Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 112
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Eur Respir J ; 63(2)2024 Feb.
Article in English | MEDLINE | ID: mdl-38123239

ABSTRACT

BACKGROUND: Short-term studies suggest that dietary nitrate (NO3 -) supplementation may improve the cardiovascular risk profile, lowering blood pressure (BP) and enhancing endothelial function. It is not clear if these beneficial effects are sustained and whether they apply in people with COPD, who have a worse cardiovascular profile than those without COPD. Nitrate-rich beetroot juice (NR-BRJ) is a convenient dietary source of nitrate. METHODS: The ON-BC trial was a randomised, double-blind, placebo-controlled parallel group study in stable COPD patients with home systolic BP (SBP) measurement ≥130 mmHg. Participants were randomly allocated (1:1) using computer-generated, block randomisation to either 70 mL NR-BRJ (400 mg NO3 -) (n=40) or an otherwise identical nitrate-depleted placebo juice (0 mg NO3 -) (n=41), once daily for 12 weeks. The primary end-point was between-group change in home SBP measurement. Secondary outcomes included change in 6-min walk distance (6MWD) and measures of endothelial function (reactive hyperaemia index (RHI) and augmentation index normalised to a heart rate of 75 beats·min-1 (AIx75)) using an EndoPAT device. Plasma nitrate and platelet function were also measured. RESULTS: Compared with placebo, active treatment lowered SBP (Hodges-Lehmann treatment effect -4.5 (95% CI -5.9- -3.0) mmHg), and improved 6MWD (30.0 (95% CI 15.7-44.2) m; p<0.001), RHI (0.34 (95% CI 0.03-0.63); p=0.03) and AIx75 (-7.61% (95% CI -14.3- -0.95%); p=0.026). CONCLUSIONS: In people with COPD, prolonged dietary nitrate supplementation in the form of beetroot juice produces a sustained reduction in BP, associated with an improvement in endothelial function and exercise capacity.


Subject(s)
Cardiovascular Diseases , Pulmonary Disease, Chronic Obstructive , Humans , Nitrates/therapeutic use , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/drug therapy , Dietary Supplements , Risk Factors , Blood Pressure , Antioxidants , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/drug therapy , Double-Blind Method , Cross-Over Studies
2.
Arterioscler Thromb Vasc Biol ; 42(1): 49-62, 2022 01.
Article in English | MEDLINE | ID: mdl-34809447

ABSTRACT

OBJECTIVE: Platelets are central to acute myocardial infarction (MI). How the platelet proteome is altered during MI is unknown. We sought to describe changes in the platelet proteome during MI and identify corresponding functional consequences. Approach and Results: Platelets from patients experiencing ST-segment-elevation MI (STEMI) before and 3 days after treatment (n=30) and matched patients with severe stable coronary artery disease before and 3 days after coronary artery bypass grafting (n=25) underwent quantitative proteomic analysis. Elevations in the proteins S100A8 and S100A9 were detected at the time of STEMI compared with stable coronary artery disease (S100A8: FC, 2.00; false discovery rate, 0.05; S100A9: FC, 2.28; false discovery rate, 0.005). During STEMI, only S100A8 mRNA and protein levels were correlated in platelets (R=0.46, P=0.012). To determine whether de novo protein synthesis occurs, activated platelets were incubated with 13C-labeled amino acids for 24 hours and analyzed by mass spectrometry. No incorporation was confidently detected. Platelet S100A8 and S100A9 was strongly correlated with neutrophil abundance at the time of STEMI. When isolated platelets and neutrophils were coincubated under quiescent and activated conditions, release of S100A8 from neutrophils resulted in uptake of S100A8 by platelets. Neutrophils released S100A8/A9 as free heterodimer, rather than in vesicles or extracellular traps. In the community-based Bruneck study (n=338), plasma S100A8/A9 was inversely associated with platelet reactivity-an effect abrogated by aspirin. CONCLUSIONS: Leukocyte-to-platelet protein transfer may occur in a thromboinflammatory environment such as STEMI. Plasma S100A8/A9 was negatively associated with platelet reactivity. These findings highlight neutrophils as potential modifiers for thrombotic therapies in coronary artery disease.


Subject(s)
Blood Platelets/metabolism , Calgranulin A/blood , Calgranulin B/blood , Neutrophil Activation , Neutrophils/metabolism , Platelet Activation , Proteome , ST Elevation Myocardial Infarction/blood , Aged , Case-Control Studies , Cell Line, Tumor , Female , Humans , Male , Middle Aged , Prospective Studies , Proteomics , ST Elevation Myocardial Infarction/diagnosis , ST Elevation Myocardial Infarction/therapy , Time Factors
3.
Platelets ; 34(1): 2200838, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37070955

ABSTRACT

Research into the natural aging process of platelets has garnered much research interest in recent years, and there have long been associations drawn between the proportion of newly formed platelets in the circulation and the risk of thrombosis. However, these observations have largely been demonstrated in patient groups in which there may be underlying systemic changes that effect platelet function. Recent advances in technology have allowed in-depth analysis of differently aged platelets isolated from the peripheral blood of healthy individuals and have demonstrated that aged platelets, often referred to as senescent platelets, undergo extensive changes in the transcriptome and proteome. Ultimately, these changes result in platelets whose functions have deteriorated such that they cannot partake in hemostatic responses to the same extent as newly formed platelets. Here, we review transcriptomic and proteomic research in platelet aging in the context of health and how this research sheds light upon alterations in platelet structure and function.


Platelets normally last within the human circulation for approximately 10 days, however there are a number of diseases in which this life span is notably reduced. People who have platelets with such reduced lifespans have higher risks of heart attacks and strokes and reduced responsiveness to standard anti-platelet drugs. Research in recent years has aimed to understand the age-related changes that occur within platelets as they circulate in the bloodstream. In this review article, the authors provide a summary of the changes that occur during the natural aging process of platelets in healthy people, highlighting reductions in the levels of RNA and proteins. Despite these general reductions in their own RNA and proteins, research has shown that as platelets circulate they take up other RNA transcripts and proteins from the bloodstream, and this too seems a hallmark of platelet aging. The authors also discuss age-related declines in various aspects of platelet function which renders them less effective participants in the formation of blood clots.


Subject(s)
Proteome , Transcriptome , Humans , Aged , Proteomics , Blood Platelets/physiology , Aging/genetics
4.
Proc Natl Acad Sci U S A ; 116(35): 17444-17449, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31405966

ABSTRACT

Trauma hemorrhage is a leading cause of death and disability worldwide. Platelets are fundamental to primary hemostasis, but become profoundly dysfunctional in critically injured patients by an unknown mechanism, contributing to an acute coagulopathy which exacerbates bleeding and increases mortality. The objective of this study was to elucidate the mechanism of platelet dysfunction in critically injured patients. We found that circulating platelets are transformed into procoagulant balloons within minutes of injury, accompanied by the release of large numbers of activated microparticles which coat leukocytes. Ballooning platelets were decorated with histone H4, a damage-associated molecular pattern released in massive quantities after severe injury, and exposure of healthy platelets to histone H4 recapitulated the changes in platelet structure and function observed in trauma patients. This is a report of platelet ballooning in human disease and of a previously unrecognized mechanism by which platelets contribute to the innate response to tissue damage.


Subject(s)
Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Hemorrhage/blood , Histones/metabolism , Wounds and Injuries/blood , Blood Coagulation , Blood Platelets/ultrastructure , Calcium/metabolism , Cell-Derived Microparticles/ultrastructure , Hemorrhage/etiology , Humans , Leukocytes/metabolism , Platelet Function Tests , Thrombin/biosynthesis , Wounds and Injuries/complications
5.
Haematologica ; 106(5): 1423-1432, 2021 05 01.
Article in English | MEDLINE | ID: mdl-32299908

ABSTRACT

We have identified a rare missense variant on chromosome 9, position 125145990 (GRCh37), in exon 8 in PTGS1 (the gene encoding cyclo-oxygenase 1, COX-1, the target of anti-thrombotic aspirin therapy). We report that in the homozygous state within a large consanguineous family this variant is associated with a bleeding phenotype and alterations in platelet reactivity and eicosanoid production. Western blotting and confocal imaging demonstrated that COX-1 was absent in the platelets of three family members homozygous for the PTGS1 variant but present in their leukocytes. Platelet reactivity, as assessed by aggregometry, lumi-aggregometry and flow cytometry, was impaired in homozygous family members, as were platelet adhesion and spreading. The productions of COX-derived eicosanoids by stimulated platelets were greatly reduced but there were no changes in the levels of urinary metabolites of COX-derived eicosanoids. The proband exhibited additional defects in platelet aggregation and spreading which may explain why her bleeding phenotype was slightly more severe than those of other homozygous affected relatives. This is the first demonstration in humans of the specific loss of platelet COX-1 activity and provides insight into its consequences for platelet function and eicosanoid metabolism. Notably despite the absence of thromboxane A2 (TXA2) formation by platelets, urinary TXA2 metabolites were in the normal range indicating these cannot be assumed as markers of in vivo platelet function. Results from this study are important benchmarks for the effects of aspirin upon platelet COX-1, platelet function and eicosanoid production as they define selective platelet COX-1 ablation within humans.


Subject(s)
Aspirin , Platelet Function Tests , Blood Platelets , Cyclooxygenase 1/genetics , Female , Humans , Platelet Aggregation/genetics , Thromboxane A2
6.
FASEB J ; 34(8): 10027-10040, 2020 08.
Article in English | MEDLINE | ID: mdl-32592197

ABSTRACT

Aspirin prevents thrombosis by inhibiting platelet cyclooxygenase (COX)-1 activity and the production of thromboxane (Tx)A2 , a pro-thrombotic eicosanoid. However, the non-platelet actions of aspirin limit its antithrombotic effects. Here, we used platelet-COX-1-ko mice to define the platelet and non-platelet eicosanoids affected by aspirin. Mass-spectrometry analysis demonstrated blood from platelet-COX-1-ko and global-COX-1-ko mice produced similar eicosanoid profiles in vitro: for example, formation of TxA2 , prostaglandin (PG) F2α , 11-hydroxyeicosatraenoic acid (HETE), and 15-HETE was absent in both platelet- and global-COX-1-ko mice. Conversely, in vivo, platelet-COX-1-ko mice had a distinctly different profile from global-COX-1-ko or aspirin-treated control mice, notably significantly higher levels of PGI2 metabolite. Ingenuity Pathway Analysis (IPA) predicted that platelet-COX-1-ko mice would be protected from thrombosis, forming less pro-thrombotic TxA2 and PGE2 . Conversely, aspirin or lack of systemic COX-1 activity decreased the synthesis of anti-aggregatory PGI2 and PGD2 at non-platelet sites leading to predicted thrombosis increase. In vitro and in vivo thrombosis studies proved these predictions. Overall, we have established the eicosanoid profiles linked to inhibition of COX-1 in platelets and in the remainder of the cardiovascular system and linked them to anti- and pro-thrombotic effects of aspirin. These results explain why increasing aspirin dosage or aspirin addition to other drugs may lessen antithrombotic protection.


Subject(s)
Aspirin/pharmacology , Blood Platelets/metabolism , Cyclooxygenase 1/physiology , Cyclooxygenase Inhibitors/pharmacology , Eicosanoids/metabolism , Membrane Proteins/physiology , Thrombosis/metabolism , Animals , Arachidonic Acid/administration & dosage , Blood Platelets/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thrombosis/drug therapy , Thrombosis/pathology
7.
Circ Res ; 125(9): 847-854, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31510878

ABSTRACT

RATIONALE: Endothelial cells (ECs) and platelets, which respectively produce antithrombotic prostacyclin and prothrombotic thromboxane A2, both express COX1 (cyclooxygenase1). Consequently, there has been no way to delineate any antithrombotic role for COX1-derived prostacyclin from the prothrombotic effects of platelet COX1. By contrast, an antithrombotic role for COX2, which is absent in platelets, is straightforward to demonstrate. This has resulted in an incomplete understanding of the relative importance of COX1 versus COX2 in prostacyclin production and antithrombotic protection in vivo. OBJECTIVE: We sought to identify the role, if any, of COX1-derived prostacyclin in antithrombotic protection in vivo and compare this to the established protective role of COX2. METHODS AND RESULTS: We developed vascular-specific COX1 knockout mice and studied them alongside endothelial-specific COX2 knockout mice. COX1 immunoreactivity and prostacyclin production were primarily associated with the endothelial layer of aortae; freshly isolated aortic ECs released >10-fold more prostacyclin than smooth muscle cells. Moreover, aortic prostacyclin production, the ability of aortic rings to inhibit platelet aggregation and plasma prostacyclin levels were reduced when COX1 was knocked out in ECs but not in smooth muscle cells. When thrombosis was measured in vivo after FeCl3 carotid artery injury, endothelial COX1 deletion accelerated thrombosis to a similar extent as prostacyclin receptor blockade. However, this effect was lost when COX1 was deleted from both ECs and platelets. Deletion of COX2 from ECs also resulted in a prothrombotic phenotype that was independent of local vascular prostacyclin production. CONCLUSIONS: These data demonstrate for the first time that, in healthy animals, endothelial COX1 provides an essential antithrombotic tone, which is masked when COX1 activity is lost in both ECs and platelets. These results help us define a new 2-component paradigm wherein thrombotic tone is regulated by both COX1 and COX2 through complementary but mechanistically distinct pathways.


Subject(s)
Cyclooxygenase 1/deficiency , Endothelium, Vascular/metabolism , Epoprostenol/metabolism , Fibrinolytic Agents/metabolism , Gene Deletion , Membrane Proteins/deficiency , Platelet Aggregation/physiology , Animals , Aorta/metabolism , Cyclooxygenase 1/genetics , Epoprostenol/genetics , Female , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic
8.
Circ Res ; 122(4): 555-559, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29298774

ABSTRACT

RATIONALE: The balance between vascular prostacyclin, which is antithrombotic, and platelet thromboxane A2, which is prothrombotic, is fundamental to cardiovascular health. Prostacyclin and thromboxane A2 are formed after the concerted actions of cPLA2α (cytosolic phospholipase A2) and COX (cyclooxygenase). Urinary 2,3-dinor-6-keto-PGF1α (PGI-M) and 11-dehydro-TXB2 (TX-M) have been taken as biomarkers of prostacyclin and thromboxane A2 formation within the circulation and used to explain COX biology and patient phenotypes, despite concerns that urinary PGI-M and TX-M originate in the kidney. OBJECTIVE: We report data from a remarkable patient carrying an extremely rare genetic mutation in cPLA2α, causing almost complete loss of prostacyclin and thromboxane A2, who was transplanted with a normal kidney resulting in an experimental scenario of whole-body cPLA2α knockout, kidney-specific knockin. By studying this patient, we can determine definitively the contribution of the kidney to the productions of PGI-M and TX-M and test their validity as markers of prostacyclin and thromboxane A2 in the circulation. METHODS AND RESULTS: Metabolites were measured using liquid chromatography-tandem mass spectrometry. Endothelial cells were grown from blood progenitors. Before kidney transplantation, the patient's endothelial cells and platelets released negligible levels of prostacyclin (measured as 6-keto-prostaglandin F1α) and thromboxane A2 (measured as TXB2), respectively. Likewise, the urinary levels of PGI-M and TX-M were very low. After transplantation and the establishment of normal renal function, the levels of PGI-M and TX-M in the patient's urine rose to within normal ranges, whereas endothelial production of prostacyclin and platelet production of thromboxane A2 remained negligible. CONCLUSIONS: These data show that PGI-M and TX-M can be derived exclusively from the kidney without contribution from prostacyclin made by endothelial cells or thromboxane A2 by platelets in the general circulation. Previous work relying on urinary metabolites of prostacyclin and thromboxane A2 as markers of whole-body endothelial and platelet function now requires reevaluation.


Subject(s)
6-Ketoprostaglandin F1 alpha/analogs & derivatives , Allografts/metabolism , Kidney Transplantation , Kidney/metabolism , Loss of Function Mutation , Phospholipases A2, Cytosolic/genetics , Thromboxane B2/analogs & derivatives , 6-Ketoprostaglandin F1 alpha/metabolism , 6-Ketoprostaglandin F1 alpha/urine , Biomarkers/urine , Cells, Cultured , Female , Humans , Middle Aged , Phenotype , Phospholipases A2, Cytosolic/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Thromboxane B2/metabolism , Thromboxane B2/urine
9.
Circ Res ; 118(3): 420-432, 2016 Feb 05.
Article in English | MEDLINE | ID: mdl-26646931

ABSTRACT

RATIONALE: Platelets shed microRNAs (miRNAs). Plasma miRNAs change on platelet inhibition. It is unclear whether plasma miRNA levels correlate with platelet function. OBJECTIVE: To link small RNAs to platelet reactivity. METHODS AND RESULTS: Next-generation sequencing of small RNAs in plasma revealed 2 peaks at 22 to 23 and 32 to 33 nucleotides corresponding to miRNAs and YRNAs, respectively. Among YRNAs, predominantly, fragments of RNY4 and RNY5 were detected. Plasma miRNAs and YRNAs were measured in 125 patients with a history of acute coronary syndrome who had undergone detailed assessment of platelet function 30 days after the acute event. Using quantitative real-time polymerase chain reactions, 92 miRNAs were assessed in patients with acute coronary syndrome on different antiplatelet therapies. Key platelet-related miRNAs and YRNAs were correlated with platelet function tests. MiR-223 (rp=0.28; n=121; P=0.002), miR-126 (rp=0.22; n=121; P=0.016), and other abundant platelet miRNAs and YRNAs showed significant positive correlations with the vasodilator-stimulated phosphoprotein phosphorylation assay. YRNAs, miR-126, and miR-223 were also among the small RNAs showing the greatest dependency on platelets and strongly correlated with plasma levels of P-selectin, platelet factor 4, and platelet basic protein in the population-based Bruneck study (n=669). A single-nucleotide polymorphism that facilitates processing of pri-miR-126 to mature miR-126 accounted for a rise in circulating platelet activation markers. Inhibition of miR-126 in mice reduced platelet aggregation. MiR-126 directly and indirectly affects ADAM9 and P2Y12 receptor expression. CONCLUSIONS: Levels of platelet-related plasma miRNAs and YRNAs correlate with platelet function tests in patients with acute coronary syndrome and platelet activation markers in the general population. Alterations in miR-126 affect platelet reactivity.


Subject(s)
Acute Coronary Syndrome/blood , Blood Platelets/metabolism , MicroRNAs/blood , Platelet Activation , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/genetics , Animals , Blood Platelets/drug effects , Cell Line, Tumor , Gene Expression Profiling/methods , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Oligonucleotides/genetics , Oligonucleotides/metabolism , Platelet Activation/drug effects , Platelet Activation/genetics , Platelet Aggregation Inhibitors/therapeutic use , Platelet Function Tests , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Transfection
10.
Arterioscler Thromb Vasc Biol ; 37(5): 949-956, 2017 05.
Article in English | MEDLINE | ID: mdl-28279968

ABSTRACT

OBJECTIVE: Aspirin together with thienopyridine P2Y12 inhibitors, commonly clopidogrel, is a cornerstone of antiplatelet therapy. However, many patients receiving this therapy display high on-treatment platelet reactivity, which is a major therapeutic hurdle to the prevention of recurrent thrombotic events. The emergence of uninhibited platelets after thrombopoiesis has been proposed as a contributing factor to high on-treatment platelet reactivity. Here, we investigate the influences of platelet turnover on platelet aggregation in the face of different dual-antiplatelet therapy strategies. APPROACH AND RESULTS: Traditional light transmission aggregometry, cytometry, advanced flow cytometric imaging, and confocal microscopy were used to follow the interactions of populations of platelets from healthy volunteers and patients with stable cardiovascular disease. Newly formed, reticulated platelets overproportionately contributed to, and clustered at, the core of forming aggregates. This phenomenon was particularly observed in samples from patients treated with aspirin plus a thienopyridine, but was absent in samples taken from patients treated with aspirin plus ticagrelor. CONCLUSIONS: Reticulated platelets are more reactive than older platelets and act as seeds for the formation of platelet aggregates even in the presence of antiplatelet therapy. This is coherent with the emergence of an uninhibited subpopulation of reticulated platelets during treatment with aspirin plus thienopyridine, explained by the short pharmacokinetic half-lives of these drugs. This phenomenon is absent during treatment with ticagrelor, because of its longer half-life and ability to act as a circulating inhibitor. These data highlight the important influences of pharmacokinetics on antiplatelet drug efficacies, especially in diseases associated with increased platelet turnover.


Subject(s)
Adenosine/analogs & derivatives , Aspirin/pharmacokinetics , Blood Platelets/drug effects , Coronary Artery Disease/drug therapy , Platelet Aggregation Inhibitors/pharmacokinetics , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Thienopyridines/pharmacokinetics , Thrombopoiesis , Adenosine/administration & dosage , Adenosine/pharmacokinetics , Adolescent , Adult , Aged , Aged, 80 and over , Aspirin/administration & dosage , Blood Platelets/metabolism , Case-Control Studies , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Drug Therapy, Combination , Half-Life , Humans , Male , Middle Aged , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Platelet Function Tests , Purinergic P2Y Receptor Antagonists/administration & dosage , Purinergic P2Y Receptor Antagonists/adverse effects , Thienopyridines/administration & dosage , Ticagrelor , Young Adult
11.
Arterioscler Thromb Vasc Biol ; 37(8): 1482-1493, 2017 08.
Article in English | MEDLINE | ID: mdl-28619996

ABSTRACT

OBJECTIVES: The liver X receptors (LXRs) and farnesoid X receptor (FXR) have been identified in human platelets. Ligands of these receptors have been shown to have nongenomic inhibitory effects on platelet activation by platelet agonists. This, however, seems contradictory with the platelet hyper-reactivity that is associated with several pathological conditions that are associated with increased circulating levels of molecules that are LXR and FXR ligands, such as hyperlipidemia, type 2 diabetes mellitus, and obesity. APPROACH AND RESULTS: We, therefore, investigated whether ligands for the LXR and FXR receptors were capable of priming platelets to the activated state without stimulation by platelet agonists. Treatment of platelets with ligands for LXR and FXR converted platelets to the procoagulant state, with increases in phosphatidylserine exposure, platelet swelling, reduced membrane integrity, depolarization of the mitochondrial membrane, and microparticle release observed. Additionally, platelets also displayed features associated with coated platelets such as P-selectin exposure, fibrinogen binding, fibrin generation that is supported by increased serine protease activity, and inhibition of integrin αIIbß3. LXR and FXR ligand-induced formation of coated platelets was found to be dependent on both reactive oxygen species and intracellular calcium mobilization, and for FXR ligands, this process was found to be dependent on cyclophilin D. CONCLUSIONS: We conclude that treatment with LXR and FXR ligands initiates coated platelet formation, which is thought to support coagulation but results in desensitization to platelet stimuli through inhibition of αIIbß3 consistent with their ability to inhibit platelet function and stable thrombus formation in vivo.


Subject(s)
Benzoates/pharmacology , Benzylamines/pharmacology , Blood Coagulation/drug effects , Blood Platelets/drug effects , Isoxazoles/pharmacology , Liver X Receptors/agonists , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Blood Platelets/metabolism , Calcium Signaling/drug effects , Cell-Derived Microparticles/drug effects , Cell-Derived Microparticles/metabolism , Cyclophilins/blood , Dose-Response Relationship, Drug , Fibrin/metabolism , Fibrinogen/metabolism , Humans , Ligands , Liver X Receptors/blood , Membrane Potential, Mitochondrial/drug effects , P-Selectin/blood , Phosphatidylserines/blood , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Reactive Oxygen Species/blood , Receptors, Cytoplasmic and Nuclear/blood
12.
Platelets ; 29(7): 650-655, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29543546

ABSTRACT

While there are many bench and bedside tests to assess platelet reactivity, ex vivo light transmission aggregometry (LTA) remains the gold standard. LTA, however, is expensive, time-consuming and requires dedicated equipment and staff, making it impractical in many situations. In addition, there is significant variability between data generated at different testing sites meaning that tests often need to be repeated if a patient is transferred to the care of a different hospital. As such, there is clearly an unmet need for standardization of platelet testing. Using the principles of LTA, aggregometry can be conducted in 96-well plates with readings being made in a standard plate reader. This approach allows for the assessment of multiple concentrations of agonists, since the volume of platelets required for each test is significantly lower than for LTA. Furthermore, the lyophilization of a set panel of agonists to a 96-well plate to produce a stable assay substrate allows the production of portable, standardized plates that can be used to generate reproducible tests at multiple sites. In this review, we will discuss the methods and uses of 96-well plate aggregometry for both research and the clinic.


Subject(s)
Platelet Aggregation , Platelet Function Tests , Animals , Humans , Platelet Aggregation/drug effects , Platelet Function Tests/instrumentation , Platelet Function Tests/methods , Research
13.
Platelets ; 29(7): 686-689, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29715047

ABSTRACT

In this short article, submitted as part of the review on platelet function testing, we illustrate the quantitative and qualitative differences between classical light transmission aggregometry (LTA) and 96-well plate aggregometry. We show that responses to platelet agonists and antagonists differ depending upon the method of aggregation testing. For example, in 96-well aggregometry, responses to collagen are strongly inhibited by P2Y12 receptor antagonists while in LTA they are much less affected. Furthermore, we explore the importance of differences in the mechanical environment upon platelet aggregation. We emphasize that LTA and 96-well aggregometry are not interchangeable assays. These two assays are best used as complementary tests to explore platelet function in depth.


Subject(s)
Platelet Aggregation , Platelet Function Tests , Adenosine Diphosphate/metabolism , Biomarkers , Humans , Platelet Activation , Platelet Function Tests/instrumentation , Platelet Function Tests/methods
14.
Platelets ; 29(2): 162-170, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28503981

ABSTRACT

Despite the interwoven nature of platelet activation and the coagulation system in thrombosis, few studies relate both analysis of protein and cellular parts of coagulation in the same population. In the present study, we use matched ex vivo samples to determine the influences of standard antiplatelet therapies on platelet function and use advanced rheological analyses to assess clot formation. Healthy volunteers were recruited following fully informed consent then treated for 7 days with single antiplatelet therapy of aspirin (75 mg) or prasugrel (10 mg) or with dual antiplatelet therapy (DAPT) using aspirin (75 mg) plus prasugrel (10 mg) or aspirin (75 mg) plus ticagrelor (90 mg). Blood samples were taken at day 0 before treatment and at day 7 following treatment. We found that aspirin plus prasugrel or aspirin plus ticagrelor inhibited platelet responses to multiple agonists and reduced P-selectin expression. Significant platelet inhibition was coupled with a reduction in fractal dimension corresponding to reductions in mean relative mass both for aspirin plus prasugrel (-35 ± 16% change, p = 0.04) and for aspirin plus ticagrelor (-45 ± 14% change, p = 0.04). Aspirin alone had no effect upon measures of clot structure, whereas prasugrel reduced fractal dimension and mean relative mass. These data demonstrate that platelets are important determinants of clot structure as assessed by fractal dimension (df) and that effective platelet inhibition is associated with a weaker, more permeable fibrin network. This indicates a strong association between the therapeutic benefits of antiplatelet therapies and their abilities to reduce thrombus density that may be useful in individual patients to determine the functional relationship between platelet reactivity, eventual clot quality, and clinical outcome. df could represent a novel risk stratification biomarker useful in individualizing antiplatelet therapies.


Subject(s)
Blood Coagulation/drug effects , Blood Platelets/metabolism , Platelet Activation/drug effects , Thrombosis/metabolism , Female , Fractals , Humans , Male
15.
J Biol Chem ; 291(26): 13448-64, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129261

ABSTRACT

Eicosanoids are important mediators of fever, pain, and inflammation that modulate cell signaling during acute and chronic disease. We show by using lipidomics that thrombin-activated human platelets generate a new type of eicosanoid that both stimulates and primes human neutrophil integrin (Mac-1) expression, in response to formylmethionylleucylphenylalanine. Detailed characterization proposes a dioxolane structure, 8-hydroxy-9,11-dioxolane eicosatetraenoic acid (dioxolane A3, DXA3). The lipid is generated in nanogram amounts by platelets from endogenous arachidonate during physiological activation, with inhibition by aspirin in vitro or in vivo, implicating cyclooxygenase-1 (COX). Pharmacological and genetic studies on human/murine platelets revealed that DXA3 formation requires protease-activated receptors 1 and 4, cytosolic phospholipase A2 (cPLA2), Src tyrosine kinases, p38 MAPK, phospholipase C, and intracellular calcium. From data generated by purified COX isoforms and chemical oxidation, we propose that DXA3 is generated by release of an intermediate from the active site followed by oxygenation at C8. In summary, a new neutrophil-activating platelet-derived lipid generated by COX-1 is presented that can activate or prime human neutrophils, suggesting a role in innate immunity and acute inflammation.


Subject(s)
Blood Platelets/enzymology , Cyclooxygenase 1/metabolism , Dioxolanes/metabolism , MAP Kinase Signaling System/physiology , Neutrophil Activation/physiology , Neutrophils/metabolism , Animals , Aspirin/pharmacology , Blood Platelets/immunology , Cyclooxygenase 1/immunology , Dioxolanes/immunology , Immunity, Innate/drug effects , Immunity, Innate/physiology , MAP Kinase Signaling System/drug effects , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/metabolism , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Neutrophil Activation/drug effects , Neutrophils/immunology , Platelet Activation/drug effects , Platelet Activation/physiology
16.
Blood ; 126(10): e11-8, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26215112

ABSTRACT

Testing of platelet function is central to the cardiovascular phenotyping of genetically modified mice. Traditional platelet function tests have been developed primarily for testing human samples and the volumes required make them highly unsuitable for the testing of mouse platelets. This limits research in this area. To address this problem, we have developed a miniaturized whole blood aggregometry assay, based on a readily accessible 96-well plate format coupled with quantification of single platelet depletion by flow cytometric analysis. Using this approach, we observed a concentration-dependent loss of single platelets in blood exposed to arachidonic acid, collagen, U46619 or protease activated receptor 4 activating peptide. This loss was sensitive to well-established antiplatelet agents and genetic manipulation of platelet activation pathways. Observations were more deeply analyzed by flow cytometric imaging, confocal imaging, and measurement of platelet releasates. Phenotypic analysis of the reactivity of platelets taken from mice lacking intercellular adhesion molecule (ICAM)-1 identified a marked decrease in fibrinogen-dependent platelet-monocyte interactions, especially under inflammatory conditions. Such findings exemplify the value of screening platelet phenotypes of genetically modified mice and shed further light upon the roles and interactions of platelets in inflammation.


Subject(s)
Blood Platelets/metabolism , Intercellular Adhesion Molecule-1/metabolism , Monocytes/metabolism , Platelet Aggregation/physiology , Platelet Function Tests/methods , Animals , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Phenotype , Sensitivity and Specificity
17.
FASEB J ; 30(12): 4256-4266, 2016 12.
Article in English | MEDLINE | ID: mdl-27633788

ABSTRACT

Regular consumption of low-dose aspirin reduces the occurrence of colorectal, esophageal, stomach, and gastrointestinal cancers. The underlying mechanism is unknown but may be linked to inhibition of angiogenesis. Because the effective doses of aspirin are consistent with the inhibition of cyclooxygenase-1 in platelets, we used liquid chromatography with tandem mass spectrometry analyses and immunoassays of human platelet releasates coupled with angiogenesis assays to search for the mediators of these effects. Blood or platelet-rich plasma from healthy volunteers stimulated with platelet activators produced a broad range of eicosanoids. Notably, preincubation of platelets with aspirin, but not with a P2Y12 receptor antagonist, caused a marked reduction in the production of 11-hydroxyeicosatetraenoic acid (HETE) and 15(S)-HETE, in addition to prostanoids such as thromboxane A2 Releasates from activated platelets caused cell migration and tube formation in cultured human endothelial cells and stimulated the sprouting of rat aortic rings in culture. These proangiogenic effects were absent when platelets were treated with aspirin but returned by coincubation with exogenous 15(S)-HETE. These results reveal 15(S)-HETE as a major platelet cyclooxygenase-1 product with strong proangiogenic effects. Thus, 15(S)-HETE represents a potential target for the development of novel antiangiogenic therapeutics, and blockade of its production may provide a mechanism for the anticancer effects of aspirin.-Rauzi, F., Kirkby, N. S., Edin, M. L., Whiteford, J. Zeldin, D. C., Mitchell, J. A., Warner, T. D. Aspirin inhibits the production of proangiogenic 15(S)-HETE by platelet cyclooxygenase-1.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Cyclooxygenase 1/metabolism , Endothelium/drug effects , Hydroxyeicosatetraenoic Acids/metabolism , Platelet Aggregation/drug effects , Cells, Cultured , Cyclooxygenase 1/drug effects , Eicosanoids/metabolism , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism
19.
Circulation ; 131(7): 633-42, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25492024

ABSTRACT

BACKGROUND: Cardiovascular side effects associated with cyclooxygenase-2 inhibitor drugs dominate clinical concern. Cyclooxygenase-2 is expressed in the renal medulla where inhibition causes fluid retention and increased blood pressure. However, the mechanisms linking cyclooxygenase-2 inhibition and cardiovascular events are unknown and no biomarkers have been identified. METHODS AND RESULTS: Transcriptome analysis of wild-type and cyclooxygenase-2(-/-) mouse tissues revealed 1 gene altered in the heart and aorta, but >1000 genes altered in the renal medulla, including those regulating the endogenous nitric oxide synthase inhibitors asymmetrical dimethylarginine (ADMA) and monomethyl-l-arginine. Cyclo-oxygenase-2(-/-) mice had increased plasma levels of ADMA and monomethyl-l-arginine and reduced endothelial nitric oxide responses. These genes and methylarginines were not similarly altered in mice lacking prostacyclin receptors. Wild-type mice or human volunteers taking cyclooxygenase-2 inhibitors also showed increased plasma ADMA. Endothelial nitric oxide is cardio-protective, reducing thrombosis and atherosclerosis. Consequently, increased ADMA is associated with cardiovascular disease. Thus, our study identifies ADMA as a biomarker and mechanistic bridge between renal cyclooxygenase-2 inhibition and systemic vascular dysfunction with nonsteroidal anti-inflammatory drug usage. CONCLUSIONS: We identify the endogenous endothelial nitric oxide synthase inhibitor ADMA as a biomarker and mechanistic bridge between renal cyclooxygenase-2 inhibition and systemic vascular dysfunction.


Subject(s)
Anti-Inflammatory Agents/adverse effects , Arginine/analogs & derivatives , Cardiovascular Diseases/blood , Cyclooxygenase 2 Inhibitors/adverse effects , Cyclooxygenase 2/deficiency , Adult , Animals , Arginine/blood , Biomarkers/blood , Cardiovascular Diseases/drug therapy , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Young Adult
20.
Blood ; 123(8): e11-22, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24408324

ABSTRACT

Up to 1% of the population have mild bleeding disorders, but these remain poorly characterized, particularly with regard to the roles of platelets. We have compared the usefulness of Optimul, a 96-well plate-based assay of 7 distinct pathways of platelet activation to characterize inherited platelet defects in comparison with light transmission aggregometry (LTA). Using Optimul and LTA, concentration-response curves were generated for arachidonic acid, ADP, collagen, epinephrine, Thrombin receptor activating-peptide, U46619, and ristocetin in samples from (1) healthy volunteers (n = 50), (2) healthy volunteers treated with antiplatelet agents in vitro (n = 10), and (3) patients with bleeding of unknown origin (n = 65). The assays gave concordant results in 82% of cases (κ = 0.62, P < .0001). Normal platelet function results were particularly predictive (sensitivity, 94%; negative predictive value, 91%), whereas a positive result was not always substantiated by LTA (specificity, 67%; positive predictive value, 77%). The Optimul assay was significantly more sensitive at characterizing defects in the thromboxane pathway, which presented with normal responses with LTA. The Optimul assay is sensitive to mild platelet defects, could be used as a rapid screening assay in patients presenting with bleeding symptoms, and detects changes in platelet function more readily than LTA. This trial was registered at www.isrctn.org as #ISRCTN 77951167.


Subject(s)
Blood Platelet Disorders/diagnosis , Drug Monitoring/methods , Hemorrhage/diagnosis , High-Throughput Screening Assays/methods , Platelet Activation/physiology , Platelet Aggregation Inhibitors/pharmacology , Adult , Blood Platelet Disorders/blood , Blood Platelet Disorders/genetics , Blood Platelets/drug effects , Blood Platelets/physiology , Female , Genetic Association Studies , Healthy Volunteers , Hemorrhage/blood , Hemorrhage/physiopathology , Humans , Male , Platelet Activation/drug effects , Predictive Value of Tests , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Sensitivity and Specificity , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL