Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
1.
J Thromb Haemost ; 16(12): 2515-2519, 2018 12.
Article in English | MEDLINE | ID: mdl-30264918

ABSTRACT

Essentials Thrombopoietin (TPO) lowers the threshold for platelet activation. TPO receptor agonists (RAs) may therefore also lead to platelet activation. Patients with chronic liver disease and thrombocytopenia participated in a randomized trial. The TPO-RA avatrombopag did not increase platelet activation in vivo or reactivity in vitro. BACKGROUND: The thrombopoietin (TPO) receptor agonist (TPO-RA) avatrombopag has recently been Food and Drug Administration-approved for the treatment of thrombocytopenia in patients with chronic liver disease (CLD) scheduled for a procedure. The TPO receptor c-mpl is expressed on the platelet surface, and TPO lowers the threshold for platelet activation. TPO-RAs may therefore also lead to platelet activation. OBJECTIVES: To evaluate the effects of avatrombopag on platelet activation. PATIENTS/METHODS: CLD patients with thrombocytopenia participated in a randomized, double-blind, placebo-controlled, parallel-group study. No patient received a platelet transfusion within 10 days of study blood draws. Platelet activation was evaluated with whole blood flow cytometry (which, unlike other methods, is accurate in thrombocytopenic samples). RESULTS: Avatrombopag, but not placebo, increased platelet counts. As measured by platelet surface P-selectin and activated glycoprotein IIb-IIIa: (i) the numbers of circulating activated platelets were not increased in avatrombopag-treated patients as compared with placebo-treated patients; and (ii) platelet reactivity to low and high concentrations of ADP and thrombin receptor-activating peptide was not increased in avatrombopag-treated patients as compared with placebo-treated patients. CONCLUSIONS: In this randomized, double-blind, placebo-controlled, parallel-group study of CLD patients with thrombocytopenia, avatrombopag increased platelet counts but did not increase platelet activation in vivo or platelet reactivity in vitro.


Subject(s)
Blood Platelets/drug effects , Hematologic Agents/therapeutic use , Liver Diseases/complications , Platelet Activation/drug effects , Thiazoles/therapeutic use , Thiophenes/therapeutic use , Thrombocytopenia/drug therapy , Thrombopoiesis/drug effects , Blood Platelets/metabolism , Chronic Disease , Double-Blind Method , Humans , Platelet Count , Thrombocytopenia/blood , Thrombocytopenia/etiology , Time Factors , Treatment Outcome , United States
3.
Thromb Haemost ; 118(1): 143-151, 2018 01.
Article in English | MEDLINE | ID: mdl-29304534

ABSTRACT

BACKGROUND: Treatment decisions for patients with immune thrombocytopenia (ITP) are difficult because patients with similarly low platelet counts differ in their bleeding tendency. We recently reported that platelet function tests, independent of platelet count, are associated with concurrent bleeding severity, suggesting that these tests may be useful indicators of future bleeding in ITP. OBJECTIVES: To test this hypothesis, we evaluated the consistency of these platelet function tests over time and their association with subsequent bleeding severity. METHODS: Bleeding score and platelet biomarkers were evaluated in a cross-sectional study of children with ITP at two visits separated by a median of 10 months. RESULTS AND CONCLUSIONS: Correlations between Visit 1 and Visit 2 results for immature platelet fraction, circulating and agonist-stimulated platelet surface P-selectin, and activated GPIIb-IIIa and GPIbα indicated consistency of the platelet phenotype over time. Consistent with our previous findings, platelet biomarkers at each visit were significantly associated with the concurrent bleeding score. Furthermore, increased P-selectin on circulating platelets and reduced agonist-stimulated P-selectin and activated GPIIb-IIIa-positive platelets at Visit 1 were significantly associated with bleeding scores at Visit 2 and remained significantly associated with bleeding severity after adjustment for platelet count. These results suggest a mechanistic link between desensitization of agonist receptors and increased bleeding severity. In summary, platelet function in ITP, independent of platelet count, is consistent over time and is associated with both concurrent and subsequent bleeding severity. These findings support further evaluation of platelet function testing to help guide patient management in ITP.


Subject(s)
Blood Platelets/physiology , Hemorrhage/physiopathology , Platelet Count , Platelet Function Tests , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/physiopathology , Adolescent , Biomarkers/metabolism , Child , Child, Preschool , Cross-Sectional Studies , Female , Humans , Infant , Male , P-Selectin/metabolism , Phenotype , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Risk Factors
4.
J Thromb Haemost ; 15(5): 858-867, 2017 05.
Article in English | MEDLINE | ID: mdl-28092426

ABSTRACT

Essentials Irreversible platelet inhibition persists after reversibly-binding ticagrelor is discontinued. Reversibility of platelet inhibition by ticagrelor and its active metabolite was assessed. Incomplete recovery was observed after prolonged exposure to ticagrelor. Activated GPIIb-IIIa and P-selectin, not platelet reactivity index, showed irreversibility. SUMMARY: Introduction Ticagrelor is described as a reversible P2Y12 antagonist. However, residual platelet inhibition persists after discontinuation of ticagrelor when plasma levels are undetectable. We assessed the reversibility of platelet inhibition by ticagrelor and its active metabolite (T-AM) in comparison with cangrelor and prasugrel's active metabolite (P-AM). Methods Whole blood was treated in vitro with ~ 50% inhibitory concentrations of ticagrelor, T-AM, cangrelor, P-AM and assessed for ADP-stimulated activated GPIIb-IIIa and P-selectin and vasodilator-stimulated phosphoprotein (VASP) platelet reactivity index (PRI) before and after 100-fold dilution. Results Platelets exposed for 30 min to ticagrelor, T-AM or cangrelor showed full recovery of activated GPIIb-IIIa but only partial recovery of P-selectin. Longer exposure (24 h) to the drug decreased reversibility of activated GPIIb-IIIa by ticagrelor (65.1% [49.5-80.6], % of vehicle with 95% confidence interval [CI]) and T-AM (88.8% [79.2-98.3]), but not by cangrelor (101.4% [96.4-106.4]). Compared with 30 min exposure, the reversibility of P-selectin further decreased after 24 h exposure to ticagrelor (from 91.8% [82.1-101.5] to 51.8% [45.5-85.0]), but not T-AM (from 79.0% [67.8-90.3] to 77.4% [61.8-93.1]) or cangrelor (from 76.0% [67.6-84.4] to 76.2% [70.6-81.8]). In contrast, 24 h exposure to ticagrelor, T-AM and cangrelor resulted in full recovery of platelet reactivity as measured by PRI. Platelets exposed to P-AM showed no recovery of ADP reactivity. Conclusions Incomplete recovery after prolonged exposure to ticagrelor, observed by activated GPIIb-IIIa and P-selectin but not upstream VASP signaling, suggests that P2Y12 regains functionality and irreversible changes occur independent of VASP signaling.


Subject(s)
Adenosine/analogs & derivatives , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Adenosine/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Blood Platelets/metabolism , Cell Adhesion Molecules/blood , Dose-Response Relationship, Drug , Humans , Kinetics , Microfilament Proteins/blood , P-Selectin/blood , Phosphoproteins/blood , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Prasugrel Hydrochloride/pharmacology , Receptors, Purinergic P2Y12/blood , Receptors, Purinergic P2Y12/drug effects , Signal Transduction/drug effects , Ticagrelor
5.
Platelets ; 27(2): 128-35, 2016.
Article in English | MEDLINE | ID: mdl-26030682

ABSTRACT

Therapeutic use of activated platelet-rich plasma (PRP) has been explored for wound healing, hemostasis and antimicrobial wound applications. Pulse electric field (PEF) stimulation may provide more consistent platelet activation and avoid complications associated with the addition of bovine thrombin, the current state of the art ex vivo activator of therapeutic PRP. The aim of this study was to compare the ability of PEF, bovine thrombin and thrombin receptor activating peptide (TRAP) to activate human PRP, release growth factors and induce cell proliferation in vitro. Human PRP was prepared in the Harvest SmartPreP2 System and treated with vehicle, PEF, bovine thrombin, TRAP or Triton X-100. Platelet activation and procoagulant markers and microparticle generation were measured by flow cytometry. Released growth factors were measured by ELISA. The releasates were tested for their ability to stimulate proliferation of human epithelial cells in culture. PEF produced more platelet-derived microparticles, P-selectin-positive particles and procoagulant annexin V-positive particles than bovine thrombin or TRAP. These differences were associated with higher levels of released epidermal growth factor after PEF than after bovine thrombin or TRAP but similar levels of platelet-derived, vascular-endothelial, and basic fibroblast growth factors, and platelet factor 4. Supernatant from PEF-treated platelets significantly increased cell proliferation compared to plasma. In conclusion, PEF treatment of fresh PRP results in generation of microparticles, exposure of prothrombotic platelet surfaces, differential release of growth factors compared to bovine thrombin and TRAP and significant cell proliferation. These results, together with PEF's inherent advantages, suggest that PEF may be a superior alternative to bovine thrombin activation of PRP for therapeutic applications.


Subject(s)
Electricity , Epidermal Growth Factor/metabolism , Platelet-Rich Plasma/cytology , Animals , Annexin A5/biosynthesis , Annexin A5/genetics , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Cattle , Cell Line , Cell Proliferation/drug effects , Cell-Derived Microparticles/metabolism , Electric Stimulation , Epidermal Growth Factor/biosynthesis , Epidermal Growth Factor/genetics , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Flow Cytometry , Gene Expression , Humans , Octoxynol/pharmacology , P-Selectin/biosynthesis , P-Selectin/genetics , Platelet Activation/drug effects , Platelet-Rich Plasma/metabolism , Receptors, Thrombin/chemistry , Thrombin/pharmacology
6.
Int J Lab Hematol ; 37(1): 112-9, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24806286

ABSTRACT

INTRODUCTION: Early and accurate identification of acute coronary syndrome (ACS) vs. noncardiac chest pain in patients presenting to the emergency department (ED) is problematic and new diagnostic markers are needed. Previous studies reported that elevated mean platelet volume (MPV) is associated with ACS and predictive of cardiovascular risk. MPV is closely related to the immature platelet fraction (IPF), and recent studies have suggested that IPF may be a more sensitive marker of ACS than MPV. The objective of the present study was to determine whether the measurement of IPF assists in the diagnosis of ACS in patients presenting to the ED with chest pain. METHODS: In this single-center, prospective, cross-sectional study, adult patients presenting to the ED with chest pain and/or suspected ACS were considered for enrollment. Blood samples from 236 ACS-negative and 44 ACS-positive patients were analyzed in a Sysmex XE-2100 for platelet count, MPV, IPF, and the absolute count of immature platelets (IPC). RESULTS: Total platelet counts, MPV, IPF, and IPC were not statistically different between ACS-negative and ACS-positive patients. The IPF was 4.6 ± 2.7% and 5.0 ± 2.8% (mean ± SD, P = 0.24), and the IPC was 10.0 ± 4.6 and 11.5 ± 7.5 × 10(3) /µL (P = 0.27) for ACS-negative and ACS-positive patients, respectively. CONCLUSION: In 280 patients presenting to the ED with chest pain and/or suspected ACS, no differences in IPF, IPC or MPV were observed in ACS-negative vs. ACS-positive patients, suggesting that these parameters do not assist in the diagnosis of ACS.


Subject(s)
Acute Coronary Syndrome/blood , Acute Coronary Syndrome/diagnosis , Blood Platelets/cytology , Chest Pain/diagnosis , Chest Pain/etiology , Emergency Service, Hospital , Platelet Count , Aged , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Prospective Studies , Risk Factors
7.
J Thromb Haemost ; 12(1): 2-13, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24406062

ABSTRACT

Polypharmacy in patients undergoing coronary artery stenting or in those presenting with an acute coronary syndrome is common. Nevertheless, the risk of drug-drug interactions in patients treated simultaneously with P2Y12 receptor inhibitors is less well considered in routine clinical practice. Whereas the irreversible P2Y12 receptor inhibitors clopidogrel and prasugrel are prodrugs requiring cytochrome P450 (CYP) enzymes for metabolic activation, such activation is not necessary for the direct-acting reversible P2Y12 receptor inhibitor ticagrelor. Several drugs frequently used in cardiology have been shown to interact with the metabolism of P2Y12 receptor inhibitors in pharmacodynamic studies. Whereas several drug-drug interactions have been described for clopidogrel and ticagrelor, prasugrel seems to have a low potential for drug-drug interactions. The clinical implications of these interactions have raised concern. In general, concomitant administration of P2Y12 receptor antagonists and strong inhibitors or inducers of CYP3A/CYP2C19 should be performed with caution in patients treated with clopidogrel/ticagrelor. Under most circumstances, clinicians have the option of prescribing alternative drugs with less risk of drug-drug interactions when used concomitantly with P2Y12 receptor inhibitors.


Subject(s)
Purinergic Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Cytochromes/metabolism , Drug Interactions , Humans
8.
J Thromb Haemost ; 2013 Apr 10.
Article in English | MEDLINE | ID: mdl-23574625

ABSTRACT

Light transmission aggregometry (LTA) is the most common method used to assess platelet function. However, there is no universal standard for its performance. The Platelet Physiology Subcommittee of the Scientific and Standardization Committee (SSC) of the International Society on Thrombosis and Haemostasis formed a working party of experts with the aim of producing a series of consensus recommendations for standardizing LTA. Due to a lack of investigations that directly compared different methodologies to perform LTA studies, there were insufficient data to develop evidence-based guidelines. Therefore, the RAND method was used, which obtains a formal consensus among experts about the appropriateness of health care interventions, particularly when scientific evidence is absent, scarce and/or heterogeneous. Using this approach, each expert scored as "appropriate", "uncertain" or "inappropriate" a series of statements about the practice of LTA, which included pre-analytical variables, blood collection, blood processing, methodological details, choice of agonists and the evaluation and reporting of results. After presentation and public discussion at SSC meetings, the assessments were further refined to produce final consensus recommendations. Before delivering the recommendations, a formal literature review was performed using a series of defined search terms about LTA. Of the 1830 potentially relevant studies identified, only 14 publications were considered to be actually relevant for review. Based upon the additional information, 6 consensus statements were slightly modified. The final statements were presented and discussed at the SSC Meeting in Cairo (2010) and formed the basis of a consensus document, which is the subject of the present report. This article is protected by copyright. All rights reserved.

10.
J Thromb Haemost ; 10(12): 2573-80, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23083103

ABSTRACT

BACKGROUND: Diadenosine 5',5'''-P(1),P(4)-tetraphosphate (Ap(4)A), a natural compound stored in platelet dense granules, inhibits ADP-induced platelet aggregation. Ap(4)A inhibits the platelet ADP receptors P2Y(1) and P2Y(12), is a partial agonist of P2Y(12), and is a full agonist of the platelet ATP-gated ion channel P2X1. Modification of the Ap(4)A tetraphosphate backbone enhances inhibition of ADP-induced platelet aggregation. However, the effects of these Ap(4)A analogs on human platelet P2Y(1), P2Y(12) and P2X1 are unclear. OBJECTIVE: To determine the agonist and antagonist activities of diadenosine tetraphosphate analogs towards P2Y(1), P2Y(12), and P2X1. METHODS: We synthesized the following Ap(4)A analogs: P(1),P(4)-dithiotetraphosphate; P(2),P(3)-chloromethylenetetraphosphate; P(1)-thio-P(2),P(3)-chloromethylenetetraphosphate; and P(1),P(4)-dithio-P(2),P(3)-chloromethylenetetraphosphate. We then measured the effects of these analogs on: (i) ADP-induced platelet aggregation; (ii) P2Y(1)-mediated changes in cytosolic Ca(2+); (iii) P2Y(12)-mediated changes in vasodilator-stimulated phosphoprotein phosphorylation; and (iv) P2X1-mediated entry of extracellular Ca(2+). RESULTS: Ap(4)A analogs with modifications in the phosphate backbone inhibited both P2Y(1) and P2Y(12), and showed no agonist activity towards these receptors. The dithio modification increased inhibition of P2Y(1), P2Y(12), and platelet aggregation, whereas the chloromethylene modification increased inhibition of P2Y(12) and platelet aggregation, but decreased P2Y(1) inhibition. Combining the dithio and chloromethylene modifications increased P2Y(1) and P2Y(12) inhibition. As compared with Ap(4)A, each modification decreased agonist activity towards P2X1, and the dual modification completely eliminated P2X1 agonist activity. CONCLUSIONS: As compared with Ap(4)A, tetraphosphate backbone analogs of Ap(4)A have diminished activity towards P2X1 but inhibit both P2Y(1) and P2Y(12) and, with greater potency, inhibit ADP-induced platelet aggregation. Thus, diadenosine tetraphosphate analogs with dual receptor selectivity may have potential as antiplatelet drugs.


Subject(s)
Adenosine Diphosphate/pharmacology , Dinucleoside Phosphates/pharmacology , Platelet Activation/drug effects , Receptors, Purinergic P2Y12/metabolism , Receptors, Purinergic P2Y1/metabolism , Humans , Phosphorylation
11.
J Thromb Haemost ; 9(11): 2302-10, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21920014

ABSTRACT

BACKGROUND: Severe thrombocytopenia is a major risk factor for hemorrhage, but platelet function and bleeding risk at low platelet counts are poorly understood, because of the limitations of platelet function testing at very low platelet counts. OBJECTIVES: To examine and compare platelet function in severely thrombocytopenic patients with acute myeloid leukemia (AML) or myelodysplasia (MDS) with that in patients with immune thrombocytopenia (ITP). METHODS: Whole blood flow cytometric measurement of platelet activation and platelet reactivity to agonists was correlated with the immature platelet fraction (IPF) and bleeding symptoms. RESULTS: Patients with AML/MDS had smaller platelets, lower IPF and substantially lower platelet surface expression of activated glycoprotein (GP)IIb-IIIa and GPIb, both with and without addition of ex vivo ADP or thrombin receptor-activating peptide, than patients with ITP. In both ITP and AML/MDS patients, increased platelet surface GPIb on circulating platelets and expression of activated GPIIb-IIIa and GPIb on ex vivo activated platelets correlated with a higher IPF. Whereas platelet reactivity was higher for AML/MDS patients with bleeding than for those with no bleeding, platelet reactivity was lower for ITP patients with bleeding than for those with no bleeding. CONCLUSIONS: AML/MDS patients have lower in vivo platelet activation and ex vivo platelet reactivity than patients with ITP. The proportion of newly produced platelets correlates with the expression of platelet surface markers of activation. These differences might contribute to differences in bleeding tendency between AML/MDS and ITP patients. This study is the first to define differences in platelet function between AML/MDS patients and ITP patients with equivalent degrees of thrombocytopenia.


Subject(s)
Blood Platelets/physiology , Leukemia, Myeloid, Acute/blood , Myelodysplastic Syndromes/blood , Purpura, Thrombocytopenic, Idiopathic/blood , Aged , Blood Platelets/pathology , Cell Shape , Female , Flow Cytometry , Hemorrhage , Humans , Male , Middle Aged , Platelet Activation , Platelet Membrane Glycoproteins/analysis
12.
J Thromb Haemost ; 9(5): 1020-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21320282

ABSTRACT

BACKGROUND: Thrombocytopenia is frequent among neonates, and 20-25% of affected infants are treated with platelet transfusions. These are frequently given for mild thrombocytopenia (platelets: 50-100 × 10(9) L(-1)), largely because of the known hyporeactivity of neonatal platelets. In tests of primary hemostasis, however, neonates have shorter bleeding and closure times (CTs) than adults. This has been attributed to their higher hematocrits, higher von Willebrand factor (VWF) concentrations, and predominance of longer VWF polymers. OBJECTIVE: To determine whether the 'transfusion' of adult (relatively hyperreactive) platelets into neonatal blood results in a hypercoagulable profile. METHODS: Cord blood (CB) and adult peripheral blood (PB) were separated (with a modified buffy coat method) to generate miniaturized platelet concentrates (PCs) and thrombocytopenic blood. PB-derived and CB-derived PCs (n = 7 per group) were then 'transfused'in vitro into thrombocytopenic CB and PB. The effects of autologous vs. allogeneic (developmentally mismatched) 'transfusions' were evaluated with whole blood aggregometry, a platelet function analyzer (PFA-100), and thromboelastography (TEG). RESULTS: Adult platelets aggregated significantly better than neonatal platelets in response to thrombin receptor-activating peptide, ADP, and collagen, regardless of the blood into which they were transfused. The 'transfusion' of adult platelets into thrombocytopenic CB resulted in shorter CTs-EPI (PFA-100) and higher clot strength and firmness (TEG) than 'transfusion' of neonatal autologous platelets. CONCLUSIONS: In vitro'transfusion' of adult platelets into neonatal blood results in shorter CTs than 'transfusion' with neonatal platelets. Our findings should raise awareness of the differences between the neonatal and adult hemostatic system and the potential 'developmental mismatch' associated with platelet transfusions for neonatal hemostasis.


Subject(s)
Platelet Transfusion/methods , Thrombocytopenia/therapy , Adult , Bleeding Time , Blood Coagulation , Blood Platelets/cytology , Fetal Blood/cytology , Hematocrit , Hemostasis , Humans , In Vitro Techniques , Infant, Newborn , Platelet Aggregation , Platelet Count , Platelet Function Tests , Thrombelastography/methods
13.
J Thromb Haemost ; 8(2): 331-40, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19922435

ABSTRACT

BACKGROUND: Release of serotonin and activation of serotonin 5HT2A receptors on platelet surfaces is a potent augmentative stimulus for platelet aggregation. However, earlier-generation serotonin receptor antagonists were not successfully exploited as antiplatelet agents, possibly owing to their lack of specificity for the 5HT2A receptor subtype. OBJECTIVE: To assess whether targeted inhibition of the serotonin 5HT2A receptor attenuates recurrent thrombosis and improves coronary patency in an in vivo canine model mimicking unstable angina. METHODS: In protocol 1, anesthetized dogs were pretreated with a novel, selective inverse agonist of the 5HT2A receptor (APD791) or saline. Recurrent coronary thrombosis was then initiated by coronary artery injury+stenosis, and coronary patency was monitored for 3 h. Protocol 2 was similar, except that: (i) treatment with APD791 or saline was begun 1 h after the onset of recurrent thrombosis; (ii) template bleeding time was measured; and (iii) blood samples were obtained for in vitro flow cytometric assessment of platelet responsiveness to serotonin. RESULTS: APD791 attenuated recurrent thrombosis, irrespective of the time of treatment: in both protocols, flow-time area (index of coronary patency; normalized to baseline coronary flow) averaged 58-59% (P<0.01) following administration of APD791 vs. 21-28% in saline controls. Moreover, the in vivo antithrombotic effect of APD791 was not accompanied by increased bleeding, but was associated with significant and selective inhibition of serotonin-mediated platelet activation. CONCLUSION: 5HT2A receptor inhibition with APD791, even when initiated after the onset of recurrent thrombosis, improves coronary patency in the in vivo canine model.


Subject(s)
Benzamides/pharmacology , Blood Platelets/drug effects , Coronary Thrombosis/drug therapy , Fibrinolytic Agents/pharmacology , Morpholines/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Pyrazoles/pharmacology , Serotonin 5-HT2 Receptor Antagonists , Serotonin Antagonists/pharmacology , Vascular Patency/drug effects , Animals , Benzamides/blood , Benzamides/toxicity , Blood Platelets/metabolism , Coronary Circulation/drug effects , Coronary Thrombosis/blood , Coronary Thrombosis/pathology , Coronary Thrombosis/physiopathology , Disease Models, Animal , Dogs , Drug Inverse Agonism , Fibrinolytic Agents/blood , Fibrinolytic Agents/toxicity , Hemodynamics/drug effects , Hemorrhage/chemically induced , Morpholines/blood , Morpholines/toxicity , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/toxicity , Pyrazoles/blood , Pyrazoles/toxicity , Receptor, Serotonin, 5-HT2A/blood , Recurrence , Serotonin Antagonists/blood , Serotonin Antagonists/toxicity , Time Factors
14.
J Thromb Haemost ; 7(6): 1029, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19422455

ABSTRACT

BACKGROUND: Light transmission aggregometry (LTA) is the most common method used in clinical and research laboratories to assess platelet function. However, the method has never been standardized. OBJECTIVES: As the first step towards development of methodological guidelines, the Platelet Physiology Subcommittee of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis (ISTH) undertook a large, detailed, global survey of LTA practices. METHODS: Members of ISTH and of External Quality Assurance in Thrombosis and Haemostasis organizations were invited to complete a 129 item, online questionnaire. Results were analyzed anonymously to participant identities. RESULTS: The online supplement for this article (http://www.isth.org/Publications/OfficialCommunications/PlateletPhysiology/LightTransmissionAggregometry/tabid/201/Default.aspx) contains the full details of the study findings. 359 (244 clinical, 115 research) laboratories from 48 countries participated in the survey. LTA was widely used to assess inherited or acquired bleeding disorders. Common practices were identified in sample collection, processing and analysis and although some are generally considered acceptable, others are not ideal. The agonist concentrations used for LTA varied, and many laboratories used ADP, collagen, epinephrine and Ristocetin, at more than one concentration, in addition to arachidonic acid. The parameters commonly used to assess LTA responses were maximal amplitude or % aggregation, which was considered particularly important, in addition to the presence of a 'secondary wave', deaggregation, shape change and a measure of the lag phase. However, many laboratories did not have appropriate reference intervals. CONCLUSIONS: This is the largest and most detailed survey of LTA practices ever undertaken. It shows a very high variability in LTA practices worldwide, and, as a consequence, methodological standardization is necessary. The information gathered in this survey will be helpful in the development of ISTH methodological guidelines for LTA.


Subject(s)
Blood Platelets/cytology , Data Collection , Humans
15.
J Thromb Haemost ; 6(12): 2035-44, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18983514

ABSTRACT

BACKGROUND: Aspirin 'resistance' is a widely used term for hyporesponsiveness to aspirin in a platelet function test. Serum thromboxane (TX) B(2) is the most specific test of aspirin's effect on platelets. OBJECTIVES: (i) To examine the role of pre-existent platelet hyperreactivity in aspirin 'resistance'. (ii) To determine the correlation between aspirin resistance defined by serum TXB(2) and other assays of platelet function. METHODS: To enable pre-aspirin samples to be drawn, platelet function was measured in normal subjects (n = 165) before and after aspirin 81 mg daily for seven days. RESULTS: The proportion of the post-aspirin platelet function predicted by the pre-aspirin platelet function was 28.3 +/- 7.5% (mean +/- asymptotic standard error) for serum TXB(2), 39.3 +/- 6.8% for urinary 11-dehydro TXB(2), 4.4 +/- 7.7% for arachidonic acid-induced platelet aggregation, 40.4 +/- 7.1% for adenosine diphosphate-induced platelet aggregation, 26.3 +/- 9.2% for the VerifyNow Aspirin Assay, and 45.0 +/- 10.9% for the TEG PlateletMapping System with arachidonic acid. There was poor agreement between aspirin-resistant subjects identified by serum TXB(2) vs. aspirin-resistant subjects identified by the other five assays, irrespective of whether the analysis was based on categorical or continuous variables. Platelet count correlated with pre-aspirin serum TXB(2) and VerifyNow Aspirin Assay, but not with any post-aspirin platelet function test. CONCLUSIONS: (i) Aspirin 'resistance' (i.e. hyporesponsiveness to aspirin in a laboratory test) is in part unrelated to aspirin but is the result of underlying platelet hyperreactivity prior to the institution of aspirin therapy. (ii) Aspirin resistance defined by serum TXB(2) shows a poor correlation with aspirin resistance defined by other commonly used assays.


Subject(s)
Aspirin/pharmacokinetics , Drug Resistance , Platelet Activation , Platelet Function Tests/standards , Adult , Cyclooxygenase Inhibitors/pharmacology , Female , Humans , Male , Platelet Function Tests/methods , Sensitivity and Specificity , Thromboxane A2/pharmacology , Young Adult
16.
J Thromb Haemost ; 6(2): 359-65, 2008 02.
Article in English | MEDLINE | ID: mdl-18021304

ABSTRACT

BACKGROUND: Prasugrel is a novel antiplatelet prodrug of the same thienopyridine class as clopidogrel and ticlopidine. Metabolism of prasugrel generates the active metabolite R-138727, an antagonist of the platelet P2Y(12) adenosine diphosphate (ADP) receptor, leading to inhibition of ADP-mediated platelet activation and aggregation. ADP also enhances the platelet response to collagen, and these two agonists contribute to the generation of platelet procoagulant activity. We therefore examined whether R-138727 inhibits ADP- and collagen-triggered platelet procoagulant activities. METHODS AND RESULTS: As shown by whole blood flow cytometry, R-138727 inhibited surface phosphatidylserine expression on ADP plus collagen-stimulated platelets and tissue factor (TF) expression on ADP-, collagen-, and ADP plus collagen-stimulated monocyte-platelet aggregates. R-138727 reduced monocyte-platelet aggregate formation, thereby further inhibiting TF expression. ADP, collagen, and ADP plus collagen accelerated the kinetics of thrombin generation in recalcified whole blood and R-138727 significantly inhibited this acceleration. Clot strength in a modified thromboelastograph system was also inhibited by R-138727 (IC50 0.7 +/- 0.1 microM). CONCLUSIONS: In addition to its previously known inhibitory effects on platelet activation and aggregation, the active metabolite of prasugrel, R-138727, inhibits platelet procoagulant activity in whole blood (as determined by phosphatidylserine expression on platelets and TF expression on monocyte-platelet aggregates), resulting in the functional consequences of delayed thrombin generation and impaired clot development.


Subject(s)
Blood Coagulation Factors/antagonists & inhibitors , Blood Platelets/drug effects , Clot Retraction/drug effects , Piperazines/pharmacology , Piperazines/pharmacokinetics , Platelet Activation/drug effects , Prodrugs/pharmacokinetics , Purinergic P2 Receptor Antagonists , Thiophenes/pharmacokinetics , Adenosine Diphosphate/pharmacology , Annexin A5/metabolism , Biotransformation , Collagen/pharmacology , Humans , Monocytes/metabolism , Phosphatidylserines/blood , Prasugrel Hydrochloride , Receptors, Purinergic P2Y12 , Thrombelastography , Thrombin/biosynthesis , Thromboplastin/biosynthesis
17.
J Thromb Haemost ; 5(4): 761-5, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17371489

ABSTRACT

AIM: To determine whether indices of platelet activation are associated with the stability of coronary artery disease (CAD). METHODS: Platelet function was examined in 677 consecutive aspirin-treated patients presenting for cardiac catheterization. Patients were grouped into recent myocardial infarction (MI), no MI but angiographically documented CAD (non-MI CAD) and no angiographically detectible CAD (no CAD), as well as additional subgroups. RESULTS: Compared with non-MI CAD or no CAD patients, more patients with recent MI had a shortened platelet function analyzer (PFA)-100 collagen-epinephrine closure time (CT) and increased circulating monocyte-platelet aggregates, neutrophil-platelet aggregates, activated platelet surface GPIIb-IIIa and plasma soluble CD40 ligand (sCD40L). More patients with non-MI CAD had shortened PFA-100 CTs and increased monocyte-platelet aggregates compared with patients with no CAD. Platelet surface P-selectin did not differ among the groups. Subgroup analysis revealed that decreasing PFA-100 CT correlated with the stability of CAD. CONCLUSIONS: Indices of platelet activation, especially the PFA-100 CT, are associated with the stability of CAD, and may reflect plaque instability, an ongoing thrombotic state and/or reduced responsiveness to aspirin.


Subject(s)
Coronary Artery Disease/blood , Coronary Artery Disease/therapy , Platelet Activation , Aged , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Aspirin/pharmacology , CD40 Ligand/metabolism , Coronary Artery Disease/diagnosis , Epinephrine/metabolism , Female , Humans , Male , Middle Aged , Neutrophils/metabolism , P-Selectin/biosynthesis , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
18.
J Thromb Haemost ; 5(1): 75-81, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17002661

ABSTRACT

BACKGROUND: Clopidogrel is a widely used antithrombotic agent that inhibits the platelet P2Y(12) adenosine diphosphate (ADP) receptor. There is increasing interest in 'clopidogrel resistance'. OBJECTIVES: To determine whether 'clopidogrel resistance' is accounted for by a pre-existent variability in platelet response to ADP. METHODS: Platelet response to 20 microm ADP was analyzed by four independent whole blood flow cytometric assays: platelet surface activated GPIIb-IIIa, platelet surface P-selectin, monocyte-platelet aggregates and neutrophil-platelet aggregates. In 25 consecutive, non-aspirin-treated healthy subjects, we studied platelet response before and after clopidogrel administration. In addition, we studied the platelet response in 613 consecutive aspirinated patients with or without coronary artery disease (CAD, as determined by angiography) who had or had not been treated with clopidogrel. In these patients, we tested for homogeneity of variance across all durations of clopidogrel exposure and severity of CAD by estimating the 'goodness of fit' of two independent models. RESULTS: In the healthy subjects, pre-clopidogrel response to ADP predicted post-clopidogrel response to ADP. In the patients, clopidogrel, as expected, inhibited the platelet response to ADP. However, irrespective of the duration of clopidogrel administration, the severity of CAD, and the dose of aspirin, clopidogrel did not increase the variance in the platelet response to ADP in any of the four assays of platelet response. CONCLUSIONS: These studies provide evidence that 'clopidogrel resistance' is accounted for by a pre-existent variability in platelet response to ADP and this variability is not increased by clopidogrel administration.


Subject(s)
Adenosine Diphosphate/pharmacology , Blood Platelets/drug effects , Drug Resistance , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Ticlopidine/analogs & derivatives , Adult , Aspirin/pharmacology , Bayes Theorem , Clopidogrel , Coronary Artery Disease/drug therapy , Coronary Artery Disease/physiopathology , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Models, Cardiovascular , Platelet Aggregation Inhibitors/administration & dosage , Platelet Function Tests , Predictive Value of Tests , Reference Values , Severity of Illness Index , Ticlopidine/administration & dosage , Ticlopidine/pharmacology , Time Factors
19.
J Thromb Haemost ; 4(12): 2670-7, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16995902

ABSTRACT

BACKGROUND: Previous studies have shown that ischemic preconditioning (PC) not only limits infarct size, but also improves arterial patency in models of recurrent thrombosis. We hypothesize that this enhanced patency is presumably because of a PC-induced attenuation of platelet-mediated thrombosis. However, there is, at present, no direct evidence that PC acts on the platelets per se and favorably down-regulates platelet reactivity. OBJECTIVES: Our goal was to test the concept that PC ischemia attenuates molecular indices of platelet activation-aggregation. METHODS: Anesthetized dogs were randomly assigned to receive 10 min of PC ischemia followed by 10 min of reperfusion or a time-matched control period. Spontaneous recurrent coronary thrombosis was then initiated in all dogs by injury + stenosis of the left anterior descending coronary artery. Coronary flow was monitored for 3 h poststenosis, and molecular indices of platelet activation-aggregation were quantified by whole blood flow cytometry. RESULTS: Coronary patency was, as expected, better-maintained following injury + stenosis in the PC group vs. controls (53% +/- 5%* vs. 23% +/- 5% of baseline flow, respectively; *P < 0.05). Moreover, PC was accompanied by: (i) a significant down-regulation of platelet-fibrinogen binding and formation of neutrophil-platelet aggregates (112% +/- 14%* vs. 177% +/- 21% and 107% +/- 8%* vs. 155% +/- 19% of baseline values in PC vs. control groups); and (ii) a trend towards a reduction in platelet P-selectin expression (148% +/- 12% vs. 190% +/- 21% of baseline; *P < 0.05 and P = 0.09 vs. control). CONCLUSION: These data provide novel, direct evidence in support of the concept that ischemic PC attenuates molecular indices of platelet activation-aggregation.


Subject(s)
Coronary Thrombosis/blood , Ischemic Preconditioning, Myocardial , Myocardial Reperfusion Injury/blood , Platelet Activation , Animals , Blood Flow Velocity , Blood Platelets/metabolism , Coronary Circulation , Coronary Thrombosis/pathology , Coronary Thrombosis/physiopathology , Coronary Thrombosis/prevention & control , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Disease Models, Animal , Dogs , Fibrinogen/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , P-Selectin/metabolism , Platelet Adhesiveness , Platelet Aggregation , Random Allocation , Vascular Patency
20.
J Thromb Haemost ; 3(11): 2563-70, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16241954

ABSTRACT

BACKGROUND: Monocytes and neutrophils form heterotypic aggregates with platelets initially via engagement of platelet surface P-selectin with leukocyte surface P-selectin glycoprotein ligand-1 (PSGL-1). The resultant intracellular signaling causes the leukocyte surface expression of tissue factor and activation of leukocyte surface Mac-1 (integrin alphaMbeta2, CD11b/CD18). The activation-dependent conformational change in monocyte surface Mac-1 results in the binding of coagulation factor Xa (FXa) and/or fibrinogen to Mac-1. The aim of this study was to develop whole blood flow cytometry assays of these procoagulant activities and to investigate the effects of platelet binding to monocytes and neutrophils. METHODS: Citrate or D-Phe-Pro-Arg-chloromethylketone (PPACK) anticoagulated whole blood was incubated with monoclonal antibodies against CD14 (PECy5), CD42a (PE), FITC-conjugated test antibody and an agonist, and then fixed with FACS lyse. Appropriate isotype negative controls were prepared in parallel. A BD FACSCalibur was used to analyze monocytes and neutrophils, which were identified based on CD14 fluorescence, forward and 90 degrees light scatter. These populations were further gated into CD42a-positive (platelet-bound) and CD42a-negative (platelet-free). Geometric mean fluorescence and per cent positive data were collected for each subpopulation to measure the binding of test antibodies directed at CD42a, tissue factor, coagulation FXa, bound fibrinogen, activated Mac-1, and CD11b. Compensation controls were prepared on six normal donors prior to the study and these settings were used throughout the 10 donor study. Negative controls verified the lack of cross talk, particularly in the quantified FITC and PE parameters. RESULTS: The physiologic agonists collagen and ADP increased monocyte-platelet and neutrophil-platelet aggregates and increased leukocyte surface Mac-1/CD11b and surface-bound tissue factor, FXa and fibrinogen. Whereas the increases in Mac-1/CD11b were mainly independent of leukocyte-platelet binding, the increases in surface-bound tissue factor, FXa and fibrinogen were mainly dependent on leukocyte-platelet binding. CONCLUSIONS: (i) We have developed novel whole blood flow cytometry assays to measure bound tissue factor, coagulation FXa, fibrinogen, activated Mac-1 and CD11b on the surface of monocytes and neutrophils, allowing independent analysis of monocytes and neutrophils with and without surface-adherent platelets. (ii) The monocyte and neutrophil surface binding of tissue factor, FXa and fibrinogen is mainly dependent on platelet adherence to monocytes and neutrophils, whereas the monocyte and neutrophil surface expression of CD11b and activated Mac-1 is mainly independent of platelet adherence to monocytes and neutrophils.


Subject(s)
Blood Coagulation/physiology , Blood Platelets/metabolism , Cell Separation , Flow Cytometry , Monocytes/metabolism , Neutrophils/metabolism , Adenosine Diphosphate/pharmacology , Adult , Blood Platelets/drug effects , Blood Platelets/immunology , CD11b Antigen/analysis , Cell Aggregation/physiology , Cell Communication/physiology , Collagen/pharmacology , Factor Xa/metabolism , Female , Fibrinogen/metabolism , Humans , Macrophage-1 Antigen/analysis , Male , Middle Aged , Monocytes/drug effects , Monocytes/immunology , Neutrophils/drug effects , Neutrophils/immunology , Platelet Activation/drug effects , Platelet Glycoprotein GPIb-IX Complex/analysis , Thromboplastin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...