Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 139
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 55(2): 224-236.e5, 2022 02 08.
Article in English | MEDLINE | ID: mdl-34995475

ABSTRACT

During gram-negative septicemia, interactions between platelets and neutrophils initiate a detrimental feedback loop that sustains neutrophil extracellular trap (NET) induction, disseminated intravascular coagulation, and inflammation. Understanding intracellular pathways that control platelet-neutrophil interactions is essential for identifying new therapeutic targets. Here, we found that thrombin signaling induced activation of the transcription factor NFAT in platelets. Using genetic and pharmacologic approaches, as well as iNFATuation, a newly developed mouse model in which NFAT activation can be abrogated in a cell-specific manner, we demonstrated that NFAT inhibition in activated murine and human platelets enhanced their activation and aggregation, as well as their interactions with neutrophils and NET induction. During gram-negative septicemia, NFAT inhibition in platelets promoted disease severity by increasing disseminated coagulation and NETosis. NFAT inhibition also partially restored coagulation ex vivo in patients with hypoactive platelets. Our results define non-transcriptional roles for NFAT that could be harnessed to address pressing clinical needs.


Subject(s)
Blood Platelets/drug effects , NFATC Transcription Factors/antagonists & inhibitors , Platelet Aggregation/drug effects , Sepsis/pathology , Animals , Blood Coagulation/drug effects , Blood Platelets/metabolism , Cell Communication/drug effects , Cytoplasmic Granules/metabolism , Disease Models, Animal , Extracellular Traps/metabolism , Humans , Inflammation , Mice , NFATC Transcription Factors/metabolism , Neutrophils/metabolism , Receptors, Thrombin/metabolism , Sepsis/metabolism
2.
Semin Thromb Hemost ; 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38653463

ABSTRACT

Platelets are the smallest blood cells, numbering 150 to 350 × 109/L in healthy individuals. The ability of activated platelets to adhere to an injured vessel wall and form aggregates was first described in the 19th century. Besides their long-established roles in thrombosis and hemostasis, platelets are increasingly recognized as pivotal players in numerous other pathophysiological processes including inflammation and atherogenesis, antimicrobial host defense, and tumor growth and metastasis. Consequently, profound knowledge of platelet structure and function is becoming more important in research and in many fields of modern medicine. This review provides an overview of platelet physiology focusing particularly on the structure, granules, surface glycoproteins, and activation pathways of platelets.

3.
Blood ; 140(13): 1470-1481, 2022 09 29.
Article in English | MEDLINE | ID: mdl-35849650

ABSTRACT

The phase 3 HESTIA3 study assessed the efficacy and safety of the reversible P2Y12 inhibitor ticagrelor vs placebo in preventing vaso-occlusive crises in pediatric patients with sickle cell disease (SCD). Patients aged 2 to 17 years were randomly assigned 1:1 to receive weight-based doses of ticagrelor or matching placebo. The primary end point was the rate of vaso-occlusive crises, a composite of painful crises and/or acute chest syndrome (ACS). Key secondary end points included number and duration of painful crises, number of ACS events, and number of vaso-occlusive crises requiring hospitalization or emergency department visits. Exploratory end points included the effect of ticagrelor on platelet activation. In total, 193 patients (ticagrelor, n = 101; placebo, n = 92) underwent randomization at 53 sites across 16 countries. The study was terminated 4 months before planned completion for lack of efficacy. Median ticagrelor exposure duration was 296.5 days. The primary end point was not met: estimated yearly incidence of vaso-occlusive crises was 2.74 in the ticagrelor group and 2.60 in the placebo group (rate ratio, 1.06; 95% confidence interval, 0.75-1.50; P = .7597). There was no evidence of efficacy for ticagrelor vs placebo across secondary end points. Median platelet inhibition with ticagrelor at 6 months was 34.9% predose and 55.7% at 2 hours' postdose. Nine patients (9%) in the ticagrelor group and eight patients (9%) in the placebo group had at least one bleeding event. In conclusion, no reduction of vaso-occlusive crises was seen with ticagrelor vs placebo in these pediatric patients with SCD. This trial was registered at www.clinicaltrials.gov as #NCT03615924.


Subject(s)
Acute Chest Syndrome , Anemia, Sickle Cell , Acute Chest Syndrome/drug therapy , Acute Chest Syndrome/etiology , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Child , Hemorrhage/drug therapy , Humans , Pain/drug therapy , Platelet Aggregation Inhibitors/adverse effects , Ticagrelor/therapeutic use
4.
Blood ; 137(15): 2010-2020, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33067606

ABSTRACT

Clinical trials in sickle cell disease (SCD) often focus on health care utilization for painful vaso-occlusive crises (VOCs). However, no objective, quantifiable pain biomarkers exist, pain is not specific to VOCs, health care utilization varies between patients, unreported at-home VOCs likely contribute to long-term outcomes, and patient-reported outcomes are seldom considered. This noninterventional, longitudinal, 6-month study aimed to develop tools to identify VOCs in SCD patients with or without health care utilization. Participants wore an actigraph device, tracking sleep and activity. Patients with SCD used an electronic patient-reported outcome (ePRO) tool to collect data on pain, medication, fatigue, and daily function. Patients self-reported when they experienced VOC pain (VOC day). Biomarkers were collected every 3 weeks (non-VOC). Self-reported VOCs triggered at-home or in-hospital blood collection. The study enrolled 37 participants with SCD; 35 completed the study. Participants reported 114 VOC events and 346 VOC days, of which 62.3% and 78.3%, respectively, were self-treated at home. The ePRO and actigraphy captured end points of pain, functionality, fatigue, activity, and sleep; each was significantly altered on VOC days compared with non-VOC days. Biomarkers collected at home or in the hospital on VOC days were significantly altered compared with non-VOC baseline values, including leukocyte-platelet aggregates, microfluidic-based blood cell adhesion, interleukin-6, C-reactive protein, interleukin-10, tumor necrosis factor-α, and thrombin-antithrombin. The Evaluation of Longitudinal Pain Study in Sickle Cell Disease (ELIPSIS) trial shows the feasibility of accurately monitoring out-of-hospital pain by using patient-reported VOC days as potential end points for clinical trials in SCD; it describes the changes in biomarkers and activity measured by actigraphy that may enable improved identification and assessment of VOCs.


Subject(s)
Anemia, Sickle Cell/complications , Pain/etiology , Actigraphy , Adolescent , Adult , Anemia, Sickle Cell/diagnosis , Anemia, Sickle Cell/drug therapy , Antisickling Agents/therapeutic use , Biomarkers/analysis , Female , Humans , Hydroxyurea/therapeutic use , Longitudinal Studies , Male , Middle Aged , Pain/diagnosis , Patient Reported Outcome Measures , Young Adult
5.
Blood ; 136(17): 1956-1967, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32693407

ABSTRACT

Gray platelet syndrome (GPS) is a rare recessive disorder caused by biallelic variants in NBEAL2 and characterized by bleeding symptoms, the absence of platelet α-granules, splenomegaly, and bone marrow (BM) fibrosis. Due to the rarity of GPS, it has been difficult to fully understand the pathogenic processes that lead to these clinical sequelae. To discern the spectrum of pathologic features, we performed a detailed clinical genotypic and phenotypic study of 47 patients with GPS and identified 32 new etiologic variants in NBEAL2. The GPS patient cohort exhibited known phenotypes, including macrothrombocytopenia, BM fibrosis, megakaryocyte emperipolesis of neutrophils, splenomegaly, and elevated serum vitamin B12 levels. Novel clinical phenotypes were also observed, including reduced leukocyte counts and increased presence of autoimmune disease and positive autoantibodies. There were widespread differences in the transcriptome and proteome of GPS platelets, neutrophils, monocytes, and CD4 lymphocytes. Proteins less abundant in these cells were enriched for constituents of granules, supporting a role for Nbeal2 in the function of these organelles across a wide range of blood cells. Proteomic analysis of GPS plasma showed increased levels of proteins associated with inflammation and immune response. One-quarter of plasma proteins increased in GPS are known to be synthesized outside of hematopoietic cells, predominantly in the liver. In summary, our data show that, in addition to the well-described platelet defects in GPS, there are immune defects. The abnormal immune cells may be the drivers of systemic abnormalities such as autoimmune disease.


Subject(s)
Cytoplasmic Granules/pathology , Genetic Heterogeneity , Gray Platelet Syndrome , Immune System/pathology , Phenotype , Biopsy , Blood Proteins/genetics , Case-Control Studies , Cohort Studies , Cytoplasmic Granules/metabolism , Diagnosis, Differential , Gene Frequency , Genetic Association Studies , Gray Platelet Syndrome/classification , Gray Platelet Syndrome/genetics , Gray Platelet Syndrome/immunology , Gray Platelet Syndrome/pathology , Humans , Immune System/physiology , Immune System Diseases/blood , Immune System Diseases/diagnosis , Immune System Diseases/genetics , Immune System Diseases/pathology , Mutation
6.
Platelets ; 33(1): 116-122, 2022 Jan 02.
Article in English | MEDLINE | ID: mdl-33284729

ABSTRACT

Our objective was to characterize platelet surface glycoprotein (GP)Ibα, activated GPIIb-IIIa, and P-selectin levels during and after extracorporeal membrane oxygenation (ECMO). We performed a single center cohort study of 10 adult patients on ECMO for cardiogenic shock. Patients had blood samples drawn on ECMO day 1 or 2, day 3, day 5, and 48-72 hours after ECMO decannulation. Platelets from untreated blood samples and samples treated with either adenosine diphosphate (ADP) or thrombin receptor agonist peptide (TRAP) had surface GPIbα, activated GPIIb-IIIa, and P-selectin levels measured using flow cytometry. Platelet surface GPIbα levels varied significantly by time on ECMO (p = .002) and were significantly higher on ECMO day 5 compared to ECMO day 1 (p = .01). GPIbα levels during ECMO did not differ significantly from levels after ECMO decannulation (p = .14). Activated GPIIb-IIIa levels did not change significantly during ECMO, but were significantly higher after ECMO decannulation (p = .04). There were no significant differences in P-selectin levels during ECMO (p = .87) or after ECMO decannulation (p = .41). Platelet surface GPIbα and P-selectin levels were similar during and after ECMO whereas activated GPIIb-IIIa levels were lower during ECMO, particularly in response to TRAP stimulation, potentially contributing to ECMO-induced coagulopathy.


Subject(s)
Extracorporeal Membrane Oxygenation/methods , P-Selectin/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Humans
7.
Cytometry A ; 99(2): 170-179, 2021 02.
Article in English | MEDLINE | ID: mdl-33399275

ABSTRACT

Platelets mediate key biological processes, including hemostasis, immunity, and inflammation. Although platelets are often treated as a homogeneous cell population, they are known to be heterogeneous in size, age, surface receptor expression, and response to agonist stimulation, raising the possibility that distinct platelet subsets perform specialized functions and that such subsets may be altered in disease settings. Attempts to identify platelet subsets by flow cytometry have had limited success due in part to limits on the number of probes that can be used at the same time and due to the challenges of compensating for probes that have large spectral overlap. We recently reported a method to identify platelet subsets by mass cytometry using a panel of 14 metal-tagged antibodies directed at platelet surface markers. Here, we describe the technical considerations and best practices for platelet sample preparation, processing, and analysis by mass cytometry. Specifically, we show that anticoagulant choice alters platelet phenotype and function and that antibody cocktail storage and sample processing are critical for reproducibility. Additionally, we optimize sample density and instrument setup for maximal platelet transmission. Lastly, we demonstrate the importance of panel design and compensation and the use of clustering and dimension reduction to map platelet heterogeneity across resting and stimulated samples.


Subject(s)
Blood Platelets , Hemostasis , Flow Cytometry , Humans , Indicators and Reagents , Platelet Activation , Reproducibility of Results
8.
J Thromb Thrombolysis ; 51(3): 675-681, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32683645

ABSTRACT

Prolonged use of dual antiplatelet therapy (DAPT) post-percutaneous coronary intervention (PCI) has been shown to reduce the risk of major adverse cardiovascular events (MACE), but with increased bleeding. It remains unknown whether biomarkers of platelet activation may be useful for identifying patients at increased risk of MACE. The DAPT study was a randomized trial of 12 versus 30 months of DAPT in patients who underwent PCI. Serum biomarkers [myeloid-related protein (MRP)-8/14, P-selectin, soluble CD-40 ligand (sCD40L)] were assessed in 1399 patients early post-PCI. On-treatment platelet reactivity index (PRI) using VASP phosphorylation was assessed in 443 patients randomized to continued DAPT at 1 year. MACE was defined as CV death, MI, or ischemic stroke. Multivariable models were adjusted for baseline characteristics, index event, and stent type. A stepwise increase in the risk of MACE was observed with increasing tertiles of both MRP-8/14 and P-selectin (p-trend = 0.04 for both). After multivariable adjustment, the adjusted HR (95% CI) for MACE in patients in the top tertile was 1.94 (1.14-3.30) for MRP-8/14 and 1.62 (0.99-2.64) for P-selectin. In contrast, baseline sCD40L was not associated with CV risk. Among patients randomized to continued DAPT, higher on-treatment platelet reactivity was not significantly associated with risk of MACE (p-trend = 0.32; adj-HR T3 vs. T1 1.54, 95% CI 0.20-12.18) or bleeding (P-trend = 0.17; adj-HR 0.25, 95% CI 0.05-1.21). MRP-8/14 and soluble P-selectin may be useful for identifying patients at increased risk of MACE after PCI. The utility of on-treatment platelet function testing requires further study.Clinical Trial Registration https://www.clinicaltrials.gov . Unique identifier NCT00977938.


Subject(s)
Calgranulin A/blood , Calgranulin B/blood , Coronary Restenosis , Hemorrhage , P-Selectin/blood , Percutaneous Coronary Intervention/adverse effects , Biomarkers/blood , CD40 Ligand/blood , Coronary Restenosis/blood , Coronary Restenosis/etiology , Coronary Restenosis/prevention & control , Drug Monitoring/methods , Dual Anti-Platelet Therapy/adverse effects , Dual Anti-Platelet Therapy/methods , Duration of Therapy , Female , Hemorrhage/blood , Hemorrhage/chemically induced , Hemorrhage/prevention & control , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/instrumentation , Percutaneous Coronary Intervention/methods , Platelet Function Tests/methods , Reproducibility of Results , Risk Assessment/methods
9.
Diabetologia ; 63(4): 733-743, 2020 04.
Article in English | MEDLINE | ID: mdl-31858186

ABSTRACT

AIMS/HYPOTHESIS: Praliciguat (IW-1973), a soluble guanylate cyclase stimulator, amplifies nitric oxide signalling. This exploratory trial investigated the safety, tolerability, pharmacokinetic profile and pharmacodynamic effects of praliciguat in individuals with type 2 diabetes and hypertension. METHODS: This Phase IIA, double-blind, placebo-controlled trial investigated praliciguat in 26 participants with type 2 diabetes and hypertension on stable glucose- and BP-lowering therapies. Participants were randomly allocated in a 3:5:5 ratio to three groups: placebo (n = 6), praliciguat 40 mg once daily for days 1-14 (n = 10), or praliciguat 20 mg twice daily for days 1-7 then 40 mg once daily for days 8-14 (n = 10). Assessments were made in clinic and included treatment-emergent adverse events, pharmacokinetics, metabolic variables, 24 h BP and heart rate, platelet function, reactive hyperaemia index (RHI) and plasma biomarkers. Participants, the sponsor, the investigator and clinic study staff (except designated pharmacy personnel) were blinded to group assignment. RESULTS: Participants treated for 14 days with praliciguat had least-square mean change-from-baseline differences vs placebo (95% CI) of -0.7 (-1.8, 0.4) mmol/l for fasting plasma glucose, -0.7 (-1.1, -0.2) mmol/l for total cholesterol, -0.5 (-1.0, -0.1) mmol/l for LDL-cholesterol, -23 (-56, 9) for HOMA-IR in those not being treated with insulin, and -5 (-10, 1) mmHg and 3 (-1, 6) beats/min for average 24 h mean arterial pressure and heart rate, respectively. Apart from one serious adverse event (SAE; upper gastrointestinal haemorrhage), praliciguat was well tolerated. Praliciguat did not affect platelet function or RHI. Among exploratory biomarkers, plasma levels of asymmetric dimethylarginine decreased in praliciguat vs placebo recipients. CONCLUSIONS/INTERPRETATION: In participants with type 2 diabetes and hypertension on standard therapies, over 14 days praliciguat was well tolerated, except for a single SAE, and showed positive trends in metabolic and BP variables. These results support further clinical investigation of praliciguat. TRIAL REGISTRATION: ClinicalTrials.gov NCT03091920. FUNDING: This trial was funded by Cyclerion Therapeutics.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypertension/drug therapy , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Adult , Aged , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/prevention & control , Double-Blind Method , Drug Therapy, Combination , Female , Guanylyl Cyclase C Agonists/pharmacokinetics , Guanylyl Cyclase C Agonists/therapeutic use , Humans , Hypertension/complications , Hypertension/metabolism , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Insulin/administration & dosage , Insulin/adverse effects , Male , Middle Aged , Treatment Outcome
10.
Br J Haematol ; 188(6): 945-961, 2020 03.
Article in English | MEDLINE | ID: mdl-31823355

ABSTRACT

Acute lymphoblastic leukaemia (ALL) is the most common malignancy in children. Although the survival rate has increased dramatically over the last decades, patients struggle with the adverse side effects of treatment. Treatment for ALL includes chemotherapy and irradiation - both of which are linked to cognitive impairments and alterations in central nervous system (CNS) structure and function detected by neuroimaging and in neurocognitive studies. The present article is a meta-analysis of the existing evidence for the mechanisms underlying changes in the CNS and neurocognitive function in ALL survivors after treatment. We found that compared with controls, ALL survivors develop: (i) cognitive sequelae in intelligence, academics, attention, memory, processing speed and executive function domains; (ii) decreased grey and white matter volume in cortical and several subcortical brain regions, with functional changes particularly in frontal regions and the hippocampus; (iii) neurocognitive impairments related to CNS changes; and (iv) reduction, but not resolution, of late neurocognitive sequelae in patients in whom prophylactic irradiation was replaced by systemic/intrathecal chemotherapy. Continued work with advanced functional magnetic resonance imaging techniques will hopefully allow the detection of early CNS changes as biomarkers to help guide early diagnosis and intervention for neurocognitive defects in patients with childhood ALL.


Subject(s)
Central Nervous System/anatomy & histology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Child , Child, Preschool , Female , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Survival Analysis
11.
Proc Natl Acad Sci U S A ; 114(29): 7695-7700, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28674018

ABSTRACT

Sudden infant death syndrome (SIDS), the leading cause of postneonatal infant mortality, likely comprises heterogeneous disorders with the common phenotype of sudden death without explanation upon postmortem investigation. Previously, we reported that ∼40% of SIDS deaths are associated with abnormalities in serotonin (5-hydroxytryptamine, 5-HT) in regions of the brainstem critical in homeostatic regulation. Here we tested the hypothesis that SIDS is associated with an alteration in serum 5-HT levels. Serum 5-HT, adjusted for postconceptional age, was significantly elevated (95%) in SIDS infants (n = 61) compared with autopsied controls (n = 15) [SIDS, 177.2 ± 15.1 (mean ± SE) ng/mL versus controls, 91.1 ± 30.6 ng/mL] (P = 0.014), as determined by ELISA. This increase was validated using high-performance liquid chromatography. Thirty-one percent (19/61) of SIDS cases had 5-HT levels greater than 2 SDs above the mean of the controls, thus defining a subset of SIDS cases with elevated 5-HT. There was no association between genotypes of the serotonin transporter promoter region polymorphism and serum 5-HT level. This study demonstrates that SIDS is associated with peripheral abnormalities in the 5-HT pathway. High serum 5-HT may serve as a potential forensic biomarker in autopsied infants with SIDS with serotonergic defects.


Subject(s)
Asphyxia/blood , Biomarkers/blood , Serotonin/blood , Sudden Infant Death/blood , Adult , Autopsy , Brain Stem/metabolism , Case-Control Studies , Chromatography, High Pressure Liquid , Cohort Studies , Female , Genotype , Humans , Hydroxyindoleacetic Acid/blood , Infant , Male , Polymorphism, Genetic , Risk Factors , Serotonin Plasma Membrane Transport Proteins/genetics
12.
Am Heart J ; 213: 97-104, 2019 07.
Article in English | MEDLINE | ID: mdl-31132584

ABSTRACT

BACKGROUND: The Fontan procedure is the final step of the 3-stage palliative procedure commonly performed in children with single ventricle physiology. Thrombosis remains an important complication in children after this procedure. To date, guideline recommendations for the type and duration of thromboprophylaxis after Fontan surgery are mainly based on extrapolation of knowledge gained from adults at risk for thrombosis in other clinical settings. Warfarin is being used off-label, and because of its multiple interactions with other drugs and food, a new alternative is highly desirable. Rivaroxaban, a direct Factor Xa inhibitor with a predictable pharmacokinetic profile, is a candidate to address this medical need. STUDY DESIGN: The UNIVERSE study is a prospective, open-label, active-controlled, multicenter study in children 2 to 8 years of age who have single ventricle physiology and had the Fontan procedure within the 4 months preceding enrollment. This study consists of 2 parts. In Part A, rivaroxaban pharmacokinetics, pharmacodynamics, safety, and tolerability are assessed to validate the pediatric dosing selected. In Part B, safety and efficacy of rivaroxaban versus acetylsalicylic acid are evaluated for thromboprophylaxis in children post-Fontan procedure. Children in each part will receive study drug for 12 months. Part A has been completed with 12 children enrolled. Enrollment into Part B is currently ongoing. CONCLUSIONS: The UNIVERSE study aims to provide dosing, pharmacokinetics/pharmacodynamics, safety, and efficacy information on the use of rivaroxaban, an oral anticoagulant, versus acetylsalicylic acid, an antiplatelet agent, in children with single ventricle physiology after the Fontan procedure.


Subject(s)
Aspirin/administration & dosage , Factor Xa Inhibitors/administration & dosage , Fibrinolytic Agents/administration & dosage , Fontan Procedure/adverse effects , Postoperative Complications/prevention & control , Randomized Controlled Trials as Topic , Rivaroxaban/administration & dosage , Thrombosis/prevention & control , Aspirin/pharmacokinetics , Child , Child, Preschool , Factor Xa Inhibitors/pharmacokinetics , Female , Fibrinolytic Agents/pharmacokinetics , Humans , Male , Multicenter Studies as Topic , Prospective Studies , Rivaroxaban/pharmacokinetics
14.
Blood ; 130(6): 713-721, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28600334

ABSTRACT

Antiplatelet therapy is of proven benefit in coronary artery disease and a number of other clinical settings. This article reviews platelet function, molecular targets of antiplatelet agents, and clinical indications for antiplatelet therapy before focusing on a frequent question to hematologists about the 2 most commonly used antiplatelet therapies: Could the patient be aspirin "resistant" or clopidogrel "resistant"? If so, should results of a platelet function test be used to guide the dose or type of antiplatelet therapy? Whether such guided therapy is of clinical benefit to patients has been a source of controversy. The present article reviews this subject in the context of 2 prototypical clinical cases. Available evidence does not support the use of laboratory tests to guide the dose of aspirin or clopidogrel in patients with so-called aspirin or clopidogrel "resistance."


Subject(s)
Aspirin/therapeutic use , Blood Platelets/drug effects , Drug Monitoring/methods , Platelet Aggregation Inhibitors/therapeutic use , Ticlopidine/analogs & derivatives , Aged , Blood Platelets/cytology , Blood Platelets/metabolism , Clopidogrel , Humans , Male , Middle Aged , Molecular Targeted Therapy/methods , Platelet Function Tests/methods , Ticlopidine/therapeutic use
15.
Blood ; 126(7): 873-9, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26138687

ABSTRACT

Immune thrombocytopenia (ITP) patients with similarly low platelet counts differ in their tendency to bleed. To determine if differences in platelet function in ITP patients account for this variation in bleeding tendency, we conducted a single-center, cross-sectional study of pediatric patients with ITP. Bleeding severity (assessed by standardized bleeding score) and platelet function (assessed by whole blood flow cytometry) with and without agonist stimulation was evaluated in 57 ITP patients (median age, 9.9 years). After adjustment for platelet count, higher levels of thrombin receptor activating peptide (TRAP)-stimulated percent P-selectin- and activated glycoprotein (GP)IIb-IIIa-positive platelets were significantly associated with a lower bleeding score, whereas higher levels of immature platelet fraction (IPF), TRAP-stimulated platelet surface CD42b, unstimulated platelet surface P-selectin, and platelet forward light scatter (FSC) were associated with a higher bleeding score. Thus, platelet function tests related to platelet age (IPF, FSC) and activation through the protease activated receptor 1 (PAR1) thrombin receptor (TRAP-stimulated P-selectin, activated GPIIb-IIIa, and CD42b), independent of platelet count, are associated with concurrent bleeding severity in ITP. These tests may be useful markers of future bleeding risk in ITP.


Subject(s)
Hemorrhage/blood , Hemorrhage/etiology , Platelet Count , Platelet Function Tests , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/complications , Adolescent , Blood Platelets/pathology , Blood Platelets/physiology , Cell Differentiation , Cell-Derived Microparticles/physiology , Child , Child, Preschool , Cross-Sectional Studies , Female , Flow Cytometry , Humans , Light , Male , Mean Platelet Volume , P-Selectin/blood , Peptide Fragments/blood , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptor, PAR-1/blood , Scattering, Radiation
16.
Blood ; 126(11): 1367-78, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26224646

ABSTRACT

UNLABELLED: Because Wiskott-Aldrich syndrome (WAS) and X-linked thrombocytopenia (XLT) patients have microthrombocytopenia, hemorrhage is a major problem. We asked whether eltrombopag, a thrombopoietic agent, would increase platelet counts, improve platelet activation, and/or reduce bleeding in WAS/XLT patients. In 9 WAS/XLT patients and 8 age-matched healthy controls, platelet activation was assessed by whole blood flow cytometry. Agonist-induced platelet surface activated glycoprotein (GP) IIb-IIIa and P-selectin in WAS/XLT patients were proportional to platelet size and therefore decreased compared with controls. In contrast, annexin V binding showed no differences between WAS/XLT and controls. Eltrombopag treatment resulted in an increased platelet count in 5 out of 8 patients. Among responders to eltrombopag, immature platelet fraction in 3 WAS/XLT patients was significantly less increased compared with 7 pediatric chronic immune thrombocytopenia (ITP) patients. Platelet activation did not improve in 3 WAS/XLT patients whose platelet count improved on eltrombopag. IN CONCLUSION: (1) the reduced platelet activation observed in WAS/XLT is primarily due to the microthrombocytopenia; and (2) although the eltrombopag-induced increase in platelet production in WAS/XLT is less than in ITP, eltrombopag has beneficial effects on platelet count but not platelet activation in the majority of WAS/XLT patients. This trial was registered at www.clinicaltrials.gov as #NCT00909363.


Subject(s)
Benzoates/therapeutic use , Genetic Diseases, X-Linked/blood , Genetic Diseases, X-Linked/drug therapy , Hydrazines/therapeutic use , Pyrazoles/therapeutic use , Thrombocytopenia/blood , Thrombocytopenia/drug therapy , Wiskott-Aldrich Syndrome/blood , Wiskott-Aldrich Syndrome/drug therapy , Adolescent , Adult , Case-Control Studies , Child , Child, Preschool , Humans , Infant , Male , Mean Platelet Volume , P-Selectin/blood , Platelet Activation/drug effects , Platelet Count , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Thrombopoietin/agonists , Young Adult
17.
Blood ; 126(4): 531-8, 2015 Jul 23.
Article in English | MEDLINE | ID: mdl-25964667

ABSTRACT

Although the biology of platelet adhesion on subendothelial matrix after vascular injury is well characterized, how the matrix biophysical properties affect platelet physiology is unknown. Here we demonstrate that geometric orientation of the matrix itself regulates platelet α-granule secretion, a key component of platelet activation. Using protein microcontact printing, we show that platelets spread beyond the geometric constraints of fibrinogen or collagen micropatterns with <5-µm features. Interestingly, α-granule exocytosis and deposition of the α-granule contents such as fibrinogen and fibronectin were primarily observed in those areas of platelet extension beyond the matrix protein micropatterns. This enables platelets to "self-deposit" additional matrix, provide more cellular membrane to extend spreading, and reinforce platelet-platelet connections. Mechanistically, this phenomenon is mediated by actin polymerization, Rac1 activation, and αIIbß3 integrin redistribution and activation, and is attenuated in gray platelet syndrome platelets, which lack α-granules, and Wiskott-Aldrich syndrome platelets, which have cytoskeletal defects. Overall, these studies demonstrate how platelets transduce geometric cues of the underlying matrix geometry into intracellular signals to extend spreading, which endows platelets spatial flexibility when spreading onto small sites of exposed subendothelium.


Subject(s)
Blood Platelets/cytology , Blood Platelets/metabolism , Exocytosis/physiology , Gray Platelet Syndrome/pathology , Platelet Adhesiveness/physiology , Wiskott-Aldrich Syndrome/pathology , Actin Cytoskeleton/metabolism , Case-Control Studies , Cell Membrane/metabolism , Cells, Cultured , Fibrinogen/metabolism , Fibronectins/metabolism , Gray Platelet Syndrome/metabolism , Humans , Immunoenzyme Techniques , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Pseudopodia , Wiskott-Aldrich Syndrome/metabolism
18.
Arterioscler Thromb Vasc Biol ; 36(3): 501-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26743169

ABSTRACT

OBJECTIVE: Unlike currently approved adenosine diphosphate receptor antagonists, the new diadenosine tetraphosphate derivative GLS-409 targets not only P2Y12 but also the second human platelet adenosine diphosphate receptor P2Y1 and may, therefore, be a promising antiplatelet drug candidate. The current study is the first to investigate the in vivo antithrombotic effects of GLS-409. APPROACH AND RESULTS: We studied (1) the in vivo effects of GLS-409 on agonist-stimulated platelet aggregation in anesthetized rats, (2) the antithrombotic activity of GLS-409 and the associated effect on the bleeding time in a canine model of platelet-mediated coronary artery thrombosis, and (3) the inhibition of agonist-stimulated platelet aggregation by GLS-409 versus selective P2Y1 and P2Y12 inhibition in vitro in samples from healthy human subjects before and 2 hours after aspirin intake. In vivo treatment with GLS-409 significantly inhibited adenosine diphosphate- and collagen-stimulated platelet aggregation in rats. Further, GLS-409 attenuated cyclic flow variation, that is, platelet-mediated thrombosis, in vivo in our canine model of unstable angina. The improvement in coronary patency was accompanied by a nonsignificant 30% increase in bleeding time. Of note, GLS-409 exerted its effects without affecting rat and canine hemodynamics. Finally, in vitro treatment with GLS-409 showed effects similar to that of cangrelor and the combination of cangrelor with the selective P2Y1 inhibitor MRS 2179 on agonist-stimulated platelet aggregation in human platelet-rich plasma and whole blood before and 2 hours after aspirin intake. CONCLUSIONS: Synergistic inhibition of both P2Y1 and P2Y12 adenosine diphosphate receptors by GLS-409 immediately attenuates platelet-mediated thrombosis and effectively blocks agonist-stimulated platelet aggregation irrespective of concomitant aspirin therapy.


Subject(s)
Blood Platelets/drug effects , Coronary Thrombosis/drug therapy , Dinucleoside Phosphates/pharmacology , Fibrinolytic Agents/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Receptors, Purinergic P2Y1/drug effects , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Adult , Animals , Aspirin/pharmacology , Blood Coagulation/drug effects , Blood Platelets/metabolism , Coronary Thrombosis/blood , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Female , Humans , Male , Platelet Function Tests , Rats, Sprague-Dawley , Receptors, Purinergic P2Y1/blood , Receptors, Purinergic P2Y12/blood , Time Factors , Young Adult
19.
Semin Thromb Hemost ; 42(3): 191-204, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26926581

ABSTRACT

Platelets are the smallest blood cells, numbering 150 to 350 × 10(9)/L in healthy individuals. The ability of activated platelets to adhere to an injured vessel wall and form aggregates was first described in the 19th century. Besides their long-established roles in thrombosis and hemostasis, platelets are increasingly recognized as pivotal players in numerous other pathophysiological processes including inflammation and atherogenesis, antimicrobial host defense, and tumor growth and metastasis. Consequently, profound knowledge of platelet structure and function is becoming more important in research and in many fields of modern medicine. This review provides an overview of platelet physiology focusing particularly on the structure, granules, surface glycoproteins, and activation pathways of platelets.


Subject(s)
Blood Platelets/physiology , Platelet Activation/physiology , Platelet Adhesiveness/physiology , Platelet Aggregation/physiology , Blood Platelets/metabolism , Hemostasis/physiology , Humans , Models, Biological , Platelet Membrane Glycoproteins/metabolism , Signal Transduction/physiology , Thrombosis/physiopathology
20.
Biomed Microdevices ; 18(4): 73, 2016 08.
Article in English | MEDLINE | ID: mdl-27464497

ABSTRACT

The vascular endothelium and shear stress are critical determinants of physiological hemostasis and platelet function in vivo, yet current diagnostic and monitoring devices do not fully incorporate endothelial function under flow in their assessment and, therefore, they can be unreliable and inaccurate. It is challenging to include the endothelium in assays for clinical laboratories or point-of-care settings because living cell cultures are not sufficiently robust. Here, we describe a microfluidic device that is lined by a human endothelium that is chemically fixed, but still retains its ability to modulate hemostasis under continuous flow in vitro even after few days of storage. This device lined with a fixed endothelium supports formation of platelet-rich thrombi in the presence of physiological shear, similar to a living arterial vessel. We demonstrate the potential clinical value of this device by showing that thrombus formation and platelet function can be measured within minutes using a small volume (0.5 mL) of whole blood taken from subjects receiving antiplatelet medications. The inclusion of a fixed endothelial microvessel will lead to biomimetic analytical devices that can potentially be used for diagnostics and point-of-care applications.


Subject(s)
Endothelium, Vascular/drug effects , Lab-On-A-Chip Devices , Thrombosis/diagnosis , Blood Platelets/drug effects , Endothelial Cells/drug effects , Fibrin/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Platelet Aggregation Inhibitors/pharmacology , Point-of-Care Systems , Stress, Mechanical , Thrombosis/blood , Thrombosis/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL