Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 440
1.
Biochem Biophys Res Commun ; 712-713: 149946, 2024 Jun 18.
Article En | MEDLINE | ID: mdl-38643717

Platelets are small anucleate cells that play a key role in thrombosis and hemostasis. Our group previously identified apolipoprotein A-IV (apoA-IV) as an endogenous inhibitor of thrombosis by competitive blockade of the αIIbß3 integrin on platelets. ApoA-IV inhibition of platelets was dependent on the N-terminal D5/D13 residues, and enhanced with absence of the C-terminus, suggesting it sterically hinders its N-terminal platelet binding site. The C-terminus is also the site of common apoA-IV polymorphisms apoA-IV-1a (T347S) and apoA-IV-2 (Q360H). Interestingly, both are linked with an increased risk of cardiovascular disease, however, the underlying mechanism remains unclear. Here, we generated recombinant apoA-IV and found that the Q360H or T347S polymorphisms dampened its inhibition of platelet aggregation in human platelet-rich plasma and gel-filtered platelets, reduced its inhibition of platelet spreading, and its inhibition of P-selectin on activated platelets. Using an ex vivo thrombosis assay, we found that Q360H and T347S attenuated its inhibition of thrombosis at both high (1800s-1) and low (300s-1) shear rates. We then demonstrate a conserved monomer-dimer distribution among apoA-IV WT, Q360H, and T347S and use protein structure modelling software to show Q360H and T347S enhance C-terminal steric hindrance over the N-terminal platelet-binding site. These data provide critical insight into increased cardiovascular risk for individuals with Q360H or T347S polymorphisms.


Apolipoproteins A , Blood Platelets , Platelet Aggregation , Thrombosis , Humans , Thrombosis/genetics , Thrombosis/metabolism , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Blood Platelets/metabolism , Blood Platelets/drug effects , Polymorphism, Genetic , Apoprotein(a)/genetics , Apoprotein(a)/metabolism , Apoprotein(a)/chemistry , P-Selectin/genetics , P-Selectin/metabolism
2.
J Int Med Res ; 52(2): 3000605241230429, 2024 Feb.
Article En | MEDLINE | ID: mdl-38420770

Strokes are the leading cause of death in most regions of the world. Epoxidase inhibitors include the drug aspirin (acetylsalicylic acid). Aspirin is widely used as first-line treatment for the prevention of cardiovascular and cerebrovascular diseases in at-risk patients. However, patients using conventional doses of aspirin can still develop ischaemic cardiovascular and cerebrovascular diseases, a phenomenon known as aspirin resistance. The occurrence of aspirin resistance hinders the prevention and treatment of ischaemic cardiovascular and cerebrovascular diseases. There are many factors affecting aspirin resistance, such as sex, drug dose, metabolic disease, genetic polymorphisms, drug interactions and pharmacokinetics. Genetic polymorphism refers to the simultaneous and frequent presence of two or more discontinuous variants or genotypes or alleles in a population of organisms. Platelets contain a large number of highly polymorphic transmembrane glycoprotein receptors encoded by two or more isomeric alleles. Changes in gene polymorphisms in various pathways during platelet aggregation can lead to aspirin resistance. This narrative review describes the gene polymorphisms that have been demonstrated to be significantly associated with aspirin resistance. Research on the mechanisms of aspirin resistance and increased knowledge should provide accurate drug guidance in individuals that require first-line antiplatelet therapy.


Oxidoreductases , Platelet Aggregation Inhibitors , Stroke , Humans , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Drug Resistance/genetics , Aspirin/pharmacology , Aspirin/therapeutic use , Polymorphism, Genetic , Platelet Aggregation/genetics , Stroke/drug therapy , Ischemia/drug therapy
3.
J Clin Invest ; 133(18)2023 09 15.
Article En | MEDLINE | ID: mdl-37471144

Protease-activated receptor 4 (PAR4) (gene F2RL3) harbors a functional dimorphism, rs773902 A/G (encoding Thr120/Ala120, respectively) and is associated with greater platelet aggregation. The A allele frequency is more common in Black individuals, and Black individuals have a higher incidence of ischemic stroke than White individuals. However, it is not known whether the A allele is responsible for worse stroke outcomes. To directly test the in vivo effect of this variant on stroke, we generated mice in which F2rl3 was replaced by F2RL3, thereby expressing human PAR4 (hPAR4) with either Thr120 or Ala120. Compared with hPAR4 Ala120 mice, hPAR4 Thr120 mice had worse stroke outcomes, mediated in part by enhanced platelet activation and platelet-neutrophil interactions. Analyses of 7,620 Black subjects with 487 incident ischemic strokes demonstrated the AA genotype was a risk for incident ischemic stroke and worse functional outcomes. In humanized mice, ticagrelor with or without aspirin improved stroke outcomes in hPAR4 Ala120 mice, but not in hPAR4 Thr120 mice. P selectin blockade improved stroke outcomes and reduced platelet-neutrophil interactions in hPAR4 Thr120 mice. Our results may explain some of the racial disparity in stroke and support the need for studies of nonstandard antiplatelet therapies for patients expressing PAR4 Thr120.


