Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Arterioscler Thromb Vasc Biol ; 43(9): 1700-1712, 2023 09.
Article in English | MEDLINE | ID: mdl-37409530

ABSTRACT

BACKGROUND: Platelets and neutrophils are the first blood cells accumulating at sites of arterial thrombus formation, and both cell types contribute to the pathology of thrombotic events. We aimed to identify key interaction mechanisms between these cells using microfluidic approaches. METHODS: Whole-blood perfusion was performed over a collagen surface at arterial shear rate. Platelet and leukocyte (in majority neutrophil) activation were microscopically visualized using fluorescent markers. The contributions of platelet-adhesive receptors (integrin, P-selectin, CD40L) and chemokines were studied by using inhibitors or antibodies and using blood from patients with GT (Glanzmann thrombasthenia) lacking platelet-expressed αIIbß3. RESULTS: We observed (1) an unknown role of activated platelet integrin αIIbß3 preventing leukocyte adhesion, which was overcome by short-term flow disturbance provoking massive adhesion; (2) that platelet-expressed CD40L controls the crawling pattern and thrombus fidelity of the cells on a thrombus; (3) that continued secretion of platelet substances promotes activation of identified neutrophils, as assessed by (fMLP [N-formylmethionyl-leucyl-phenylalanine, a potent chemotactic agent and leukocyte activator] induced) [Ca2+]i rises and antigen expression; (4) and that platelet-released chemokines activate the adhered cells in the order of CXCL7>CCL5>CXCL4. Furthermore, postsilencing of the platelets in a thrombus suppressed the leukocyte activation. However, the leukocytes on thrombi did no more than limitedly form neutrophil extracellular traps, unless stimulated with phorbol ester or lipopolysaccharide. CONCLUSIONS: Together, these findings reveal a multifaceted regulation of adhesion and activation of neutrophils by platelets in a thrombus, with a balanced role of several platelet-adhesive receptors and a promoting role of platelet-released substances. This multivalent nature of neutrophil-thrombus interactions offers novel prospects for pharmacological intervention.


Subject(s)
Arteries , Blood Platelets , Chemokines , Neutrophil Activation , Neutrophils , Thrombosis , Blood Platelets/immunology , Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Chemokines/metabolism , Thrombosis/immunology , CD40 Ligand , Neutrophils/immunology , Neutrophils/metabolism , Cell Adhesion , Humans
2.
Arterioscler Thromb Vasc Biol ; 40(3): e65-e77, 2020 03.
Article in English | MEDLINE | ID: mdl-31893947

ABSTRACT

OBJECTIVE: In patients with diabetes mellitus, increased platelet reactivity predicts cardiac events. Limited evidence suggests that DPP-4 (dipeptidyl peptidase 4) influences platelets via GLP-1 (glucagon-like peptide 1)-dependent effects. Because DPP-4 inhibitors are frequently used in diabetes mellitus to improve the GLP-1-regulated glucose metabolism, we characterized the role of DPP-4 inhibition and of native intact versus DPP-4-cleaved GLP-1 on flow-dependent thrombus formation in mouse and human blood. Approach and Results: An ex vivo whole blood microfluidics model was applied to approach in vivo thrombosis and study collagen-dependent platelet adhesion, activation, and thrombus formation under shear-flow conditions by multiparameter analyses. In mice, in vivo inhibition or genetic deficiency of DPP-4 (Dpp4-/-), but not of GLP-1-receptors (Glp1r-/-), suppressed flow-dependent platelet aggregation. In human blood, GLP-1(7-36), but not DPP-4-cleaved GLP-1(9-36), reduced thrombus volume by 32% and impaired whole blood thrombus formation at both low/venous and high/arterial wall-shear rates. These effects were enforced upon ADP costimulation and occurred independently of plasma factors and leukocytes. Human platelets did not contain detectable levels of GLP-1-receptor transcripts. Also, GLP-1(7-36) did not inhibit collagen-induced aggregation under conditions of stirring or stasis of platelets, pointing to a marked flow-dependent role. CONCLUSIONS: Native, intact GLP-1 is a natural suppressor of thrombus growth under physiological flow conditions, with DPP-4 inhibition and increased intact GLP-1 suppressing platelet aggregation under flow without a main relevance of GLP-1-receptor on platelets.


Subject(s)
Blood Platelets/drug effects , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Fibrinolytic Agents/pharmacology , Glucagon-Like Peptide 1/metabolism , Linagliptin/pharmacology , Sitagliptin Phosphate/pharmacology , Thrombosis/prevention & control , Animals , Blood Platelets/metabolism , Dipeptidyl Peptidase 4/genetics , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Peptide Fragments/metabolism , Platelet Aggregation/drug effects , Signal Transduction , Thrombosis/enzymology , Thrombosis/genetics
3.
Blood ; 132(24): e35-e46, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30275110

ABSTRACT

Antithrombotic therapies reduce cardiovascular diseases by preventing arterial thrombosis and thromboembolism, but at expense of increased bleeding risks. Arterial thrombosis studies using genetically modified mice have been invaluable for identification of new molecular targets. Because of low sample sizes and heterogeneity in approaches or methodologies, a formal meta-analysis to compare studies of mice with single-gene defects encountered major limitations. To overcome these, we developed a novel synthesis approach to quantitatively scale 1514 published studies of arterial thrombus formation (in vivo and in vitro), thromboembolism, and tail-bleeding of genetically modified mice. Using a newly defined consistency parameter (CP), indicating the strength of published data, comparisons were made of 431 mouse genes, of which 17 consistently contributed to thrombus formation without affecting hemostasis. Ranking analysis indicated high correlations between collagen-dependent thrombosis models in vivo (FeCl3 injury or ligation/compression) and in vitro. Integration of scores and CP values resulted in a network of protein interactions in thrombosis and hemostasis (PITH), which was combined with databases of genetically linked human bleeding and thrombotic disorders. The network contained 2946 nodes linked to modifying genes of thrombus formation, mostly with expression in megakaryocytes. Reactome pathway analysis and network characteristics revealed multiple novel genes with potential contribution to thrombosis/hemostasis. Studies with additional knockout mice revealed that 4 of 8 (Apoe, Fpr2, Ifnar1, Vps13a) new genes were modifying in thrombus formation. The PITH network further: (i) revealed a high similarity of murine and human hemostatic and thrombotic processes and (ii) identified multiple new candidate proteins regulating these processes.


Subject(s)
Hemorrhage , Thrombosis , Animals , Disease Models, Animal , Hemorrhage/genetics , Hemorrhage/metabolism , Hemorrhage/pathology , Humans , Mice , Mice, Knockout , Thrombosis/genetics , Thrombosis/metabolism , Thrombosis/pathology
4.
Blood ; 130(26): 2819-2828, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29018081

ABSTRACT

Traditionally, in vitro flow chamber experiments and in vivo arterial thrombosis studies have been proved to be of vital importance to elucidate the mechanisms of platelet thrombus formation after vessel wall injury. In recent years, it has become clear that platelets also act as modulators of inflammatory processes, such as atherosclerosis. A key element herein is the complex cross talk between platelets, the coagulation system, leukocytes, and the activated endothelium. This review provides insight into the platelet-endothelial interface, based on in vitro flow chamber studies and cross referenced with in vivo thrombosis studies. The main mechanisms of platelet interaction with the activated endothelium encompass (1) platelet rolling via interaction of platelet glycoprotein Ib-IX-V with endothelial-released von Willebrand factor with a supporting role for the P-selectin/P-selectin glycoprotein ligand 1 axis, followed by (2) firm platelet adhesion to the endothelium via interaction of platelet αIIbß3 with endothelial αvß3 and intercellular adhesion molecule 1, and (3) a stimulatory role for thrombin, the thrombospondin-1/CD36 axis and cyclooxygenase 1 in subsequent platelet activation and stable thrombus formation. In addition, the molecular mechanisms underlying the stimulatory effect of platelets on leukocyte transendothelial migration, a key mediator of atheroprogression, are discussed. Throughout the review, emphasis is placed on recommendations for setting up, reporting, interpreting, and comparing endothelial-lined flow chamber studies and suggestions for future studies.


Subject(s)
Blood Platelets/metabolism , Endothelium, Vascular/metabolism , Microfluidics/methods , Transendothelial and Transepithelial Migration , Cell Communication/physiology , Humans , Microfluidics/trends , Receptor Cross-Talk/physiology
5.
Haematologica ; 103(3): 540-549, 2018 03.
Article in English | MEDLINE | ID: mdl-29242293

ABSTRACT

In patients with dysfunctions of the Ca2+ channel ORAI1, stromal interaction molecule 1 (STIM1) or integrin-regulating kindlin-3 (FERMT3), severe immunodeficiency is frequently linked to abnormal platelet activity. In this paper, we studied platelet responsiveness by multiparameter assessment of whole blood thrombus formation under high-shear flow conditions in 9 patients, including relatives, with confirmed rare genetic mutations of ORAI1, STIM1 or FERMT3. In platelets isolated from 5 out of 6 patients with ORAI1 or STIM1 mutations, store-operated Ca2+ entry (SOCE) was either completely or partially defective compared to control platelets. Parameters of platelet adhesion and aggregation on collagen microspots were impaired for 4 out of 6 patients, in part related to a low platelet count. For 4 patients, platelet adhesion/aggregation and procoagulant activity on von Willebrand Factor (VWF)/rhodocytin and VWF/fibrinogen microspots were impaired independently of platelet count, and were partly correlated with SOCE deficiency. Measurement of thrombus formation at low shear rate confirmed a greater impairment of platelet functionality in the ORAI1 patients than in the STIM1 patient. For 3 patients/relatives with a FERMT3 mutation, all parameters of thrombus formation were strongly reduced regardless of the microspot. Bone marrow transplantation, required by 2 patients, resulted in overall improvement of platelet function. We concluded that multiparameter assessment of whole blood thrombus formation in a surface-dependent way can detect: i) additive effects of low platelet count and impaired platelet functionality; ii) aberrant ORAI1-mediated Ca2+ entry; iii) differences in platelet activation between patients carrying the same ORAI1 mutation; iv) severe platelet function impairment linked to a FERMT3 mutation and bleeding history.


Subject(s)
Immunologic Deficiency Syndromes/blood , Platelet Activation/genetics , Calcium/metabolism , Humans , Immunologic Deficiency Syndromes/genetics , Membrane Proteins/genetics , Mutation , Neoplasm Proteins/genetics , ORAI1 Protein/genetics , Platelet Adhesiveness , Platelet Aggregation , Platelet Function Tests , Stromal Interaction Molecule 1/genetics , Thrombosis/etiology
6.
Mol Cell Proteomics ; 15(10): 3154-3169, 2016 10.
Article in English | MEDLINE | ID: mdl-27535140

ABSTRACT

The Scott syndrome is a very rare and likely underdiagnosed bleeding disorder associated with mutations in the gene encoding anoctamin-6. Platelets from Scott patients are impaired in various Ca2+-dependent responses, including phosphatidylserine exposure, integrin closure, intracellular protein cleavage, and cytoskeleton-dependent morphological changes. Given the central role of anoctamin-6 in the platelet procoagulant response, we used quantitative proteomics to understand the underlying molecular mechanisms and the complex phenotypic changes in Scott platelets compared with control platelets. Therefore, we applied an iTRAQ-based multi-pronged strategy to quantify changes in (1) the global proteome, (2) the phosphoproteome, and (3) proteolytic events between resting and stimulated Scott and control platelets. Our data indicate a limited number of proteins with decreased (70) or increased (64) expression in Scott platelets, among those we confirmed the absence of anoctamin-6 and the strong up-regulation of aquaporin-1 by parallel reaction monitoring. The quantification of 1566 phosphopeptides revealed major differences between Scott and control platelets after stimulation with thrombin/convulxin or ionomycin. In Scott platelets, phosphorylation levels of proteins regulating cytoskeletal or signaling events were increased. Finally, we quantified 1596 N-terminal peptides in activated Scott and control platelets, 180 of which we identified as calpain-regulated, whereas a distinct set of 23 neo-N termini was caspase-regulated. In Scott platelets, calpain-induced cleavage of cytoskeleton-linked and signaling proteins was downregulated, in accordance with an increased phosphorylation state. Thus, multipronged proteomic profiling of Scott platelets provides detailed insight into their protection against detrimental Ca2+-dependent changes that are normally associated with phosphatidylserine exposure.


Subject(s)
Blood Coagulation Disorders/blood , Blood Platelets/physiology , Phosphoproteins/analysis , Proteomics/methods , Blood Coagulation Disorders/metabolism , Blood Platelets/drug effects , Calcium/metabolism , Crotalid Venoms/pharmacology , Gene Expression Regulation , Humans , Ionomycin/pharmacology , Lectins, C-Type , Phosphoproteins/drug effects , Proteolysis , Signal Transduction , Thrombin/pharmacology
8.
FASEB J ; 30(2): 727-37, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26481309

ABSTRACT

Scott syndrome is a rare bleeding disorder, characterized by altered Ca(2+)-dependent platelet signaling with defective phosphatidylserine (PS) exposure and microparticle formation, and is linked to mutations in the ANO6 gene, encoding anoctamin (Ano)6. We investigated how the complex platelet phenotype of this syndrome is linked to defective expression of Anos or other ion channels. Mice were generated with heterozygous of homozygous deficiency in Ano6, Ano1, or Ca(2+)-dependent KCa3.1 Gardos channel. Platelets from these mice were extensively analyzed on molecular functions and compared with platelets from a patient with Scott syndrome. Deficiency in Ano1 or Gardos channel did not reduce platelet responses compared with control mice (P > 0.1). In 2 mouse strains, deficiency in Ano6 resulted in reduced viability with increased bleeding time to 28.6 min (control 6.4 min, P < 0.05). Platelets from the surviving Ano6-deficient mice resembled platelets from patients with Scott syndrome in: 1) normal collagen-induced aggregate formation (P > 0.05) with reduced PS exposure (-65 to 90%); 2) lowered Ca(2+)-dependent swelling (-80%) and membrane blebbing (-90%); 3) reduced calpain-dependent protein cleavage (-60%); and 4) moderately affected apoptosis-dependent PS exposure. In conclusion, mouse deficiency of Ano6 but not of other channels affects viability and phenocopies the complex changes in platelets from hemostatically impaired patients with Scott syndrome.


Subject(s)
Blood Coagulation Disorders/metabolism , Blood Platelets/metabolism , Phospholipid Transfer Proteins/metabolism , Phospholipids/metabolism , Proteolysis , Animals , Anoctamin-1 , Anoctamins , Blood Coagulation Disorders/genetics , Blood Coagulation Disorders/pathology , Blood Platelets/pathology , Calcium/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Cell Membrane/pathology , Chloride Channels/genetics , Chloride Channels/metabolism , Female , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Mice , Mice, Knockout , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipids/genetics
9.
Circ Res ; 116(4): 587-99, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25472975

ABSTRACT

RATIONALE: Besides their essential role in hemostasis, platelets also have functions in inflammation. In platelets, junctional adhesion molecule (JAM)-A was previously identified as an inhibitor of integrin αIIbß3-mediated outside-in signaling and its genetic knockdown resulted in hyperreactivity. OBJECTIVE: This gain-of-function was specifically exploited to investigate the role of platelet hyperreactivity in plaque development. METHODS AND RESULTS: JAM-A-deficient platelets showed increased aggregation and cellular and sarcoma tyrosine-protein kinase activation. On αIIbß3 ligation, JAM-A was shown to be dephosphorylated, which could be prevented by protein tyrosine phosphatase nonreceptor type 1 inhibition. Mice with or without platelet-specific (tr)JAM-A-deficiency in an apolipoprotein e (apoe(-/-)) background were fed a high-fat diet. After ≤12 weeks of diet, trJAM-A(-/-)apoe-/- mice showed increased aortic plaque formation when compared with trJAM-A(+/+) apoe(-/-) controls, and these differences were most evident at early time points. At 2 weeks, the plaques of the trJAM-A(-/-) apoe(-/-) animals revealed increased macrophage, T cell, and smooth muscle cell content. Interestingly, plasma levels of chemokines CC chemokine ligand 5 and CXC-chemokine ligand 4 were increased in the trJAM-A(-/-) apoe(-/-)mice, and JAM-A-deficient platelets showed increased binding to monocytes and neutrophils. Whole-blood perfusion experiments and intravital microscopy revealed increased recruitment of platelets and monocytes to the inflamed endothelium in blood of trJAM-A(-/-) apoe(-/-)mice. Notably, these proinflammatory effects of JAM-A-deficient platelets could be abolished by the inhibition of αIIbß3 signaling in vitro. CONCLUSIONS: Deletion of JAM-A causes a gain-of-function in platelets, with lower activation thresholds and increased inflammatory activities. This leads to an increase of plaque formation, particularly in early stages of the disease.


Subject(s)
Aorta/metabolism , Aortic Diseases/etiology , Atherosclerosis/etiology , Blood Platelets/metabolism , Carotid Artery Diseases/etiology , Cell Adhesion Molecules/deficiency , Hyperlipidemias/complications , Platelet Aggregation , Receptors, Cell Surface/deficiency , Animals , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Artery Diseases/blood , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Cell Adhesion , Cell Adhesion Molecules/blood , Cell Adhesion Molecules/genetics , Cells, Cultured , Chemotaxis, Leukocyte , Diet, High-Fat , Disease Models, Animal , Disease Progression , Female , Genotype , Humans , Hyperlipidemias/blood , Hyperlipidemias/genetics , Inflammation Mediators/metabolism , Leukocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plaque, Atherosclerotic , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Receptors, Cell Surface/blood , Receptors, Cell Surface/genetics , Thrombosis/blood , Thrombosis/etiology , Time Factors , src-Family Kinases/metabolism
10.
Arterioscler Thromb Vasc Biol ; 36(4): 692-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26848157

ABSTRACT

OBJECTIVE: Platelet- and fibrin-dependent thrombus formation is regulated by blood flow and exposure of collagen and tissue factor. However, interactions between these blood-borne and vascular components are not well understood. APPROACH AND RESULTS: Here, we developed a method to assess whole-blood thrombus formation on microspots with defined amounts of collagen and tissue factor, allowing determination of the mechanical properties and intrathrombus composition. Confining the collagen content resulted in diminished platelet deposition and fibrin formation at high shear flow conditions, but this effect was compensated by a larger thrombus size and increased accumulation of fibrin in the luminal regions of the thrombi at the expense of the base regions. These thrombi were more dependent on tissue factor-triggered thrombin generation. Microforce nanoindentation analysis revealed a significantly increased microelasticity of thrombi with luminal-oriented fibrin. At a low shear rate, fibrin fibers tended to luminally cover the thrombi, again resulting in a higher microelasticity. Studies with blood from patients with distinct hemostatic insufficiencies indicated an impairment in the formation of a platelet-fibrin thrombus in the cases of dilutional coagulopathy, thrombocytopenia, Scott syndrome, and hemophilia B. CONCLUSIONS: Taken together, our data indicate that (1) thrombin increases the platelet thrombus volume; (2) tissue factor drives the formation of fibrin outside of the platelet thrombus; (3) limitation of platelet adhesion redirects fibrin from bottom to top of the thrombus; (4) a lower shear rate promotes thrombus coverage with fibrin; (5) the fibrin distribution pattern determines thrombus microelasticity; and (6) the thrombus-forming process is reduced in patients with diverse hemostatic defects.


Subject(s)
Blood Coagulation , Blood Platelets/metabolism , Fibrin/metabolism , Thrombosis/blood , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/physiopathology , Blood Coagulation Tests , Blood Flow Velocity , Case-Control Studies , Collagen/blood , Elasticity , Hemophilia B/blood , Hemophilia B/physiopathology , Humans , Regional Blood Flow , Thrombocytopenia/blood , Thrombocytopenia/physiopathology , Thromboplastin/metabolism , Thrombosis/physiopathology , Time Factors
11.
Circulation ; 132(15): 1414-24, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26330411

ABSTRACT

BACKGROUND: Platelets are central to the process of hemostasis, rapidly aggregating at sites of blood vessel injury and acting as coagulation nidus sites. On interaction with the subendothelial matrix, platelets are transformed into balloonlike structures as part of the hemostatic response. It remains unclear, however, how and why platelets generate these structures. We set out to determine the physiological relevance and cellular and molecular mechanisms underlying platelet membrane ballooning. METHODS AND RESULTS: Using 4-dimensional live-cell imaging and electron microscopy, we show that human platelets adherent to collagen are transformed into phosphatidylserine-exposing balloonlike structures with expansive macro/microvesiculate contact surfaces, by a process that we termed procoagulant spreading. We reveal that ballooning is mechanistically and structurally distinct from membrane blebbing and involves disruption to the platelet microtubule cytoskeleton and inflation through fluid entry. Unlike blebbing, procoagulant ballooning is irreversible and a consequence of Na(+), Cl(-), and water entry. Furthermore, membrane ballooning correlated with microparticle generation. Inhibition of Na(+), Cl(-), or water entry impaired ballooning, procoagulant spreading, and microparticle generation, and it also diminished local thrombin generation. Human Scott syndrome platelets, which lack expression of Ano-6, also showed a marked reduction in membrane ballooning, consistent with a role for chloride entry in the process. Finally, the blockade of water entry by acetazolamide attenuated ballooning in vitro and markedly suppressed thrombus formation in vivo in a mouse model of thrombosis. CONCLUSIONS: Ballooning and procoagulant spreading of platelets are driven by fluid entry into the cells, and are important for the amplification of localized coagulation in thrombosis.


Subject(s)
Blood Platelets/ultrastructure , Acetazolamide/pharmacology , Actomyosin/metabolism , Amides/pharmacology , Animals , Anoctamins , Blood Coagulation Disorders/blood , Blood Platelets/drug effects , Blood Platelets/metabolism , Carotid Artery Thrombosis/blood , Carotid Artery Thrombosis/chemically induced , Carotid Artery Thrombosis/drug therapy , Cell Adhesion , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Shape/drug effects , Cell Shape/physiology , Cell Size/drug effects , Cell-Derived Microparticles , Chlorides/metabolism , Collagen , Cytochalasin D/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Mice , Microtubules/drug effects , Phospholipid Transfer Proteins/deficiency , Phospholipid Transfer Proteins/physiology , Pyridines/pharmacology , Sodium/metabolism , Thrombin/biosynthesis , Thrombosis/prevention & control , Water/metabolism
12.
Haematologica ; 101(4): 427-36, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26721892

ABSTRACT

Coated platelets, formed by collagen and thrombin activation, have been characterized in different ways: i) by the formation of a protein coat of α-granular proteins; ii) by exposure of procoagulant phosphatidylserine; or iii) by high fibrinogen binding. Yet, their functional role has remained unclear. Here we used a novel transglutaminase probe, Rhod-A14, to identify a subpopulation of platelets with a cross-linked protein coat, and compared this with other platelet subpopulations using a panel of functional assays. Platelet stimulation with convulxin/thrombin resulted in initial integrin α(IIb)ß3 activation, the appearance of a platelet population with high fibrinogen binding, (independently of active integrins, but dependent on the presence of thrombin) followed by phosphatidylserine exposure and binding of coagulation factors Va and Xa. A subpopulation of phosphatidylserine-exposing platelets bound Rhod-A14 both in suspension and in thrombi generated on a collagen surface. In suspension, high fibrinogen and Rhod-A14 binding were antagonized by combined inhibition of transglutaminase activity and integrin α(IIb)ß3 Markedly, in thrombi from mice deficient in transglutaminase factor XIII, platelet-driven fibrin formation and Rhod-A14 binding were abolished by blockage of integrin α(IIb)ß3. Vice versa, star-like fibrin formation from platelets of a patient with deficiency in α(IIb)ß3(Glanzmann thrombasthenia) was abolished upon blockage of transglutaminase activity. We conclude that coated platelets, with initial α(IIb)ß3 activation and high fibrinogen binding, form a subpopulation of phosphatidylserine-exposing platelets, and function in platelet-dependent star-like fibrin fiber formation via transglutaminase factor XIII and integrin α(IIb)ß3.


Subject(s)
Blood Platelets/metabolism , Factor XIII/metabolism , Fibrin/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombasthenia/blood , Animals , Blood Coagulation , Blood Platelets/drug effects , Blood Platelets/pathology , Crotalid Venoms/pharmacology , Factor Va/chemistry , Factor Va/metabolism , Factor XIII/chemistry , Factor Xa/chemistry , Factor Xa/metabolism , Fibrin/chemistry , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Lectins, C-Type , Mice , Mice, Knockout , Molecular Probes/chemistry , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Platelet Activation/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Primary Cell Culture , Protein Binding , Thrombasthenia/pathology , Thrombin/pharmacology
13.
Arterioscler Thromb Vasc Biol ; 35(12): 2554-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26471268

ABSTRACT

OBJECTIVE: Platelets are increasingly implicated in processes beyond hemostasis and thrombosis, such as vascular remodeling. Members of the matrix metalloproteinase (MMP) family not only remodel the extracellular matrix but also modulate platelet function. Here, we made a systematic comparison of the roles of MMP family members in acute thrombus formation under flow conditions and assessed platelet-dependent collagenolytic activity over time. APPROACH AND RESULTS: Pharmacological inhibition of MMP-1 or MMP-2 (human) or deficiency in MMP-2 (mouse) suppressed collagen-dependent platelet activation and thrombus formation under flow, whereas MMP-9 inhibition/deficiency stimulated these processes. The absence of MMP-3 was without effect. Interestingly, MMP-14 inhibition led to the formation of larger thrombi, which occurred independently of its capacity to activate MMP-2. Platelet thrombi exerted local collagenolytic activity capable of cleaving immobilized dye-quenched collagen and fibrillar collagen fibers within hours, with loss of the majority of the platelet adhesive properties of collagen as a consequence. This collagenolytic activity was redundantly mediated by platelet-associated MMP-1, MMP-2, MMP-9, and MMP-14 but occurred independently of platelet α-granule release (Nbeal2(-/-) mice). The latter was in line with subcellular localization experiments, which indicated a granular distribution of MMP-1 and MMP-2 in platelets, distinct from α-granules. Whereas MMP-9 protein could not be detected inside platelets, activated platelets did bind plasma-derived MMP-9 to their plasma membrane. Overall, platelet MMP activity was predominantly membrane-associated and influenced by platelet activation status. CONCLUSIONS: Platelet-associated MMP-1, MMP-2, MMP-9, and MMP-14 differentially modulate acute thrombus formation and at later time points limit thrombus formation by exerting collagenolytic activity.


Subject(s)
Blood Platelets/enzymology , Collagen/metabolism , Collagenases/blood , Thrombosis/enzymology , Animals , Blood Platelets/drug effects , Blood Proteins/genetics , Blood Proteins/metabolism , Collagenases/deficiency , Collagenases/genetics , Disease Models, Animal , Humans , Matrix Metalloproteinase Inhibitors/pharmacology , Mice, Inbred C57BL , Mice, Knockout , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , Proteolysis , Thrombosis/blood , Thrombosis/genetics , Time Factors
14.
Arterioscler Thromb Vasc Biol ; 35(6): 1374-81, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25908768

ABSTRACT

OBJECTIVE: To investigate the roles and signaling pathways of CD40L and CD40 in platelet-platelet interactions and thrombus formation under conditions relevant for atherothrombosis. APPROACH AND RESULTS: Platelets from mice prone to atherosclerosis lacking CD40L (Cd40lg(-/-)Apoe(-/-)) showed diminished αIIbß3 activation and α-granule secretion in response to glycoprotein VI stimulation, whereas these responses of CD40-deficient platelets (Cd40(-/-)Apoe(-/-)) were not decreased. Using blood from Cd40lg(-/-)Apoe(-/-) and Cd40(-/-)Apoe(-/-) mice, the glycoprotein VI-dependent formation of dense thrombi was impaired on atherosclerotic plaque material or on collagen, in comparison with Apoe(-/-) blood. In all genotypes, addition of CD40L to the blood enhanced the growth of dense thrombi on plaques and collagen. Similarly, CD40L enhanced glycoprotein VI-induced platelet aggregation, even with platelets deficient in CD40. This potentiation was antagonized in Pik3cb(R/R) platelets or by inhibiting phosphatidylinositol 3-kinase ß (PI3Kß). Addition of CD40L also enhanced collagen-induced Akt phosphorylation, which was again antagonized by absence or inhibition of PI3Kß. Finally, platelets from Chuk1(A/A)Apoe(-/-) mice deficient in IκB kinase α (IKKα), implicated in CD40 signaling to nuclear factor (NF) κB, showed unchanged responses to CD40L in aggregation or thrombus formation. CONCLUSIONS: Under atherogenic conditions, CD40L enhances collagen-induced platelet-platelet interactions by supporting integrin αIIbß3 activation, secretion and thrombus growth via PI3Kß, but not via CD40 and IKKα/NFκB. This role of CD40L exceeds the no more than modest role of CD40 in thrombus formation.


Subject(s)
Atherosclerosis/metabolism , Blood Platelets/metabolism , CD40 Antigens/metabolism , CD40 Ligand/metabolism , I-kappa B Kinase/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Thrombosis/metabolism , Animals , Atherosclerosis/pathology , Collagen/metabolism , Mice , Platelet Activation , Signal Transduction , Thrombosis/pathology
15.
Arterioscler Thromb Vasc Biol ; 34(8): 1674-80, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24855058

ABSTRACT

OBJECTIVE: Atherothrombosis is the main cause of myocardial infarction and ischemic stroke. Although the extrinsic (tissue factor-factor VIIa [FVIIa]) pathway is considered as a major trigger of coagulation in atherothrombosis, the role of the intrinsic coagulation pathway via coagulation FXII herein is unknown. Here, we studied the roles of the extrinsic and intrinsic coagulation pathways in thrombus formation on atherosclerotic plaques both in vivo and ex vivo. APPROACH AND RESULTS: Plaque rupture after ultrasound treatment evoked immediate formation of subocclusive thrombi in the carotid arteries of Apoe(-/-) mice, which became unstable in the presence of structurally different FXIIa inhibitors. In contrast, inhibition of FVIIa reduced thrombus size at a more initial stage without affecting embolization. Genetic deficiency in FXII (human and mouse) or FXI (mouse) reduced ex vivo whole-blood thrombus and fibrin formation on immobilized plaque homogenates. Localization studies by confocal microscopy indicated that FXIIa bound to thrombi and fibrin particularly in luminal-exposed thrombus areas. CONCLUSIONS: The FVIIa- and FXIIa-triggered coagulation pathways have distinct but complementary roles in atherothrombus formation. The tissue factor-FVIIa pathway contributes to initial thrombus buildup, whereas FXIIa bound to thrombi ensures thrombus stability.


Subject(s)
Aortic Diseases/complications , Atherosclerosis/complications , Blood Coagulation , Blood Platelets/metabolism , Carotid Artery Diseases/complications , Factor XII/metabolism , Plaque, Atherosclerotic , Thrombosis/etiology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Diseases/blood , Aortic Diseases/genetics , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Diseases/blood , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Cholesterol, Dietary , Disease Models, Animal , Factor VIIa/metabolism , Factor XI/metabolism , Factor XII/genetics , Factor XII Deficiency/blood , Factor XII Deficiency/genetics , Factor XIIa/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Rupture, Spontaneous , Thromboplastin/metabolism , Thrombosis/blood , Thrombosis/genetics , Thrombosis/pathology , Time Factors
16.
Arterioscler Thromb Vasc Biol ; 34(6): 1187-92, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24675658

ABSTRACT

OBJECTIVE: Platelets abundantly express the membrane receptor CD36 and store its ligand thrombospondin-1 (TSP1) in the α-granules. We investigated whether released TSP1 can support platelet adhesion and thrombus formation via interaction with CD36. APPROACH AND RESULTS: Mouse platelets deficient in CD36 showed reduced adhesion to TSP1 and subsequent phosphatidylserine expression. Deficiency in either CD36 or TSP1 resulted in markedly increased dissolution of thrombi formed on collagen, although thrombus buildup was unchanged. In mesenteric vessels in vivo, deficiency in CD36 prolonged the time to occlusion and enhanced embolization, which was in agreement with earlier observations in TSP1-deficient mice. Thrombi formed using wild-type blood stained positively for secreted TSP1. Releasate from wild-type but not from TSP1-deficient platelets enhanced platelet activation, phosphatidylserine expression, and thrombus formation on collagen. The enhancement was dependent on CD36 because it was without effect on thrombus formation by CD36-deficient platelets. CONCLUSIONS: These results demonstrate an anchoring role of platelet-released TSP1 via CD36 in platelet adhesion and collagen-dependent thrombus stabilization. Thus, the TSP1-CD36 tandem is another platelet ligand-receptor axis contributing to the maintenance of a stable thrombus.


Subject(s)
CD36 Antigens/physiology , Collagen/metabolism , Thrombosis/etiology , Thrombospondin 1/physiology , Animals , Mice , Mice, Inbred C57BL , Platelet Activation , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/physiology
17.
Arterioscler Thromb Vasc Biol ; 34(1): 61-71, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24233488

ABSTRACT

OBJECTIVE: Platelet activation is essential for primary hemostasis and acute thrombotic vascular occlusions. On activation, platelets release their prothrombotic granules and expose phosphatidylserine, thus fostering thrombin generation and thrombus formation. In other cell types, both degranulation and phosphatidylserine exposure are modified by sphingomyelinase-dependent formation of ceramide. The present study thus explored whether acid sphingomyelinase participates in the regulation of platelet secretion, phosphatidylserine exposure, and thrombus formation. APPROACH AND RESULTS: Collagen-related peptide-induced or thrombin-induced ATP release and P-selectin exposure were significantly blunted in platelets from Asm-deficient mice (Smpd1(-/-)) when compared with platelets from wild-type mice (Smpd1(+/+)). Moreover, phosphatidylserine exposure and thrombin generation were significantly less pronounced in Smpd1(-/-) platelets than in Smpd1(+/+) platelets. In contrast, platelet integrin αIIbß3 activation and aggregation, as well as activation-dependent Ca(2+) flux, were not significantly different between Smpd1(-/-) and Smpd1(+/+) platelets. In vitro thrombus formation at shear rates of 1700 s(-1) and in vivo thrombus formation after FeCl3 injury were significantly blunted in Smpd1(-/-) mice while bleeding time was unaffected. Asm-deficient platelets showed significantly reduced activation-dependent ceramide formation, whereas exogenous ceramide rescued diminished platelet secretion and thrombus formation caused by Asm deficiency. Treatment of Smpd1(+/+) platelets with bacterial sphingomyelinase (0.01 U/mL) increased, whereas treatment with functional acid sphingomyelinase-inhibitors, amitriptyline or fluoxetine (5 µmol/L), blunted activation-dependent platelet degranulation, phosphatidylserine exposure, and thrombus formation. Impaired degranulation and thrombus formation of Smpd1(-/-) platelets were again overcome by exogenous bacterial sphingomyelinase. CONCLUSIONS: Acid sphingomyelinase is a completely novel element in the regulation of platelet plasma membrane properties, secretion, and thrombus formation.


Subject(s)
Blood Platelets/enzymology , Cell Degranulation , Cell Membrane/enzymology , Platelet Activation , Sphingomyelin Phosphodiesterase/blood , Thrombosis/enzymology , Adenosine Triphosphate/blood , Animals , Blood Platelets/drug effects , Calcium/blood , Cell Degranulation/drug effects , Cell Membrane/drug effects , Ceramides/blood , Chlorides , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Ferric Compounds , Fibrinolytic Agents/pharmacology , Male , Mice , Mice, Knockout , P-Selectin/blood , Phosphatidylserines/blood , Platelet Activation/drug effects , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Thrombin/metabolism , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/genetics , Thrombosis/prevention & control , Time Factors
18.
J Biol Chem ; 288(19): 13325-36, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23519467

ABSTRACT

BACKGROUND: Inactivation of integrin αIIbß3 reverses platelet aggregate formation upon coagulation. RESULTS AND CONCLUSION: Platelets from patient (Scott) and mouse (Capn1(-/-) and Ppif(-/-)) blood reveal a dual mechanism of αIIbß3 inactivation: by calpain-2 cleavage of integrin-associated proteins and by cyclophilin D/TMEM16F-dependent phospholipid scrambling. SIGNIFICANCE: These data provide novel insight into the switch mechanisms from aggregating to procoagulant platelets. Aggregation of platelets via activated integrin αIIbß3 is a prerequisite for thrombus formation. Phosphatidylserine-exposing platelets with a key role in the coagulation process disconnect from a thrombus by integrin inactivation via an unknown mechanism. Here we show that αIIbß3 inactivation in procoagulant platelets relies on a sustained high intracellular Ca(2+), stimulating intracellular cleavage of the ß3 chain, talin, and Src kinase. Inhibition of calpain activity abolished protein cleavage, but only partly suppressed αIIbß3 inactivation. Integrin αIIbß3 inactivation was unchanged in platelets from Capn1(-/-) mice, suggesting a role of the calpain-2 isoform. Scott syndrome platelets, lacking the transmembrane protein TMEM16F and having low phosphatidylserine exposure, displayed reduced αIIbß3 inactivation with the remaining activity fully dependent on calpain. In platelets from Ppif(-/-) mice, lacking mitochondrial permeability transition pore (mPTP) formation, agonist-induced phosphatidylserine exposure and αIIbß3 inactivation were reduced. Treatment of human platelets with cyclosporin A gave a similar phenotype. Together, these data point to a dual mechanism of αIIbß3 inactivation via calpain(-2) cleavage of integrin-associated proteins and via TMEM16F-dependent phospholipid scrambling with an assistant role of mPTP formation.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Animals , Anoctamins , Blood Platelets/drug effects , Blood Platelets/physiology , CD36 Antigens/metabolism , Calcium Signaling , Calpain/antagonists & inhibitors , Calpain/metabolism , Cell Membrane/metabolism , Crotalid Venoms/pharmacology , Dipeptides/pharmacology , Humans , Lectins, C-Type , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Platelet Aggregation , Protein Structure, Quaternary , Proteolysis , Talin/metabolism , Thrombin/pharmacology , Thrombin/physiology , src-Family Kinases/metabolism
19.
Thromb Res ; 236: 179-190, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38460307

ABSTRACT

Endothelialized in vitro models for cardiovascular disease have contributed greatly to our current understanding of the complex molecular mechanisms underlying thrombosis. To further elucidate these mechanisms, it is important to consider which fundamental aspects to incorporate into an in vitro model. In this review, we will focus on the design of in vitro endothelialized models of thrombosis. Expanding our understanding of the relation and interplay between the different pathways involved will rely in part on complex models that incorporate endothelial cells, blood, the extracellular matrix, and flow. Importantly, the use of tissue-specific endothelial cells will help in understanding the heterogeneity in thrombotic responses between different vascular beds. The dynamic and complex responses of endothelial cells to different shear rates underlines the importance of incorporating appropriate shear in in vitro models. Alterations in vascular extracellular matrix composition, availability of bioactive molecules, and gradients in concentration and composition of these molecules can all regulate the function of both endothelial cells and perivascular cells. Factors modulating these elements in in vitro models should therefore be considered carefully depending on the research question at hand. As the complexity of in vitro models increases, so can the variability. A bottom-up approach to designing such models will remain an important tool for researchers studying thrombosis. As new techniques are continuously being developed and new pathways are brought to light, research question-dependent considerations will have to be made regarding what aspects of thrombosis to include in in vitro models.


Subject(s)
Endothelial Cells , Thrombosis , Humans , Endothelial Cells/metabolism , Endothelium, Vascular , Thrombosis/metabolism
20.
Semin Thromb Hemost ; 39(3): 306-14, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23378253

ABSTRACT

Multimeric glycoprotein von Willebrand factor (VWF) exhibits a unique triplet structure of individual oligomers, resulting from ADAMTS-13 (a disintegrin and metalloproteinase with thrombospondin type 1 motifs 13) cleavage. The faster and slower migrating triplet bands of a given VWF multimer have one shorter or longer N-terminal peptide sequence, respectively. Within this peptide sequence, the A1 domain regulates interaction of VWF with platelet glycoprotein (GP)Ib. Therefore, platelet-adhesive properties of two VWF preparations with similar multimeric distribution but different triplet composition were investigated for differential functional activities. Preparation A was enriched in intermediate triplet bands, whereas preparation B predominantly contained larger triplet bands. Binding studies revealed that preparation A displayed a reduced affinity for recombinant GPIb but an unchanged affinity for collagen type III when compared to preparation B. Under high-shear flow conditions, preparation A was less active in recruiting platelets to collagen type III. Furthermore, when added to blood from patients with von Willebrand disease (VWD), defective thrombus formation was less restored. Thus, VWF forms lacking larger-size triplet bands appear to have a decreased potential to recruit platelets to collagen-bound VWF under arterial flow conditions. By implication, changes in triplet band distribution observed in patients with VWD may result in altered platelet adhesion at high-shear flow.


Subject(s)
Blood Platelets/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombosis/blood , von Willebrand Factor/metabolism , Blood Platelets/cytology , Enzyme-Linked Immunosorbent Assay , Humans , Platelet Adhesiveness/physiology , Surface Plasmon Resonance , Thrombosis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL