 |
Exchange of cyclooxygenase dependent metabolites between vessel wall and platelets in arterial thrombosis.
In this investigation, transport of endoperoxides from the vessel wall to the platelets in an in vivo model for the induction and quantification of platelet thrombosis is described. Thrombi are induced in a branch of the mesenteric artery of the male white Wistar rat through topical superfusion of adenosine diphosphate solutions following focal de-endothelialization by means of a small electrical current in two groups, comparing untreated rats to acetylsalicylic acid (ASA) treated ones. In the latter group the cyclooxygenase activity of the platelets is completely inhibited but not so that of the endothelial cells. In both groups arachidonic acid enhances adenosine diphosphate induced arterial platelet thrombosis most likely through the conveyance of endoperoxides generated in the vessel wall to the platelets as thromboxane synthetase antagonists as well as thromboxane receptor blocking agents completely inhibit this enhancement in both groups. Furthermore it is demonstrated that ASA treated animals show an increased propensity to thrombosis induced by adenosine diphosphate. Topics: Adenosine Diphosphate; Animals; Arachidonic Acid; Arachidonic Acids; Aspirin; Blood Platelets; Cyclooxygenase Inhibitors; Epoprostenol; Imidazoles; Male; Mesenteric Arteries; Platelet Aggregation; Propionates; Prostaglandin-Endoperoxide Synthases; Rats; Rats, Inbred Strains; Thrombosis; Thromboxane A2; Tranylcypromine | 1988 |
Effects of dietary palm oil on arterial thrombosis, platelet responses and platelet membrane fluidity in rats.
Wistar rats were fed a control diet containing 5 energy % (en %) sunflowerseed oil or diets containing 50 en % of either palm oil, rich in saturated fatty acids, or sunflowerseed oil, high in linoleic acid, for at least eight weeks. Arterial thrombosis tendency, measured by the aorta loop technique, tended to be lowered by the palm oil diet and was lowered significantly by the sunflowerseed oil diet, compared with the control. Aggregation of platelets in whole blood activated with collagen was not altered by palm oil feeding, but was enhanced in the sunflowerseed oil group, compared with the control. The concomitant formation of thromboxane A2 was decreased by palm oil feeding, although formation of prostacyclin did not change; the ratio of thromboxane/prostacyclin formed was decreased significantly in the palm oil group. Compared with the control diet, platelet membrane fluidity, measured by fluorescence polarization, was not altered in the palm oil group and was significantly increased only by sunflowerseed-oil feeding. Thus, although palm oil contains about 50% saturated fatty acids, it did not increase arterial thrombosis tendency and tended to decrease platelet aggregation, as compared with highly polyunsaturated sunflowerseed oil. Topics: Animals; Blood Platelets; Dietary Fats, Unsaturated; Male; Membrane Fluidity; Palm Oil; Plant Oils; Rats; Rats, Inbred Strains; Sunflower Oil; Thrombosis; Thromboxane A2 | 1988 |
Preventive effect of ONO-3708 on thrombosis and vasospasms in vitro and in vivo.
Topics: Animals; Dogs; Humans; In Vitro Techniques; Ischemic Attack, Transient; Platelet Aggregation; Thrombosis; Thromboxane A2; Vasoconstriction | 1987 |
Effect of thromboxane receptor antagonists on renal artery thrombosis in the cynomolgus monkey.
The effect of the thromboxane (Tx) A2-receptor antagonists SQ 28,668 and SQ 30,741 on platelet function and renal artery thrombosis was studied in the dialurethane-anesthetized cynomolgus monkey. Both antagonists competitively inhibited the aggregation of platelet rich plasma in vitro to arachidonic acid and U-46,619, a Tx-mimetic. SQ 30,741 was 4 to 7 times more potent than SQ 28,668 against either of these agonists. Thrombotic cyclical blood flow reductions (CFRs) were elicited by placing a critical stenosis at a crush injury site on the left renal artery. After allowing 10 consecutive CFRs, of which 95% required shaking of the vessel to restore flow, a single i.v. injection of either SQ 28,668 (1 mg/kg, n = 6), SQ 30,741 (1 mg/kg, n = 8) or vehicle (2 ml of 0.2% Na2CO3 + 10% ethanol, n = 4) was administered. Antithrombotic activity was defined as the spontaneous restoration of flow and was accompanied by a reduction in the rate of flow decline during the CFRs. Spontaneous flow restoration was observed in animals treated with SQ 28,668 (five of six) and SQ 30,741 (six of eight) but not vehicle (zero of four). The rate of flow decline was reduced only with SQ 28,668 (56 +/- 8%) and SQ 30,741 (53 +/- 10%) treatments. The antithrombotic activities of SQ 28,668 and SQ 30,741 lasted 68 +/- 6 and 224 +/- 21 min, respectively. The threshold antithrombotic dose was found to be lower for SQ 30,741 (0.20 +/- 0.03 mg/kg) than SQ 28,668 (0.61 +/- 0.09 mg/kg) in additional experiments.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Dose-Response Relationship, Drug; Ketanserin; Macaca fascicularis; Platelet Aggregation; Receptors, Prostaglandin; Receptors, Thromboxane; Renal Artery Obstruction; Renal Circulation; Thrombosis; Thromboxane A2 | 1987 |
Anti-thrombotic and anti-atherogenic action of eicosapentaenoic acid.
Effects of dietary supplementation with highly purified EPA (1.8-2.7 g/day) for 16 weeks on platelet and red blood cell function and serum lipids concentration were investigated in patients with various thrombotic diseases. Decreases in platelet aggregation, thromboxane formation in platelets, platelet retention and whole blood viscosity, increased red blood cell deformation and prolongation of bleeding time were observed in the present study. In addition a reduction in serum cholesterol and triglyceride concentrations was noted in patients with hyperlipidemia after EPA ingestion. Some clinical improvements such as improvement of diabetic gangrene or peripheral vascular occlusive disease were observed. These results indicate that dietary supplementation of purified EPA may be beneficial for prevention and treatment of cerebro- and cardiovascular diseases. Topics: Arteriosclerosis; Bleeding Time; Blood Viscosity; Capsules; Drug Evaluation; Eicosapentaenoic Acid; Erythrocyte Deformability; Female; Humans; Lipids; Male; Middle Aged; Platelet Aggregation; Thrombosis; Thromboxane A2 | 1987 |
The effect of dietary saturated fat and cholesterol on platelet function, platelet survival and response to continuous aortic injury in rats.
Induction of hypercholesterolemia in rats by diets containing milk fat, cholesterol and taurocholate caused increased sensitivity of platelets to thrombin-induced aggregation and release, but not to ADP- or collagen-induced aggregation or release. This hypersensitivity to thrombin persisted in the presence of CP/CPK to convert released ADP to ATP, and aspirin to block formation of thromboxane A2. The increased sensitivity of platelets to thrombin in hypercholesterolemic animals was associated with an increase in 18:1 omega 9, 18:2 omega 6 and 20:3 omega 6 and a decrease in 20:4 omega 6 and 22:4 omega 6 in their phospholipids. Hypercholesterolemic animals also had a shortened platelet survival that did not appear to be due to an alteration in the lipid composition of the platelets. The diet-induced changes in platelet function were not associated with enhanced thrombosis in animals with indwelling aortic catheters, but were associated with increased platelet accumulation on the exposed subendothelium. Topics: Animals; Aorta; Blood Platelets; Cell Survival; Cholesterol; Cholesterol, Dietary; Collagen; Dietary Fats; Hypercholesterolemia; Male; Milk; Platelet Aggregation; Rats; Rats, Inbred Strains; Thrombin; Thrombosis; Thromboxane A2 | 1987 |
Synergism between a phosphodiesterase inhibitor and modulators of thromboxane formation on thrombin-induced sudden death in rabbits.
Topics: Animals; Blood Pressure; Bridged Bicyclo Compounds, Heterocyclic; Death, Sudden; Drug Synergism; Fatty Acids, Unsaturated; Hydrazines; Imidazoles; Indomethacin; Isoquinolines; Male; Pyridines; Rabbits; Tetrahydroisoquinolines; Thrombin; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1987 |
Leukocyte mediated vein injury and thrombosis is reduced by a lipoxygenase inhibitor.
We have previously demonstrated an in vivo model of deep vein thrombosis which suggests that the neutrophil promotes vascular injury and thrombosis following blood flow stasis. Since leukotrienes are potent mediators of vascular injury and neutrophil (PMN) chemotaxis, we wished to determine if in vivo inhibition of 5-lipoxygenase would reduce neutrophil mediated events in our model. Lipoxygenase was inhibited in vivo with 2,3-diethyl-4-methoxy,1-naphthalenol acetate (U-66,855). The in vivo activity of U-66,855 was demonstrated in 4 cats. Each animal was treated with 5 mg/kg of U-66,855 intravenously. Blood cell leukotriene B4 (LTB4) and thromboxane A2, via its metabolite thromboxane B2 (TBX2) was assessed before and 30, 60, and 120 min after dosing. Blood cell LTB4 and TBX2 production was stimulated by A23187 (24 microM) and assayed by radioimmunoassay. We exposed and isolated a 3-cm segment of the jugular veins from 10 additional cats 5 of which were treated with U-66,855 (5 mg/kg, iv). In order to assess the effect of stasis, the jugular veins were ligated at the thoracic inlet for 2 hr after which the veins were perfused, fixed in 2.5% glutaraldehyde, and prepared for electron microscopy. U-66,855 reduced LTB4 production significantly (P less than 0.01), but not TBX2. In untreated cats, PMNs adhered to and migrated underneath the venous endothelium. Additionally, platelets, fibrin and formed thrombi were found on the basement membrane exposed by the migrating neutrophils. In contrast, we observed significantly reduced PMN adhesion as well as no fibrin deposition in veins obtained from cats treated with U-66,855. The results suggest that 5-lipoxygenase inhibition can significantly reduce undesirable neutrophil/vessel wall interactions. Topics: Animals; Arachidonate Lipoxygenases; Cats; Cell Adhesion; Cell Movement; Endothelium; Jugular Veins; Leukocytes; Leukotriene B4; Lipoxygenase Inhibitors; Microscopy, Electron; Microscopy, Electron, Scanning; Naphthols; Neutrophils; Thrombosis; Thromboxane A2; Thromboxane B2 | 1986 |
Efficacy of tissue plasminogen activator and urokinase in a canine model of prosthetic graft thrombosis.
Tissue plasminogen activator and urokinase were evaluated in a model of prosthetic graft thrombosis. In addition, the effects of thrombus age on lysability and the effect of thrombolytic agents on endothelium were examined. Polytef (polytetrafluoroethylene [PTFE]) grafts (3 mm X 3.5 cm) were placed in femoral arteries of dogs and graft thrombosis was induced. Grafts were treated with a local infusion of either urokinase or tissue plasminogen activator (4000 units/min) and the times for initial flow, complete thrombolysis, and anastomotic bleeding were noted. The luminal surfaces of the grafts and the proximal arterial segments were assayed for the production of thromboxane A2 and prostacyclin and examined with scanning electron microscopy. No difference in the ease of graft lysis was observed, but 50% of tissue plasminogen activator-treated vs 0% of urokinase treated grafts had extravasation of blood through the wall. Grafts treated with tissue plasminogen activator produced less thromboxane A2 and had less thrombus than those treated with urokinase. No differences between arteries exposed to either agent and control arteries were seen. Grafts treated 1,3,5, and 7 days after thrombosis were progressively more difficult to lyse. We conclude that tissue plasminogen activator is an effective thrombolytic agent, but has a potential for local bleeding complications. Grafts of PTFE are thrombogenic after lysis, but may be less so with tissue plasminogen activator than with urokinase. No effect on arterial endothelium was seen, and our studies confirm the clinical impression that older thrombi are more difficult to lyse. Topics: Animals; Arteries; Blood Vessel Prosthesis; Disease Models, Animal; Dogs; Epoprostenol; Graft Occlusion, Vascular; Hemorrhage; Microscopy, Electron, Scanning; Polytetrafluoroethylene; Thrombosis; Thromboxane A2; Time Factors; Tissue Plasminogen Activator; Urokinase-Type Plasminogen Activator | 1986 |
The effect of predetermined thrombotic potential of the recipient on small-caliber graft performance.
This study was designed to establish the influence of the recipient's thrombotic potential on the patency of small-caliber prostheses and to evaluate the subsequent improvement of graft performance by medicinally altering the prostaglandin balance in subjects predisposed to graft occlusion. Mongrel dogs were pretested and classified as low and high responders according to their thrombotic potential, measured as prostaglandin metabolite balance and platelet aggregability. High responders were randomly divided into two groups. Those assigned to serve as the medicated subjects were pretreated 1 week before surgery with a single oral daily administration of combined dazmegrel (UK-38,485) and aspirin in equal dosages of 3 mg/kg. Medication was continued throughout the experiment. Dacron grafts were implanted bilaterally in the carotid artery site in all subjects. Following a 3-week implantation period, the patency rate for the group with low thrombotic potential was 100%. In the animals with high thrombotic potential the patency rate was 10% for nonmedicated subjects and 100% for medicated subjects. These data support the concept that the thrombotic potential largely determines the capacity of the recipient's blood to thrombose small-caliber prostheses. Effective medicinal alteration of prostaglandin balance results in exceptionally increased patency of synthetic grafts. Topics: Animals; Aspirin; Blood Cell Count; Blood Vessel Prosthesis; Carotid Arteries; Dogs; Drug Therapy, Combination; Epoprostenol; Graft Occlusion, Vascular; Imidazoles; Male; Malondialdehyde; Platelet Aggregation; Prostaglandins; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1986 |
Oral acetylsalicylic acid improves patency rates in small-vessel anastomoses.
Because platelet aggregation is the critical step in vascular thrombosis, this study investigated the possibility that acetylsalicylic acid given orally would improve patency of anastomoses in small arteries. Under clean conditions, in male Sprague-Dawley rats, the superficial femoral arteries were divided and reanastomosed using the sleeve technique with 10-0 nylon suture. Fifty-one rats received a full laboratory diet and acetylsalicylic acid in their drinking water (1.08 mg/ml), before and after operation. A control group of 54 rats was matched for weight, water intake and duration of vessel occlusion. Vessel patency was assessed on postoperative day 7, at sacrifice, by dye angiography. In the experimental group 7 (13.7%) anastomoses became occluded, compared with 20 (37.0%) in the control group (chi 2 = 5.99, df = 1, p less than 0.025). Serum thromboxane B2 levels in five rats given acetylsalicylic acid orally (33.1 +/- 6.1 ng/ml) were significantly (p less than 0.001) lower than in five control rats (86.6 +/- 49.1 ng/ml). The authors concluded that acetylsalicylic acid administered in the drinking water to Sprague-Dawley rats improved the patency rate of femoral artery anastomoses probably because of a reduction in platelet aggregation. Orally administered acetylsalicylic acid may be of clinical benefit to patients who undergo small-vessel anastomoses. Topics: Administration, Oral; Animals; Aspirin; Femoral Artery; Male; Platelet Aggregation; Postoperative Complications; Rats; Rats, Inbred Strains; Suture Techniques; Thrombosis; Thromboxane A2; Vascular Patency | 1986 |
Platelet active concentration profiles near growing thrombi. A mathematical consideration.
When blood contacts foreign material surfaces, platelets usually adhere and form aggregates on those surfaces, generating mural thrombi. The mechanism of mural thrombogenesis is not completely understood, but one hypothesis states that the local release of certain platelet-active substances from the platelets composing an initial small thrombus stimulates additional platelet recruitment to that thrombus, resulting in growth of the cell aggregate. The purpose of this paper is to investigate the feasibility of this hypothesis. Concentration profiles of adenosine diphosphate (ADP), thromboxane A2 (TxA2), and thrombin were computed in the vicinity of growing model thrombi 10 and 20 micron long. Wall shear rates of 100, 500, and 1,500 s-1 were considered for blood flowing through a thin rectangular slit 200 micron wide coated with collagen, a predominant subendothelial protein. The local concentrations of ADP and TxA2 were marginally large enough to stimulate platelet activation individually, while local thrombin levels can be much greater than required for stimulation. Antithrombin III, a natural thrombin inhibitor, did not significantly reduce the thrombin concentrations, but antithrombin III accelerated by heparin greatly reduced the local thrombin concentrations. The reduced thrombin levels may, however, still be large enough to activate platelets. Topics: Adenosine Diphosphate; Blood Platelets; Humans; Mathematics; Models, Biological; Platelet Aggregation; Thrombin; Thrombosis; Thromboxane A2 | 1986 |
Sex-related differences in the effects of aspirin on the interaction of platelets with subendothelium.
Using the Baumgartner perfusion technique, marked sex-related differences in the extent of platelet-subendothelium interaction and in the effect of aspirin (ASA) have been observed. The administration of ASA (150 mg daily for 15 days) to two groups of healthy volunteers, one composed of males and the other of females, proved to block the generation of TXB2 in both cases. The basic pattern of platelet subendothelium interaction, however, was found to be markedly different in both groups studied. In men, aspirin treatment induced a significant reduction in the percentage of platelet thrombi, whereas in women, post ASA values remained at the same level as in control experiments. These results show that in the Baumgartner perfusion system women display a less thrombogenic tendency than men and that 150 mg of ASA administered daily are effective in reducing the extent of platelet-subendothelium interaction in the male group but not in the female group. These findings could explain the absence of benefit observed for women in clinical trials with aspirin. Topics: Administration, Oral; Adult; Aspirin; Blood Platelets; Blood Vessels; Endothelium; Female; Humans; Male; Platelet Adhesiveness; Platelet Aggregation; Sex Factors; Thrombosis; Thromboxane A2 | 1986 |
Altered metabolism of thromboxane A2 and prostacyclin in the pathogenesis of coronary vasospasm and thrombosis.
This chapter documents the significance of TXA2 and PGI2 in the pathologic events associated with active coronary artery disease. We suggest that altered metabolism of these prostanoids may result in disintegration of platelet and vascular wall reactivities accompanying coronary spasm or intravascular thrombosis associated with myocardial cell necrosis. Further studies should be carried out to determine if coronary circulatory disorders and subsequent myocardial cell injury can be affected by disintegration of the lipoxygenase system that plays a pivotal role in maintaining homeostasis in vascular endothelium and blood components. Topics: Coronary Vasospasm; Epoprostenol; Humans; Myocardial Infarction; Thrombosis; Thromboxane A2 | 1985 |
Potentiation of the inhibitory effect of a thromboxane A2 antagonist (L-640,035) on arterial thrombosis formation in rabbit by the angiotensin converting enzyme inhibitor enalapril.
The angiotensin converting enzyme inhibitor enalapril (0.5 mg/kg i.v.) potentiated significantly the inhibitory effect of the thromboxane A2 antagonist L-640,035 (1 mg/kg i.v.) on electrically induced platelet accumulation in the rabbit in vivo. Enalapril had no effect upon platelet accumulation when given alone. The hypotensive effects of enalapril did not account for the potentiation because a combination of hexamethonium (5 mg/kg i.v.) and hydralazine (1 mg/kg i.v.), which decreased blood pressure similarly to enalapril, did not augment the effect of L-640,035. Determination by radioimmunoassay of plasma levels of immunoreactive 6-keto-PGF1 alpha, suggested that increases in PGI2 levels after combined administration of enalapril and L-640,035 could explain the observed potentiation. Topics: 6-Ketoprostaglandin F1 alpha; Angiotensin-Converting Enzyme Inhibitors; Animals; Dibenzothiepins; Dipeptides; Drug Synergism; Enalapril; Female; Hexamethonium Compounds; Hydralazine; Male; Prostaglandin Antagonists; Rabbits; Radioimmunoassay; Thrombosis; Thromboxane A2; Thromboxanes | 1985 |
Blockade of thromboxane and the prevention of eicosanoid-induced sudden death in mice.
We studied the effects of thromboxane-receptor antagonism and thromboxane synthetase inhibition in a thrombotic model of sudden death in mice. Intravenous injection of arachidonic acid (AA; 80 mg/kg) or the prostaglandin-endoperoxide analog U-46,619 (2.3 mg/kg) results in sudden death in approximately 90% of the animals. Pretreatment with the thromboxane receptor antagonist SQ-29,548 (0.3-10 mg/kg) protects dose-dependently against AA and U-46,619-induced sudden death. In contrast, CGS-13,080, a thromboxane synthetase inhibitor, shows a dose-dependent beneficial effect in AA-induced sudden death only. Although PTA2 has partial thromboxane agonistic properties in the rabbit, it protected the mice against AA-induced sudden death, thus demonstrating TxA2 antagonistic properties in this species. These data emphasize the importance of thromboxane A2 as a major mediator of arachidonic acid-induced sudden death and the effectiveness of thromboxane-receptor antagonists in endoperoxide-induced sudden death. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Animals; Arachidonic Acid; Arachidonic Acids; Bicyclic Monoterpenes; Bridged Bicyclo Compounds, Heterocyclic; Death, Sudden; Fatty Acids, Unsaturated; Fibrinolytic Agents; Hydrazines; Imidazoles; Injections, Intravenous; Male; Mice; Platelet Aggregation; Prostaglandin Endoperoxides, Synthetic; Pyridines; Receptors, Cell Surface; Receptors, Prostaglandin; Receptors, Thromboxane; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1985 |
Supplementation of the diet with eicosapentaenoic acid: a possible approach to the treatment of thrombosis and inflammation.
Topics: Dietary Fats; Eicosapentaenoic Acid; Epoprostenol; Humans; Inflammation; Leukocytes; Leukotriene B4; Myocardial Infarction; Thrombosis; Thromboxane A2 | 1985 |
Platelet function and thrombus in diabetes.
Patients with diabetes mellitus are several fold more prone to various forms of vascular diseases than are the non-diabetic subjects. Because platelets are in the key position in thrombus formation and possibly in atherogenesis, much interest has focused on the role of platelets in the development of diabetic vascular disease. Most studies on this topic have suggested increased adhesiveness and aggregability of the platelets from diabetic patients. The increased production of von Willebrand factor may account for the enhanced adhesion. The shift of the balance between proaggregatory thromboxane A2 and antiaggregatory prostacyclin to the dominance of thromboxane A2 could explain the increased aggregability of diabetic platelets, but the data available at the moment do not allow the conclusion that such a change really exist in human in vivo. One recent work has suggested that the increased glycosylation of connective tissue proteins in diabetes would increase their aggregating potency, but also this finding needs further confirmation. Topics: Blood Platelets; Diabetes Complications; Diabetes Mellitus; Epoprostenol; Humans; Platelet Adhesiveness; Platelet Aggregation; Thrombosis; Thromboxane A2 | 1985 |
Impaired arteriolar vasodilation induced by thrombosis of a coronary arterial stenosis.
This study tested the hypothesis that aggregation of platelets and release of thromboxane A2 at the site of a coronary arterial stenosis may contribute to myocardial ischemia by impairing flow regulation of the distal coronary bed. Measurements of hemodynamics, flow (microspheres), lactate metabolism, and prostaglandin (PG) metabolites (thromboxane B2 and 6-keto-PGF1 alpha) were made in closed-chest anesthetized pigs instrumented with a stenosis (65% diam reduction) in the left anterior descending (LAD) coronary artery. Data were acquired prior to stenosis insertion (control) and 5 and 15 min after insertion, during which time thrombotic occlusion of the device was occurring. Heart rate was controlled by atrial pacing. Distal LAD zone endocardial flow (ml X min-1 X g-1) declined versus control (1.15 +/- 0.20, mean +/- SD) at 5 min (0.89 +/- 0.40, P less than 0.05) and 15 min (0.41 +/- 0.36, P less than 0.01) of occlusion. Distal endocardial resistance (mmHg X ml-1 X min X g), however, did not change versus control (72 +/- 12) at 5 (66 +/- 12) or 15 min (61 +/- 38). Distal epicardial resistance (mmHg X ml-1 X min X g) declined versus control (90 +/- 17) at 5 (66 +/- 35, P less than 0.05) and 15 min (43 +/- 26, P less than 0.01) postinsertion. Finally, lactate extraction (%) at control (42 +/- 19) changed to production 15 min postinsertion (-36 +/- 93, P less than 0.05) and arterial-anterior interventricular vein thromboxane B2 difference (pg/0.1 ml) changed from 13.1 +/- 17.8 pre to -15.8 +/- 30.0 at 5 min post (P less than 0.05). Thus platelet aggregation and release at a spontaneously thrombosing stenosis contribute to ischemia not only by reduction of stenosis diameter but also by impairment of flow regulation in endocardial layers distal to it. Topics: Animals; Arterial Occlusive Diseases; Aspirin; Coronary Circulation; Heart Rate; Hemodynamics; Imidazoles; Lactates; Lactic Acid; Prostaglandin Endoperoxides, Synthetic; Regional Blood Flow; Swine; Thrombosis; Thromboxane A2; Vascular Resistance; Vasodilation | 1985 |
Anti-thrombotic effect of curcumin.
Topics: Animals; Aspirin; Blood Platelets; Catechols; Collagen; Curcumin; Epinephrine; Macaca mulatta; Male; Malondialdehyde; Mice; Thrombosis; Thromboxane A2 | 1985 |
Prostaglandin production and platelet reactivity of small-diameter grafts.
This article examines the relationship of platelet deposition to thromboxane and prostacyclin (PGI2) production in arterial autografts (n = 8), para-anastomotic native artery (n = 40), nonseeded control (n = 6), and endothelial cell-seeded (n = 17) small-diameter Dacron grafts implanted in the carotid and femoral arteries of dogs. Platelet deposition was measured by a dual-isotope subtraction platelet-imaging technique that expresses platelet deposition as percent indium excess (%IE). PGI2 and thromboxane assays were performed with the use of an immunoreactive assay. The %IE in the nonseeded grafts was significantly higher than in the seeded prostheses (p less than 0.001). Arterial autografts accumulated significantly less platelets than did seeded grafts (p less than 0.05) or nonseeded grafts (p less than 0.001). The thromboxane A2 (TXA2) production in nonseeded grafts was significantly higher than in seeded grafts (p less than 0.001), arterial autografts (p less than 0.001), or in para-anastomotic native artery (p less than 0.001). The PGI2 production by the arterial autografts was significantly higher than by the nonseeded grafts (p less than 0.005), seeded grafts (p less than 0.001), or para-anastomotic native artery (p less than 0.025). The PGF1 alpha/TXB2 ratio was significantly higher in the arterial autografts when compared with the nonseeded grafts (p less than 0.001), endothelial cell-seeded grafts (p less than 0.001), or para-anastomotic native artery (p less than 0.025). We conclude that platelet deposition can be significantly decreased by endothelial cell seeding of small-diameter grafts. The transmural production of TXA2 by native arteries and prosthetic grafts may have an important influence on platelet deposition and patency.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Arteries; Blood Platelets; Blood Vessel Prosthesis; Cells, Cultured; Dogs; Endothelium; Epoprostenol; Indium; Polyethylene Terephthalates; Prostaglandins; Radioisotopes; Thrombosis; Thromboxane A2; Thromboxane B2 | 1984 |
Effects of AVS (1,2-bis(nicotinamido)propane) on platelet function and vascular endothelium.
The effects of 1,2-bis(nicotinamido)propane (AVS) on platelet function and vascular endothelium were investigated using various experimental thrombosis and vascular endothelial injury models. Neither in vitro platelet aggregation induced by ADP, collagen or arachidonate nor ex vivo platelet aggregation by ADP or collagen could be antagonized by AVS. On the other hand, AVS prevented mice, rats and rabbits from death induced by acute cerebral or pulmonary thromboembolism following the injection of arachidonate or collagen. These activities were as potent as those of acetylsalicylic acid. The disrupting actions of citrate and/or lipidperoxide (13-hydroperoxy linoleic acid) on endothelium were well inhibited by the pretreatment of AVS. AVS did not inhibit cyclooxygenase, increased prostacyclin (PGI2)/thromboxane A2 (TXA2) ratio in the coupled system of platelets and aortic microsomes. In conclusion, AVS inhibited thrombus formation in vivo while it was ineffective in vitro platelet alone system, which may result from the actions of this agent on both platelets and vascular endothelium. The above-mentioned results clearly show that AVS may be a new potent anti-vascular damaging agent with both endothelium stabilizing and PGI2 enhancing activities. Topics: Animals; Arachidonic Acid; Arachidonic Acids; Aspirin; Blood Platelets; Blood Vessels; Cerebrovascular Disorders; Collagen; Dinoprostone; Endothelium; In Vitro Techniques; Mice; Microscopy, Electron, Scanning; Niacinamide; Platelet Aggregation; Prostaglandins E; Rabbits; Rats; Rats, Inbred Strains; Thromboembolism; Thrombosis; Thromboxane A2 | 1984 |
The aspirin dilemma revisited.
Topics: Aspirin; Epoprostenol; Humans; Thrombosis; Thromboxane A2 | 1984 |
The pharmacological profile of the thromboxane A2 antagonist BM 13.177. A new anti-platelet and anti-thrombotic drug.
BM 13.177 (4-[2-(benzenesulfonamido)-ethyl]-phenoxyacetic acid) is a representative of a new class of sulfonamidophenylcarboxylic acids which possess platelet-inhibitory and anti-thrombotic activity and inhibits the contraction of rabbit aorta stimulated by PG endoperoxides and TXA2. BM 13.177 5 mg/kg body weight p.o. protected rabbits from arachidonate-induced sudden death and greater than or equal to 10 mg/kg dose-dependently reduced the experimental thrombus formation induced in the rabbit aorta by perivascular administration of silver nitrate. In guinea-pigs, the collagen-induced bronchoconstriction was inhibited in a dose- and time-dependent fashion. The formation of TXA2 and the TXA2-induced platelet aggregation and smooth muscle contraction are probably crucial events in these experimental models. The protective effect of BM 13.177 may, therefore, be due to the TXA2-antagonizing effect of BM 13.177, which has been conclusively demonstrated in human platelets (PATSCHEKE and STEGMEIER, Thrombosis Res., 33, 277-288 (1984). The antagonism of TXA2 is supported by the observation that BM 13.177 also specifically inhibits the contraction of isolated arterial strips from rabbits which were stimulated with the thromboxane A2 mimetic U 46619. Schild-plot with a slope close to unity suggests a competitive type of antagonism. BM 13.177 exhibited neither anti-inflammatory nor ulcer-inducing activity of cyclooxygenase inhibitors. Furthermore it did not block the TXB2 formation in spontaneously clotting blood from rabbits and did not inhibit the release of prostacyclin-like activity from rabbit aortas. The lack of toxicological effects in long-term toxicity studies in rat and dog, together with the absence of objective and subjective side effects in the first human studies have encouraged us to initiate clinical trials in order to evaluate the therapeutic benefit of this new approach in humans. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Animals; Epoprostenol; Female; Fibrinolytic Agents; Guinea Pigs; Male; Muscle Contraction; Muscle, Smooth, Vascular; Platelet Aggregation; Prostaglandin Endoperoxides, Synthetic; Rabbits; Sulfonamides; Thrombosis; Thromboxane A2; Thromboxane B2; Thromboxanes | 1984 |
Inhibition of prostanoid-mediated platelet aggregation in vivo and in vitro by 3-hydroxymethyl-dibenzo(b,f)thiepin 5,5-dioxide (L-640,035).
Platelet aggregation was studied in vitro with human platelets and in vivo in guinea pigs and rabbits. L-640,035 and its acetic acid metabolite, L-636,499, significantly inhibited human platelet aggregation induced by arachidonic acid, collagen and a prostaglandin endoperoxide analog (U44069) but not ADP. In guinea pigs, circulating platelets labeled with 111indium were monitored with probes in the lung and abdominal regions. Platelet aggregation was indicated by an increase in the ratio of gamma-radiation of the lung vs. the abdominal region, as aggregates of platelets accumulate in the microvascular network in the lung. L-640,035 (1 and 3 mg/kg i.v.) inhibited platelet aggregation induced in this model by U-44069 but not by ADP. In rabbits acute thrombosis was induced in the carotid artery by local electrical stimulation and platelet accumulation at the stimulus site was quantitated using 111indium-labeled autologous platelets. L-640,035 (1 and 3 mg/kg i.v.) significantly reduced electrically evoked platelet accumulation. It is concluded that L-640,035 is a novel and selective antagonist of platelet aggregation induced by thromboxane A2 or prostaglandin endoperoxides and that it may have utility as an antiplatelet drug. Topics: Adenosine Diphosphate; Animals; Arachidonic Acid; Arachidonic Acids; Collagen; Dibenzothiepins; Electric Stimulation; Female; Guinea Pigs; Humans; Male; Platelet Aggregation; Prostaglandin Endoperoxides, Synthetic; Rabbits; Thrombosis; Thromboxane A2; Thromboxane B2; Thromboxanes | 1984 |
Antagonism of thromboxane A2/prostaglandin H2 by 13-azaprostanoic acid prevents platelet deposition to the de-endothelialized rabbit aorta in vivo.
The present study evaluated the direct involvement of thromboxane A2/prostaglandin H2 (TXA2/PGH2) in the process of thrombus formation at a site of vascular damage. De-endothelialization of the rabbit aorta was performed by a balloon catheter technique. Platelet deposition to the injured vessel was measured using 111Indium-labeled autologous platelets. Studies using the radiolabeled TXA2/PGH2 antagonist, 13-azaprostanoic acid (13-APA), indicated that 13-APA has an in vivo half-life of approximately 35 min and is excreted by the kidney in the metabolized form. Addition of 13-APA to rabbit plasma samples in vitro produced a dose-dependent inhibition of arachidonic acid-induced platelet aggregation. When comparable plasma levels of 13-APA were achieved by infusion of 13-APA (300 micrograms/kg/min for 90 min), a similar dose-dependency of inhibition of ex vivo aggregation was observed. Furthermore, at a plasma concentration of 40 microM, 13-APA was found to inhibit platelet deposition to the de-endothelialized rabbit aorta by 45%. Because 13-APA does not interfere with arachidonic acid metabolism, the ability of 13-APA to suppress thrombus formation is presumably due to direct antagonism of TXA2/PGH2 at the platelet receptor level. These findings, therefore, provide evidence that TXA2 and/or PGH2 have a major role in platelet deposition at a site of vascular damage. Topics: Animals; Arachidonic Acid; Arachidonic Acids; Blood Platelets; Fatty Acids; Male; Platelet Aggregation; Prostaglandin Endoperoxides; Prostaglandin Endoperoxides, Synthetic; Prostaglandin H2; Prostaglandins H; Prostanoic Acids; Rabbits; Thrombosis; Thromboxane A2; Thromboxanes | 1984 |
Eicosanoids and the blood vessel wall.
Topics: Arachidonic Acid; Arachidonic Acids; Blood Platelets; Cardiovascular Diseases; Coronary Disease; Eicosanoic Acids; Endothelium; Heart Failure; Humans; Leukocytes; Lipoxygenase; Muscle, Smooth, Vascular; Myocardial Infarction; Myocarditis; Prostaglandin-Endoperoxide Synthases; Prostaglandins; Thrombosis; Thromboxane A2 | 1984 |
Prostaglandin endoperoxides and thromboxane A2 in thrombus formation in the hamster cheek pouch in vivo.
Electrical stimulation in the presence of ADP of arterioles of the hamster cheek pouch caused endothelial damage and white thrombus formation. The thrombus formation was inhibited by cyclo-oxygenase inhibitors aspirin and sulphinpyrazone and its thioether derivative G 25671. Thromboxane synthetase inhibitors N-(7-carboxyheptyl) imidazole and butylimidazole failed to inhibit thrombus formation, although in the same doses both compounds inhibited serum levels of thromboxane. These results indicate that thromboxane is not important in thrombus formation in the hamster, but that prostaglandin endoperoxides are more significant. However, it is possible that the inhibition of white thrombus formation by aspirin, sulphinpyrazone and G 25671 may be mediated by a different mechanism altogether. Topics: Animals; Arachidonic Acid; Arachidonic Acids; Aspirin; Cheek; Cricetinae; Cyclooxygenase Inhibitors; Dipyridamole; Epoprostenol; Male; Mesocricetus; Microscopy, Electron; Platelet Aggregation; Sulfinpyrazone; Thrombosis; Thromboxane A2; Thromboxane B2; Thromboxane-A Synthase; Thromboxanes | 1984 |
[Thrombocyte interaction with a collagen substrate: the role of thrombocyte-synthesized prostaglandin endoperoxides and thromboxane A2].
The effects of (i) the exogenous arachidonic acid (AA), (ii) stable prostaglandin endoperoxide analogue--U46619, and (iii) cyclooxygenase inhibitor--aspirin on the interaction of platelets with a surface coated with fibrillar calf skin collagen were studied using scanning electron microscopy. AA and U46619 stimulate massive spreading of platelets (on the collagen substrate and formation of surface-bound multilayer (thrombi-like) aggregates. The stimulation of spreading and formation of thrombi-like aggregates by AA correlate with the thromboxane A2 (TXA2) synthesis in platelets. Unlike AA, U46619 induces these processes without transformation into TXA2 and stimulation of its synthesis in platelets. Cyclooxygenase inhibitor--aspirin prevents the AA-induced platelet spreading, formation of the surface-bound thrombi-like aggregates, and TXA2 synthesis. In the absence of soluble platelet inducers, aspirin inhibits the substrate-induced spreading, but doesn't affect the initial attachment of nonactivated platelets to the collagen substrate. Topics: 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid; Arachidonic Acid; Arachidonic Acids; Aspirin; Blood Platelets; Collagen; Humans; Microscopy, Electron, Scanning; Platelet Aggregation; Prostaglandin Endoperoxides; Prostaglandin Endoperoxides, Synthetic; Thrombosis; Thromboxane A2; Thromboxanes | 1984 |
Arterial wall thromboxane: dominance after surgery predisposes to thrombosis.
Ibuprofen reduces early occlusion and surface thrombogenicity of small-diameter polytetrafluoroethylene (PTFE) grafts in sheep. The following experiments were designed to determine the effects of cyclooxygenase inhibition by ibuprofen on long-term patency and arterial wall and graft neointima prostaglandin synthesis. Crossover PTFE grafts (internal diameter 4 mm) were interposed between the carotid arteries of 18 sheep. Nine animals were used as controls, and nine received ibuprofen, 10 mm/kg intravenously, 1 hour prior to surgery and then every 8 hours for the duration of the experiment. Grafts were removed either when they became occluded or routinely at 60 days. Arterial segments adjacent to anastomoses were removed at graft insertion and at graft removal for assay of arterial wall production of the prostaglandins 6-keto-PGF1 alpha and thromboxane B2 (TxB2). There was no difference in patency between control animals (four out of nine) and ibuprofen-treated (five out of nine) animals. Arterial segments removed before the administration of ibuprofen produced 2.59 +/- 1.48 nM/mg protein/20 min of 6-keto-PGF1 alpha and 2.59 +/- 1.38 nM/mg protein/20 min of TxB2. A single dose of ibuprofen reduced the levels of 6-keto-PGF1 alpha to 0.30 +/- 0.12 nM/mg protein/20 min and the levels of TxB2 to 0.78 +/- 0.41 nM/mg protein/20 min (p less than 0.01). When the grafts were removed, control animals showed a reduction of 6-keto-PGF1 alpha to 1.69 +/- 0.86 nM/mg protein/20 min, whereas TxB2 levels remained unchanged at 2.54 +/- 1.23 nM/mg protein/20 min.(ABSTRACT TRUNCATED AT 250 WORDS) Topics: Animals; Blood Vessel Prosthesis; Carotid Arteries; Graft Occlusion, Vascular; Ibuprofen; Sheep; Thrombosis; Thromboxane A2; Thromboxanes | 1984 |
[Alteration of platelet population in thrombotic diseases].
Topics: Blood Platelets; Blood Volume; Humans; Myocardial Infarction; Platelet Aggregation; Platelet Count; Thrombosis; Thromboxane A2 | 1984 |
Pharmacological studies on OKY-1581: a selective thromboxane synthetase inhibitor.
Topics: Acrylates; Animals; Arachidonic Acid; Arachidonic Acids; Aspirin; Biological Assay; Blood Platelets; Cerebral Infarction; Haplorhini; Kinetics; Methacrylates; Muscle Contraction; Oxidoreductases; Platelet Aggregation; Rabbits; Rats; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1983 |
[Prostaglandins in coronary disease].
Topics: Arteriosclerosis; Blood Platelets; Coronary Disease; Dietary Fats; Epoprostenol; Humans; Myocardial Infarction; Prostaglandins; Thrombosis; Thromboxane A2 | 1983 |
Prostanoids in platelet-vascular interactions.
Prostacyclin, the labile prostanoid product of arachidonic acid metabolism in vascular endothelium, is the most potent known inhibitor of platelet aggregation and is highly effective in relaxing vascular smooth muscle. Its production is probably critically important in the maintenance of an intact vasculature. Although there is some evidence that prostacyclin circulates as a hormone, it is probably most important as a locally active agent in preventing thrombosis and maintaining patent vessels. Several factors can influence prostacyclin production, the most important of which probably act locally at sites of vessel wall injury. The most promising therapeutic approaches toward using prostacyclin's beneficial effects in vascular disease may lie in the use of drugs aimed at increasing prostacyclin production. Among these are thromboxane synthesis inhibitors, which act by diverting prostaglandin endoperoxides through the prostacyclin synthetase pathway, and lipoxygenase inhibitors, which might act chiefly by preventing formation of metabolites capable of inhibiting prostacyclin synthetase. Topics: Blood Platelets; Blood Vessels; Endothelium; Epoprostenol; Humans; Muscle Relaxation; Muscle, Smooth, Vascular; Platelet Aggregation; Prostaglandins; Thrombosis; Thromboxane A2; Thromboxane-A Synthase | 1983 |
Plaques and platelets. The vascular surgeon's challenge and dilemma.
Vascular surgeons have not paid enough attention to all of the host factors responsible for the pathogenesis of atherosclerosis. With a clearer understanding of the role of platelets and endothelial cells, improved results and better long-term palliation can be realized by combining newer pharmacologic approaches with well-established surgical procedures. Prostaglandin chemistry research may provide promising therapeutic adjuncts to current techniques for managing atherosclerosis. Topics: Animals; Arteriosclerosis; Blood Platelets; Epoprostenol; Humans; Platelet Aggregation; Risk; Thrombosis; Thromboxane A2 | 1983 |
Inhibition of platelet aggregation by a new agent, 2,2'-dithiobis-(N-2-hydroxypropyl benzamide) (KF4939).
KF4939, 2,2'-dithiobis-(N-2-hydroxypropylbenzamide), is a potent inhibitor of platelet aggregation in vitro in rabbit and human PRP. This agent inhibited both cyclooxygenase product-dependent (collagen and arachidonate) and independent (ADP and thrombin)-platelet aggregations. This action carried over to ex vivo situation following intraduodenal dosing as demonstrated in rabbits. KF4939 inhibited experimentally induced thrombocytopenias in rats and pulmonary thrombosis in mice following oral doses in a range of 25-300mg/kg. These results indicate that KF4939 is a new orally active inhibitor of platelet aggregation possessing a different mode of action from cyclooxygenase inhibition. Topics: Adenosine Diphosphate; Animals; Arachidonic Acid; Arachidonic Acids; Aspirin; Benzamides; Collagen; Dose-Response Relationship, Drug; Fibrinolytic Agents; Humans; In Vitro Techniques; Male; Mice; Papaverine; Platelet Aggregation; Rabbits; Rats; Rats, Inbred Strains; Thrombin; Thrombocytopenia; Thrombosis; Thromboxane A2 | 1983 |
Protective effects of thromboxane A2 synthetase inhibitors on endotoxin shock.
To elucidate the role of thromboxane A2 in the development of endotoxin shock following administration of endotoxin, the effects of three thromboxane A2 synthetase inhibitors, (E)-3-(4-(1-imidazolyl)phenyl)-2-propenoic acid hydrochloride monohydrate (OKY-046), sodium (E)-3-(4-(3-pyridylmethyl)phenyl)-2-methylacrylate (OKY-1581) and imidazole were examined. Intravenous administration of E. Coli endotoxin (3 mg/kg) produced shock and all rats died within ten hours. Pretreatment with thromboxane A2 synthetase inhibitors markedly improved the survival rates. The untreated endotoxin shock group showed marked increase in thromboxane B2 levels in the venous blood, while no such changes were seen in the pretreated groups. There were no statistically significant differences in 6-keto prostaglandin F1 alpha levels in the venous blood. In the untreated shock group, microthrombi were observed in 64% of the glomeruli in the kidneys two hours after endotoxin injection. In the groups pretreated with OKY-046, OKY-1581 and imidazole, microthrombi were seen only in 22, 19 and 24%, respectively. Thus, thromboxane A2 plays an important role in the development of endotoxin shock and thromboxane A2 synthetase inhibitors, in particular OKY-046 and -1581, are prophylactic. Topics: Animals; Fibrinogen; Glucuronidase; Kidney; Male; Methacrylates; Oxidoreductases; Platelet Count; Rats; Rats, Inbred Strains; Shock, Septic; Thrombosis; Thromboxane A2; Thromboxane B2; Thromboxane-A Synthase; Thromboxanes | 1983 |
[Prostaglandins, thrombosis and hemorrhage].
Topics: Animals; Arachidonic Acids; Chemical Phenomena; Chemistry; Epoprostenol; Hemorrhage; Hemostasis; Humans; Prostaglandins; Rats; Thrombosis; Thromboxane A2 | 1982 |
Enhancement of arterial thromboformation by uric acid, a free radical scavenger.
Topics: Adenosine Diphosphate; Animals; Arterial Occlusive Diseases; Epoprostenol; Free Radicals; Photometry; Prostaglandin-Endoperoxide Synthases; Rats; Rats, Inbred Strains; Thrombosis; Thromboxane A2; Tranylcypromine; Uric Acid | 1982 |
Prostacyclin in therapeutics.
Topics: Blood Platelets; Epoprostenol; Hemodynamics; Humans; Prostaglandins; Thrombosis; Thromboxane A2 | 1981 |
Platelet - vessel wall interaction: role of blood clotting.
Vascular damage initiates not only the adhesion and aggregation of blood platelets but also coagulation, which is of mixed (intrinsic and extrinsic) origin. Evidence is presented that thrombin, generated as a result of the injury, is a prerequisite for platelet aggregation. Platelets, after activation, in their turn promote coagulation. Prostaglandin I2 (PGI2 or prostacyclin) inhibits coagulation induced by damaged vascular tissue. This effect of PGI2 is mediated by the inhibition of platelets in their participation in the generation of factor Xa and thrombin. Dietary cod liver oil, by changing plasma coagulability, decreases the procoagulation activity of vessel walls, and arterial thrombosis. Another fish oil with similar effects on plasma coagulability and some other haemostatic parameters does not modify vessel wall-induced clotting, nor does it significantly lower arterial thrombosis tendency; this indicates the physiological relevance of vessel wall-induced clotting in arterial thrombus formation. Some evidence is also given for the importance of vessel wall-induced clotting in primary haemostasis. Topics: Adenosine Diphosphate; Animals; Aorta; Blood Coagulation; Blood Platelets; Cod Liver Oil; Collagen; Endothelium; Epoprostenol; Factor VII; In Vitro Techniques; Rats; Thrombin; Thromboplastin; Thrombosis; Thromboxane A2 | 1981 |
[The role of prostacycline, thromboxane A2 and prostaglandins in platelet aggregation (author's transl)].
Topics: Arteriosclerosis; Aspirin; Epoprostenol; Hemostasis; Platelet Aggregation; Prostaglandin Antagonists; Prostaglandins; Thrombosis; Thromboxane A2; Thromboxanes | 1981 |
[Prostacyclin and thromboxane in thrombosing atherosclerotic diseases].
Topics: Arteriosclerosis; Epoprostenol; Humans; Prostaglandins; Thrombosis; Thromboxane A2; Thromboxanes | 1981 |
[Prevention of thrombosis by thrombocyte inhibitors. 1. Cell regulation mechanisms and pharmacological modification].
Topics: Adenosine Diphosphate; Aspirin; Blood Platelets; Cyclic AMP; Dextrans; Dipyridamole; Humans; Hydroxychloroquine; Indomethacin; Platelet Aggregation; Prostaglandins; Sulfinpyrazone; Thrombosis; Thromboxane A2 | 1980 |
The interactions of lipids, platelets and endothelial cells in thrombogenesis.
Topics: Adenosine Diphosphate; Animals; Blood Platelets; Blood Vessels; Coronary Disease; Diet; Disease Models, Animal; Endothelium; Fatty Acids; Humans; Hyperlipoproteinemia Type II; Lipids; Lipoproteins, HDL; Lipoproteins, LDL; Thrombosis; Thromboxane A2 | 1980 |
[Function of prostaglandins in platelet aggregation].
Topics: Animals; Epoprostenol; Fibrinolytic Agents; Platelet Aggregation; Prostaglandins; Rabbits; Rats; Thrombosis; Thromboxane A2; Thromboxanes | 1980 |
The anti-thrombotic effects of prostacyclin.
Topics: Animals; Arteries; Aspirin; Cardiovascular Diseases; Dipyridamole; Epoprostenol; Fatty Acids; Hemostasis; Homeostasis; Humans; Lipid Peroxides; Platelet Aggregation; Prostaglandins; Thrombosis; Thromboxane A2 | 1980 |
Prostacyclin, thromboxane A2 interactions in haemostasis and thrombosis.
Prostacyclin and thromboxane A2 are products of arachidonic acid which play a role in the regulation of haemostatic plug and thrombus formation. Aspirin inhibits the synthesis of both compounds but is more active in blocking TXA2 formation; based on this, aspirin is suggested to have an anti-thrombotic effect. Other possible approaches to the development of anti-thrombotic drugs are discussed. Topics: Animals; Aspirin; Blood Platelets; Dose-Response Relationship, Drug; Epoprostenol; Humans; Platelet Aggregation; Prostaglandins; Thrombosis; Thromboxane A2; Thromboxanes | 1979 |
Eicosapentaenoic acid and prevention of thrombosis and atherosclerosis?
Unlike arachidonic acid (eicosatetraenoic acid, C20:4omega-6, A.A.), eicosapentaenoic acid (C20:5omega-3, E.P.A.) does not induce platelet aggregation in human platelet-rich plasma (P.R.P.), probably because of the formation of thromboxane A3 (T.X.A3) which does not have platelet aggregating properties. Moreover, E.P.A., like A.A., can be utilised by the vessel wall to make an anti-aggregating substance, probably a delta17-prostacyclin (P.G.I3). This finding suggests that, in vivo, high levels of E.P.A. and low levels of A.A. could lead to an antithrombotic state in which an active P.G.I3 and a non-active T.X.A3 are formed. Eskimos have high levels of E.P.A. and low levels of A.A. and they also have a low incidence of myocardial infarction and a tendency to bleed. It is possible that dietary enrichment with E.P.A. will protect against thrombosis. Topics: Arachidonic Acids; Arteriosclerosis; Blood Vessels; Denmark; Epoprostenol; Fatty Acids, Unsaturated; Greenland; Humans; Inuit; Models, Chemical; Myocardial Infarction; Platelet Aggregation; Thrombosis; Thromboxane A2 | 1978 |
Lipid-membrane interactions of platelets, coagulation and the arterial wall at the molecular level. Summary of workshop 2b: lipid-membrane interactions of platelets and coagulation with the arterial wall.
Topics: Animals; Blood Coagulation; Blood Platelets; Calcium; Cell Membrane; Female; Humans; Male; Prostaglandin Endoperoxides; Thrombosis; Thromboxane A2 | 1978 |
[Significance of arachidonic acid cascade in cardiology].
Topics: Arachidonic Acids; Humans; Models, Biological; Myocardial Infarction; Platelet Aggregation; Prostaglandins; Thrombosis; Thromboxane A2 | 1978 |
Unstable metabolites of arachidonic acid aspirin and the formation of the haemostatic plug.
Topics: Animals; Arachidonic Acids; Aspirin; Blood Coagulation Tests; Dose-Response Relationship, Drug; Hemostasis; Linoleic Acids; Male; Prostaglandins G; Rabbits; Thrombosis; Thromboxane A2 | 1978 |