Ischemic Stroke , Stroke , Humans , Animals , Mice , Receptors, Thrombin/genetics , Platelet Aggregation/genetics , Blood Platelets/physiology , Platelet Aggregation Inhibitors/pharmacology , Stroke/genetics , Receptor, PAR-1
4.
Biomolecules ; 14(1)2023 Dec 27.
Article En | MEDLINE | ID: mdl-38254638

Growth differentiation factor-15 (GDF-15) is proposed to be strongly associated with several cardiovascular diseases, such as heart failure and atherosclerosis. Moreover, some recent studies have reported an association between GDF-15 and platelet activation. In this study, we isolated peripheral blood platelets from healthy volunteers and evaluated the effect of GDF-15 on adenosine diphosphate (ADP)-induced platelet activation using the platelet aggregation assay. Subsequently, we detected the expression of GDF-15-related receptors on platelets, including the epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), human epidermal growth factor receptor 3 (HER3), transforming growth factor-beta receptor I (TGF-ßRI), transforming growth factor-beta receptor II (TGF-ßRII), glial-cell-line-derived neurotrophic factor family receptor α-like (GFRAL), and those rearranged during transfection (RET). Then, we screened for GDF-15 receptors using the GDF-15-related receptor microarray comprising these recombinant proteins. We also performed the immunoprecipitation assay to investigate the interaction between GDF-15 and the receptors on platelets. For the further exploration of signaling pathways, we investigated the effects of GDF-15 on the extracellular signal-regulated kinase (ERK), protein kinase B (AKT), and Janus kinase 2 (JAK2) pathways. We also investigated the effects of GDF-15 on the ERK and AKT pathways and platelet aggregation in the presence or absence of RET agonists or inhibition. Our study revealed that GDF-15 can dose-independently inhibit ADP-induced human platelet aggregation and that the binding partner of GDF-15 on platelets is GFRAL. We also found that GDF-15 inhibits ADP-induced AKT and ERK activation in platelets. Meanwhile, our results revealed that the inhibitory effects of GDF-15 can be mediated by the GFRAL/RET complex. These findings reveal the novel inhibitory mechanism of ADP-induced platelet activation by GDF-15.


Glial Cell Line-Derived Neurotrophic Factor Receptors , Growth Differentiation Factor 15 , Platelet Aggregation , Proto-Oncogene Proteins c-ret , Humans , Adenosine Diphosphate/pharmacology , ErbB Receptors , Extracellular Signal-Regulated MAP Kinases , Growth Differentiation Factor 15/pharmacology , Platelet Aggregation/genetics , Proto-Oncogene Proteins c-akt , Transforming Growth Factors , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism
5.
Pharmacogenomics ; 23(11): 639-648, 2022 07.
Article En | MEDLINE | ID: mdl-35880550

Background: Patients might still experience major adverse cardiovascular events even with dual antiplatelet therapy after percutaneous coronary intervention. Our study aimed to explore the impact of gene polymorphism on clinical outcomes in one-year follow-up. Methods: A total of 171 patients treated with dual antiplatelet therapy after percutaneous coronary intervention from April to December 2020 in the first hospital of Jilin University enrolled in this study. Results: PEAR1 genetic polymorphisms was associated with the arachidonic acid (AA) and adenosine diphosphate (ADP) platelet aggregation. Hyperglycemia was associated with the rate of major adverse cardiovascular events. PEAR1 GA+AA genetic genetic polymorphisms is associated with hyperglycemia. Conclusion:PEAR1 GG is a risk factor for AA and ADP platelet aggregation. Hyperglycemia can effect the one-year outcome. PEAR1 GA+AA genetic polymorphisms are associated with hyperglycemia.


Cardiovascular Diseases , Hyperglycemia , Percutaneous Coronary Intervention , Adenosine Diphosphate/pharmacology , Aspirin/adverse effects , Cardiovascular Diseases/drug therapy , China , Clopidogrel/adverse effects , Humans , Hyperglycemia/chemically induced , Hyperglycemia/drug therapy , Hyperglycemia/genetics , Percutaneous Coronary Intervention/adverse effects , Platelet Aggregation/genetics , Platelet Aggregation Inhibitors/adverse effects , Polymorphism, Genetic/genetics , Receptors, Cell Surface/genetics
6.
Int J Mol Sci ; 23(2)2022 Jan 11.
Article En | MEDLINE | ID: mdl-35054930

Inhalation of particulate matter in polluted air causes direct, size-restricted passage in the circulation and pronounced lung inflammation, provoking platelet activation and (non)-fatal cardiovascular complications. To determine potency and mechanism of platelet sensitization via neutrophil enzymes, we performed in vitro aggregation studies in washed human platelets and in murine and human blood, in the presence of elastase, cathepsin G and regular platelet agonists, present in damaged arteries. The impact of both enzymes on in vivo thrombogenicity was studied in the same thrombosis mouse model, previously having demonstrated that neutrophil activation enhances peripheral thrombogenicity. At 0.05 U/mL, cathepsin G activated washed human platelets via PAR1, whereas at 0.35 U/mL, aggregation occurred via PAR4. In Swiss mouse platelet-rich plasma no aggregation occurred by cathepsin G at 0.4 U/mL. In human and murine blood, aggregations by 0.05-0.1 U/mL cathepsin G were similar and not PAR-mediated, but platelet aggregation was inhibited by ADP antagonists, advocating cathepsin G-released ADP in blood as the true agonist of sustained platelet activation. In the mouse thrombosis model, cathepsin G and elastase amplified mild thrombogenicity at blood concentrations that activated platelets in vitro. This study shows that cathepsin G and elastase secreted in the circulation during mild air pollution-induced lung inflammation lyse red blood cell membrane proteins, leading to ADP-leakage into plasma, sensitizing platelets and amplifying their contribution to cardiovascular complications of ambient particle inhalation.


Arteries/metabolism , Blood Platelets/metabolism , Cathepsin G/metabolism , Neutrophils/metabolism , Platelet Activation , Thrombosis/etiology , Thrombosis/metabolism , Adenosine Diphosphate/metabolism , Animals , Arteries/pathology , Biomarkers , Cathepsin G/genetics , Disease Susceptibility , Humans , Mice , Mice, Knockout , Neutrophil Activation , Pancreatic Elastase/metabolism , Platelet Activation/genetics , Platelet Aggregation/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Thrombosis/pathology
7.
Blood ; 139(9): 1374-1388, 2022 03 03.
Article En | MEDLINE | ID: mdl-34905618

Genetic variants within the fibrinogen Aα chain encoding the αC-region commonly result in hypodysfibrinogenemia in patients. However, the (patho)physiological consequences and underlying mechanisms of such mutations remain undefined. Here, we generated Fga270 mice carrying a premature termination codon within the Fga gene at residue 271. The Fga270 mutation was compatible with Mendelian inheritance for offspring of heterozygous crosses. Adult Fga270/270 mice were hypofibrinogenemic with ∼10% plasma fibrinogen levels relative to FgaWT/WT mice, linked to 90% reduction in hepatic Fga messenger RNA (mRNA) because of nonsense-mediated decay of the mutant mRNA. Fga270/270 mice had preserved hemostatic potential in vitro and in vivo in models of tail bleeding and laser-induced saphenous vein injury, whereas Fga-/- mice had continuous bleeding. Platelets from FgaWT/WT and Fga270/270 mice displayed comparable initial aggregation following adenosine 5'-diphosphate stimulation, but Fga270/270 platelets quickly disaggregated. Despite ∼10% plasma fibrinogen, the fibrinogen level in Fga270/270 platelets was ∼30% of FgaWT/WT platelets with a compensatory increase in fibronectin. Notably, Fga270/270 mice showed complete protection from thrombosis in the inferior vena cava stasis model. In a model of Staphylococcus aureus peritonitis, Fga270/270 mice supported local, fibrinogen-mediated bacterial clearance and host survival comparable to FgaWT/WT, unlike Fga-/- mice. Decreasing the normal fibrinogen levels to ∼10% with small interfering RNA in mice also provided significant protection from venous thrombosis without compromising hemostatic potential and antimicrobial function. These findings both reveal novel molecular mechanisms underpinning fibrinogen αC-region truncation mutations and highlight the concept that selective fibrinogen reduction may be efficacious for limiting thrombosis while preserving hemostatic and immune protective functions.


Afibrinogenemia , Blood Platelets/metabolism , Fibrinogen , Hemostasis/genetics , Mutation , Platelet Aggregation/genetics , Thrombosis , Afibrinogenemia/genetics , Afibrinogenemia/metabolism , Animals , Fibrinogen/genetics , Fibrinogen/metabolism , Mice , Mice, Knockout , Thrombosis/genetics , Thrombosis/metabolism
8.
Clin Appl Thromb Hemost ; 27: 10760296211044212, 2021.
Article En | MEDLINE | ID: mdl-34617458

The sticky platelet syndrome (SPS) was described by Mammen in 1983. Since then, scientists in several countries have identified the condition and published cases or series of patients, thus enabling the description of the prevalence of the inherited condition, its salient clinical features, and the treatment of the disease. The diagnosis of the SPS phenotype requires fresh blood samples and special equipment which is not available in all coagulation laboratories. In the era of molecular biology, up to now it has not been possible to define a clear association of the SPS phenotype with a specific molecular marker. Some molecular changes which have been described in platelet proteins in some persons with the phenotype of the SPS are here discussed. Nowadays, the SPS phenotype may be considered as a risk factor for thrombosis and most cases of the SPS developing vaso-occlussive episodes are the result of its coexistence with other thrombosis-prone conditions, some of the inherited and some of them acquired, thus leading to the concept of multifactorial thrombophilia. Ignoring all these evidence-based concepts is inappropriate, same as stating that the SPS is a nonentity simply because not all laboratories are endowed with adequate equipment to support the diagnosis.


Blood Platelet Disorders/genetics , Platelet Aggregation/genetics , Thrombophilia/etiology , Animals , Genotype , Humans , Mice , Phenotype , Syndrome , Thrombophilia/pathology
9.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article En | MEDLINE | ID: mdl-34638997

One of the mechanisms by which PI3 kinase can regulate platelet function is through phosphorylation of downstream substrates, including glycogen synthase kinase-3 (GSK3)α and GSK3ß. Platelet activation results in the phosphorylation of an N-terminal serine residue in GSK3α (Ser21) and GSK3ß (Ser9), which competitively inhibits substrate phosphorylation. However, the role of phosphorylation of these paralogs is still largely unknown. Here, we employed GSK3α/ß phosphorylation-resistant mouse models to explore the role of this inhibitory phosphorylation in regulating platelet activation. Expression of phosphorylation-resistant GSK3α/ß reduced thrombin-mediated platelet aggregation, integrin αIIbß3 activation, and α-granule secretion, whereas platelet responses to the GPVI agonist collagen-related peptide (CRP-XL) were significantly enhanced. GSK3 single knock-in lines revealed that this divergence is due to differential roles of GSK3α and GSK3ß phosphorylation in regulating platelet function. Expression of phosphorylation-resistant GSK3α resulted in enhanced GPVI-mediated platelet activation, whereas expression of phosphorylation-resistant GSK3ß resulted in a reduction in PAR-mediated platelet activation and impaired in vitro thrombus formation under flow. Interestingly, the latter was normalised in double GSK3α/ß KI mice, indicating that GSK3α KI can compensate for the impairment in thrombosis caused by GSK3ß KI. In conclusion, our data indicate that GSK3α and GSK3ß have differential roles in regulating platelet function.


Blood Platelets/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3/metabolism , Platelet Activation/genetics , Platelet Aggregation/genetics , Signal Transduction/genetics , Thrombosis/metabolism , Animals , Blood Donors , Cells, Cultured , Disease Models, Animal , Gene Knock-In Techniques , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta/genetics , Humans , Integrins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/metabolism , Thrombin/metabolism , Thrombosis/genetics
10.
Int J Mol Sci ; 22(9)2021 May 03.
Article En | MEDLINE | ID: mdl-34063730

Based on recent findings that show that depletion of factor XII (FXII) leads to better posttraumatic neurological recovery, we studied the effect of FXII-deficiency on post-traumatic cognitive and behavioral outcomes in female and male mice. In agreement with our previous findings, neurological deficits on day 7 after weight-drop traumatic brain injury (TBI) were significantly reduced in FXII-/- mice compared to wild type (WT) mice. Also, glycoprotein Ib (GPIb)-positive platelet aggregates were more frequent in brain microvasculature of WT than FXII-/- mice 3 months after TBI. Six weeks after TBI, memory for novel object was significantly reduced in both female and male WT but not in FXII-/- mice compared to sham-operated mice. In the setting of automated home-cage monitoring of socially housed mice in IntelliCages, female WT mice but not FXII-/- mice showed decreased exploration and reacted negatively to reward extinction one month after TBI. Since neuroendocrine stress after TBI might contribute to trauma-induced cognitive dysfunction and negative emotional contrast reactions, we measured peripheral corticosterone levels and the ration of heart, lung, and spleen weight to bodyweight. Three months after TBI, plasma corticosterone levels were significantly suppressed in both female and male WT but not in FXII-/- mice, while the relative heart weight increased in males but not in females of both phenotypes when compared to sham-operated mice. Our results indicate that FXII deficiency is associated with efficient post-traumatic behavioral and neuroendocrine recovery.


Brain Injuries, Traumatic/genetics , Cognitive Dysfunction/genetics , Factor XII Deficiency/genetics , Factor XII/genetics , Animals , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/blood , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/pathology , Cognitive Dysfunction/blood , Cognitive Dysfunction/complications , Cognitive Dysfunction/pathology , Corticosterone/blood , Disease Models, Animal , Factor XII Deficiency/blood , Factor XII Deficiency/complications , Factor XII Deficiency/pathology , Humans , Memory/physiology , Mice , Mice, Knockout , Platelet Aggregation/genetics , Platelet Glycoprotein GPIb-IX Complex
11.
Int J Mol Sci ; 22(8)2021 Apr 14.
Article En | MEDLINE | ID: mdl-33920051

Atherothrombosis exposes vascular components to blood. Currently, new antithrombotic therapies are emerging. Herein we investigated thrombogenesis of human arteries with/without atherosclerosis, and the interaction of coagulation and vascular components, we and explored the anti-thrombogenic efficacy of blockade of the P2X purinoceptor 7 (P2X7). A confocal blood flow videomicroscopy system was performed on cryosections of internal mammary artery (IMA) or carotid plaque (CPL) determining/localizing platelets and fibrin. Blood from healthy donors elicited thrombi over arterial layers. Confocal microscopy associated thrombus with tissue presence of collagen type I, laminin, fibrin(ogen) and tissue factor (TF). The addition of antibodies blocking TF (aTF) or factor XI (aFXI) to blood significantly reduced fibrin deposition, variable platelet aggregation and aTF + aFXI almost abolished thrombus formation, showing synergy between coagulation pathways. A scarce effect of aTF over sub-endothelial regions, more abundant in tissue TF and bundles of laminin and collagen type I than deep intima, may suggest tissue thrombogenicity as molecular structure-related. Consistently with TF-related vascular function and expression of P2X7, the sections from CPL but not IMA tissue cultures pre-treated with the P2X7 antagonist A740003 demonstrated poor thrombogenesis in flow experiments. These data hint to local targeting studies on P2X7 modulation for atherothrombosis prevention/therapy.


Atherosclerosis/diagnostic imaging , Blood Platelets/ultrastructure , Microscopy, Video , Receptors, Purinergic P2X7/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Blood Circulation/physiology , Blood Coagulation/genetics , Blood Platelets/metabolism , Carotid Arteries/diagnostic imaging , Carotid Arteries/ultrastructure , Fibrin/genetics , Humans , Microscopy, Confocal , Platelet Aggregation/genetics , Thrombosis/diagnostic imaging , Thrombosis/pathology
12.
Int J Mol Sci ; 22(6)2021 Mar 11.
Article En | MEDLINE | ID: mdl-33799528

Obstructive sleep apnoea (OSA) is a common disease which is characterised by repetitive collapse of the upper airways during sleep resulting in chronic intermittent hypoxaemia and frequent microarousals, consequently leading to sympathetic overflow, enhanced oxidative stress, systemic inflammation, and metabolic disturbances. OSA is associated with increased risk for cardiovascular morbidity and mortality, and accelerated coagulation, platelet activation, and impaired fibrinolysis serve the link between OSA and cardiovascular disease. In this article we briefly describe physiological coagulation and fibrinolysis focusing on processes which could be altered in OSA. Then, we discuss how OSA-associated disturbances, such as hypoxaemia, sympathetic system activation, and systemic inflammation, affect these processes. Finally, we critically review the literature on OSA-related changes in markers of coagulation and fibrinolysis, discuss potential reasons for discrepancies, and comment on the clinical implications and future research needs.


Acute Coronary Syndrome/metabolism , Fibrinolysis/genetics , Hypoxia/metabolism , Sleep Apnea, Obstructive/metabolism , Stroke/metabolism , Venous Thrombosis/metabolism , Acute Coronary Syndrome/complications , Acute Coronary Syndrome/genetics , Acute Coronary Syndrome/physiopathology , Blood Coagulation Factors/genetics , Blood Coagulation Factors/metabolism , Blood Platelets/metabolism , Blood Platelets/pathology , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression Regulation , Humans , Hypoxia/complications , Hypoxia/genetics , Hypoxia/physiopathology , Inflammation , Oxidative Stress , Platelet Activation/genetics , Platelet Aggregation/genetics , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/genetics , Sleep Apnea, Obstructive/physiopathology , Stroke/complications , Stroke/genetics , Stroke/physiopathology , Venous Thrombosis/complications , Venous Thrombosis/genetics , Venous Thrombosis/physiopathology
13.
Haematologica ; 106(5): 1423-1432, 2021 05 01.
Article En | MEDLINE | ID: mdl-32299908

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.


Aspirin , Platelet Function Tests , Blood Platelets , Cyclooxygenase 1/genetics , Female , Humans , Platelet Aggregation/genetics , Thromboxane A2
14.
Semin Cell Dev Biol ; 112: 1-7, 2021 04.
Article En | MEDLINE | ID: mdl-32563678

The ability to study the behavior of cells, proteins, and cell-cell or cell-protein interactions under dynamic forces such as shear stress under fluid flow, provides a more accurate understanding of the physiopathology of hemostasis. This review touches upon the traditional methods for studying blood coagulation and platelet aggregation and provides an overview on cellular and protein response to shear stress. We also elaborate on the biological aspects of how cells recognize mechanical forces and convert them into biochemical signals that can drive various signaling pathways. We give a detailed description of the various types of microfluidic devices that are employed to study the complex processes of platelet aggregation and blood coagulation under flow conditions as well as to investigate endothelial shear-response. We also highlight works mimicking artificial vessels as platforms to study the mechanisms of coagulation, and finish our review by describing anticipated clinical uses of microfluidics devices and their standardization.


Blood Coagulation/physiology , Hemostasis/physiology , Lab-On-A-Chip Devices , Thrombosis/genetics , Blood Coagulation/genetics , Hemostasis/genetics , Humans , Platelet Aggregation/genetics , Platelet Aggregation/physiology , Signal Transduction/genetics , Thrombosis/physiopathology
15.
Platelets ; 32(4): 507-515, 2021 May 19.
Article En | MEDLINE | ID: mdl-32449466

Advanced glycation end products (AGEs) arising from dietary intake have been associated with numerous chronic diseases including cardiovascular diseases. The interaction between platelets and AGEs has been proposed to play a role in the etiology of cardiovascular diseases. However, the effects of the interaction between platelets and Maillard reaction products generated from glyoxal (Gly) or methylglyoxal (MG) are poorly understood. In this work, the effects of AGEs generated by the reaction between Gly or MG with Lys or bovine serum albumin (BSA) on platelet activation and aggregation were assessed. AGEs were generated incubating Gly or MG with Lys or BSA during 5 hours or 14 days, respectively. AGEs generation were characterized by kinetic studies and by amino acid analysis. Human platelet-rich plasma (PRP) was incubated with different concentrations of AGEs from Lys-MG or Lys-Gly and BSA-MG or BSA-Gly. Platelet activation was determined quantifying the expression of CD62 (P-selectin) in PRP exposed to different AGEs concentrations. It was found that Lys-MG and Lys-Gly induced an increase in P-selectin expression (p < .05), being 33.9% higher for Lys-MG when compared to Lys-Gly. Platelets incubated in the presence of BSA-MG and BSA-Gly did not show an increase in the P-selectin expression. Platelet aggregation was significantly higher for the mixture Lys-MG (in all the range of concentrations evaluated), whereas for Lys-Gly it was only significant the highest concentration (Lys 168 µM/Gly 168 µM). It was observed a significant increase in platelet aggregation induced by ADP for samples BSA-Gly. AGEs formed with MG-Lys induce a higher activation and aggregation of platelets when compared to those formed from Gly-Lys.


Glycation End Products, Advanced/adverse effects , Glyoxal/therapeutic use , Platelet Activation/genetics , Platelet Aggregation/genetics , Pyruvaldehyde/therapeutic use , Glyoxal/pharmacology , Humans , Pyruvaldehyde/pharmacology
16.
Methods Mol Biol ; 2217: 237-249, 2021.
Article En | MEDLINE | ID: mdl-33215384

Platelets are small, anucleate cells that play oversized roles in hemostasis, immunity, and inflammation. An important mediator of platelet function is integrin αIIbß3, which is required for fibrinogen-dependent platelet aggregation during hemostasis. This platelet response is dependent on conformational changes in the integrin induced by "inside-out" biochemical signals that are triggered by platelet agonists. In turn, fibrinogen binding to αIIbß3 initiates "outside-in" biochemical and mechanical signals that regulate the platelet cytoskeleton and help to promote full platelet aggregation and secretory responses. Without a nucleus, there is a limited range of experimental manipulations that are possible with human platelets to study the molecular basis of integrin signaling in these primary cells. Consequently, many studies of αIIbß3 function use genetic approaches that rely on heterologous expression systems or platelets from gene-targeted mice, sometimes with uncertain applicability to human platelets. This chapter will detail a method for genetic manipulation of megakaryocytes and platelets derived from human induced pluripotent stem cells for molecular studies of αIIbß3 signaling and for modeling of human platelet functions potentially relevant to hemostasis, immunity, and inflammation.


Blood Platelets/metabolism , Cell Engineering/methods , Megakaryocytes/metabolism , Platelet Aggregation/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIb-IX Complex/genetics , Platelet Membrane Glycoprotein IIb/genetics , Blood Platelets/cytology , Cell Differentiation , Cell Line , Cytoskeleton/metabolism , Cytoskeleton/ultrastructure , Fibrinogen/genetics , Fibrinogen/metabolism , Gene Expression Regulation , Hemostasis/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Megakaryocytes/cytology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Membrane Glycoprotein IIb/metabolism , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Ubiquitins/antagonists & inhibitors , Ubiquitins/genetics , Ubiquitins/metabolism
17.
Int J Mol Sci ; 21(23)2020 Nov 25.
Article En | MEDLINE | ID: mdl-33255747

Distinct membrane receptors activate platelets by Src-family-kinase (SFK)-, immunoreceptor-tyrosine-based-activation-motif (ITAM)-dependent stimulation of spleen tyrosine kinase (Syk). Recently, we reported that platelet activation via glycoprotein (GP) VI or GPIbα stimulated the well-established Syk tyrosine (Y)-phosphorylation, but also stoichiometric, transient protein kinase C (PKC)-mediated Syk serine(S)297 phosphorylation in the regulatory interdomain-B, suggesting possible feedback inhibition. The transient nature of Syk S297 phosphorylation indicated the presence of an unknown Syk pS297 protein phosphatase. In this study, we hypothesize that the S-protein phosphatase 2A (PP2A) is responsible for Syk pS297 dephosphorylation, thereby affecting Syk Y-phosphorylation and activity in human washed platelets. Using immunoblotting, we show that specific inhibition of PP2A by okadaic acid (OA) alone leads to stoichiometric Syk S297 phosphorylation, as analyzed by Zn2+-Phos-tag gels, without affecting Syk Y-phosphorylation. Pharmacological inhibition of Syk by PRT060318 or PKC by GF109203X only minimally reduced OA-induced Syk S297 phosphorylation. PP2A inhibition by OA preceding GPVI-mediated platelet activation induced by convulxin extended Syk S297 phosphorylation but inhibited Syk Y-phosphorylation. Our data demonstrate a novel biochemical and functional link between the S-protein phosphatase PP2A and the Y-protein kinase Syk in human platelets, and suggest that PP2A represents a potential enhancer of GPVI-mediated Syk activity caused by Syk pS297 dephosphorylation.


Blood Platelets/metabolism , Platelet Activation/genetics , Protein Phosphatase 2/genetics , Syk Kinase/genetics , Humans , Phosphorylation , Platelet Aggregation/genetics , Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics
18.
Int J Mol Sci ; 21(22)2020 Nov 23.
Article En | MEDLINE | ID: mdl-33238433

Inflammatory processes are triggered by the fibrinolytic enzyme plasmin. Tissue-type plasminogen activator, which cleaves plasminogen to plasmin, can be activated by the cross-ß-structure of misfolded proteins. Misfolded protein aggregates also represent substrates for plasmin, promoting their degradation, and are potent platelet agonists. However, the regulation of plasmin-mediated platelet activation by misfolded proteins and vice versa is incompletely understood. In this study, we hypothesize that plasmin acts as potent agonist of human platelets in vitro after short-term incubation at room temperature, and that the response to thrombospondin-1 and the bona fide misfolded proteins Eap and SCN--denatured IgG interfere with plasmin, thereby modulating platelet activation. Plasmin dose-dependently induced CD62P surface expression on, and binding of fibrinogen to, human platelets in the absence/presence of plasma and in citrated whole blood, as analyzed by flow cytometry. Thrombospondin-1 pre-incubated with plasmin enhanced these plasmin-induced platelet responses at low concentration and diminished them at higher dose. Platelet fibrinogen binding was dose-dependently induced by the C-terminal thrombospondin-1 peptide RFYVVMWK, Eap or NaSCN-treated IgG, but diminished in the presence of plasmin. Blocking enzymatically catalyzed thiol-isomerization decreased plasmin-induced platelet responses, suggesting that plasmin activates platelets in a thiol-dependent manner. Thrombospondin-1, depending on the concentration, may act as cofactor or inhibitor of plasmin-induced platelet activation, and plasmin blocks platelet activation induced by misfolded proteins and vice versa, which might be of clinical relevance.


Blood Platelets/metabolism , Inflammation/genetics , Platelet Aggregation/genetics , Thrombospondin 1/blood , Fibrinogen/genetics , Fibrinogen/metabolism , Fibrinolysin/metabolism , Humans , Inflammation/blood , Inflammation/metabolism , Isomerases/genetics , Isomerases/metabolism , P-Selectin/blood , P-Selectin/genetics , Peptides/genetics , Peptides/pharmacology , Plasminogen/genetics , Plasminogen/metabolism , Platelet Activation/genetics , Protein Aggregates/genetics , Protein Conformation, beta-Strand , Protein Folding/drug effects , Sulfhydryl Compounds/blood , Sulfhydryl Compounds/metabolism , Thrombospondin 1/genetics
19.
Int J Mol Sci ; 21(21)2020 Nov 03.
Article En | MEDLINE | ID: mdl-33153214

Diabetes is associated with platelet hyper-reactivity and enhanced risk of thrombosis development. Here we compared protein expression in platelets from healthy donors and diabetic patients to identify differentially expressed proteins and their possible function in platelet activation. Mass spectrometry analyses identified cyclin Y (CCNY) in platelets and its reduced expression in platelets from diabetic patients, a phenomenon that could be attributed to the increased activity of calpains. To determine the role of CCNY in platelets, mice globally lacking the protein were studied. CCNY-/- mice demonstrated lower numbers of circulating platelets but platelet responsiveness to thrombin and a thromboxane A2 analogue were comparable with that of wild-type mice, as was agonist-induced α and dense granule secretion. CCNY-deficient platelets demonstrated enhanced adhesion to fibronectin and collagen as well as an attenuated spreading and clot retraction, indicating an alteration in "outside in" integrin signalling. This phenotype was accompanied by a significant reduction in the agonist-induced tyrosine phosphorylation of ß3 integrin. Taken together we have shown that CCNY is present in anucleated platelets where it is involved in the regulation of integrin-mediated outside in signalling associated with thrombin stimulation.


Blood Platelets/metabolism , Cyclins/genetics , Integrins/metabolism , Adult , Animals , Cyclins/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Platelet Activation/genetics , Platelet Adhesiveness/genetics , Platelet Aggregation/genetics , Signal Transduction/genetics , Young Adult
20.
Front Immunol ; 11: 1522, 2020.
Article En | MEDLINE | ID: mdl-32765527

The cross-talk between the inflammatory complement system and hemostasis is becoming increasingly recognized. The interaction between complement C1q, initiation molecule of the classical pathway, and von Willebrand factor (vWF), initiator molecule of primary hemostasis, has been shown to induce platelet rolling and adhesion in vitro. As vWF disorders result in prolonged bleeding, a lack of C1q as binding partner for vWF might also lead to an impaired hemostasis. Therefore, this study aimed to investigate the in vivo relevance of C1q-dependent binding of vWF in hemostasis. For this purpose, we analyzed parameters of primary and secondary hemostasis and performed bleeding experiments in wild type (WT) and C1q-deficient (C1qa-/-) mice, with reconstitution experiments of C1q in the latter. Bleeding tendency was examined by quantification of bleeding time and blood loss. First, we found that complete blood counts and plasma vWF levels do not differ between C1qa-/- mice and WT mice. Moreover, platelet aggregation tests indicated that the platelets of both strains of mice are functional. Second, while the prothrombin time was comparable between both groups, the activated partial thromboplastin time was shorter in C1qa-/- mice. In contrast, tail bleeding times of C1qa-/- mice were prolonged accompanied by an increased blood loss. Upon reconstitution of C1qa-/- mice with C1q, parameters of increased bleeding could be reversed. In conclusion, our data indicate that C1q, a molecule of the first-line of immune defense, actively participates in primary hemostasis by promoting arrest of bleeding. This observation might be of relevance for the understanding of thromboembolic complications in inflammatory disorders, where excess of C1q deposition is observed.


Complement C1q/immunology , Hemostasis/immunology , Animals , Biomarkers , Blood Cell Count , Blood Coagulation/genetics , Blood Coagulation/immunology , Blood Coagulation Tests , Blood Platelets/immunology , Blood Platelets/metabolism , Complement C1q/genetics , Hemostasis/genetics , Mice , Mice, Knockout , Platelet Activation/immunology , Platelet Aggregation/genetics , Platelet Aggregation/immunology , von Willebrand Factor/metabolism
...