Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters











Publication year range
1.
TH Open ; 7(4): e294-e302, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37964899

ABSTRACT

Introduction Atrial fibrillation (AF) increases the risk of ischemic stroke (IS). We hypothesized that the functional form of platelet receptor glycoprotein (GP) VI, GPVI-dimer, which binds to collagen and fibrin causing platelet activation, is overexpressed in patients with AF who have not had a stroke. Methods A total of 75 inpatients with AF were recruited. None were admitted with or had previously had thrombotic events, including IS or myocardial infarction. Platelet surface expression of total GPVI, GPVI-dimer, and the platelet activation marker P-selectin were quantitated by whole blood flow cytometry. Serum biomarkers were collected in AF patients. Results were compared against patients contemporaneously admitted to hospital with similar age and vascular risk-factor profiles without AF (noAF, n = 30). Results Patients with AF have similar total GPVI surface expression ( p = 0.58) and P-selectin exposure ( p = 0.73) on their platelets compared with noAF patients but demonstrate significantly higher GPVI-dimer expression ( p = 0.02 ). Patients with paroxysmal AF express similar GPVI-dimer levels compared with permanent AF and GPVI-dimer levels were not different between anticoagulated groups. Serum N-terminal pro b-type natriuretic peptide ( p < 0.0001 ) and high sensitivity C-reactive protein ( p < 0.0001 ) were significantly correlated with GPVI-dimer expression in AF platelets. AF was the only vascular risk factor that was independently associated with higher GPVI-dimer expression in the whole population ( p = 0.02 ) . Conclusion GPVI inhibition is being explored in clinical trials as a novel target for IS treatment. As GPVI-dimer is elevated in AF patients' platelets, the exploration of targeted GPVI-dimer inhibition for stroke prevention in patients at high risk of IS due to AF is supported.

2.
Res Pract Thromb Haemost ; 6(3): e12697, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35494504

ABSTRACT

Background: In the fibrin-forming process, thrombin cleaves fibrinogen to fibrin, which form fibrils and then fibers, producing a gel-like clot. Thrombin also activates coagulation factor XIII (FXIII), which crosslinks fibrin γ-chains and α-chains, stabilizing the clot. Many proteins bind to fibrin, including FXIII, an established regulation of clot structure, and platelet glycoprotein VI (GPVI), whose contribution to clot function is largely unknown. FXIII is present in plasma, but the abundant FXIII in platelet cytosol becomes exposed to the surface of strongly activated platelets. Objectives: We determined if GPVI interacts with FXIII and how this might modulate clot formation. Methods: We measured interactions between recombinant proteins of the GPVI extracellular domain: GPVI-dimer (GPVI-Fc2) or monomer (GPVIex) and FXIII proteins (nonactivated and thrombin-activated FXIII, FXIII subunits A and B) by ELISA. Binding to fibrin clots and fibrin γ-chain crosslinking were analyzed by immunoblotting. Results: GPVI-dimer, but not GPVI-monomer, bound to FXIII. GPVI-dimer selectively bound to the FXIII A-subunit, but not to the B-subunit, an interaction that was decreased or abrogated by the GPVI-dimer-specific antibody mFab-F. The GPVI-dimer-FXIII interaction decreased the extent of γ-chain crosslinking, indicating a role in the regulation of clot formation. Conclusions: This is the first report of the specific interaction between GPVI-dimer and the A-subunit of FXIII, as determined in an in vitro system with defined components. GPVI-dimer-FXIII binding was inhibitory toward FXIII-catalyzed crosslinking of fibrin γ-chains in fibrin clots. This raises the possibility that GPVI-dimer may negatively modulate fibrin crosslinking induced by FXIII, lessening clot stability.

3.
PLoS One ; 17(1): e0262695, 2022.
Article in English | MEDLINE | ID: mdl-35041713

ABSTRACT

OBJECTIVES: Platelet activation underpins thrombus formation in ischemic stroke. The active, dimeric form of platelet receptor glycoprotein (GP) VI plays key roles by binding platelet ligands collagen and fibrin, leading to platelet activation. We investigated whether patients presenting with stroke expressed more GPVI on their platelet surface and had more active circulating platelets as measured by platelet P-selectin exposure. METHODS: 129 ischemic or hemorrhagic stroke patients were recruited within 8h of symptom onset. Whole blood was analyzed for platelet-surface expression of total GPVI, GPVI-dimer, and P-selectin by flow cytometry at admission and day-90 post-stroke. Results were compared against a healthy control population (n = 301). RESULTS: The platelets of stroke patients expressed significantly higher total GPVI and GPVI-dimer (P<0.0001) as well as demonstrating higher resting P-selectin exposure (P<0.0001), a measure of platelet activity, compared to the control group, suggesting increased circulating platelet activation. GPVI-dimer expression was strongly correlated circulating platelet activation [r2 = 0.88, P<0.0001] in stroke patients. Furthermore, higher platelet surface GPVI expression was associated with increased stroke severity at admission. At day-90 post-stroke, GPVI-dimer expression and was further raised compared to the level at admission (P<0.0001) despite anti-thrombotic therapy. All ischemic stroke subtypes and hemorrhagic strokes expressed significantly higher GPVI-dimer compared to controls (P<0.0001). CONCLUSIONS: Stroke patients express more GPVI-dimer on their platelet surface at presentation, lasting at least until day-90 post-stroke. Small molecule GPVI-dimer inhibitors are currently in development and the results of this study validate that GPVI-dimer as an anti-thrombotic target in ischemic stroke.


Subject(s)
Biomarkers/blood , Platelet Activation , Platelet Adhesiveness , Platelet Membrane Glycoproteins/analysis , Stroke/diagnosis , Aged , Aged, 80 and over , Case-Control Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/metabolism , Prognosis , Protein Multimerization , Stroke/metabolism
4.
J Thromb Haemost ; 19(8): 2056-2067, 2021 08.
Article in English | MEDLINE | ID: mdl-34032355

ABSTRACT

OBJECTIVE: The platelet collagen receptor glycoprotein VI (GPVI) has an independent role as a receptor for fibrin produced via the coagulation cascade. However, various reports of GPVI binding to immobilized fibrin(ogen) are not consistent. As a collagen receptor, GPVI-dimer is the functional form, but whether GPVI dimers or monomers bind to fibrin remains controversial. To resolve this, we analyzed GPVI binding to nascent fibrin clots, which more closely approximate physiological conditions. METHODS AND RESULTS: ELISA using biotinyl-fibrinogen immobilized on streptavidin-coated wells indicated that GPVI dimers do not bind intact fibrinogen. Clots were formed by adding thrombin to a mixture of near-plasma level of fibrinogen and recombinant GPVI ectodomain: GPVI dimer (GPVI-Fc2 or Revacept) or monomer (GPVI-His: single chain of Revacept GPVI domain, with His tag). Clot-bound proteins were analyzed by SDS-PAGE/immunoblotting. GPVI-dimer bound to noncrosslinked fibrin clots with classical one-site binding kinetics, with µM-level KD , and to crosslinked clots with higher affinity. Anti-GPVI-dimer (mFab-F) inhibited the binding. However, GPVI-His binding to either type of clot was nonsaturable and nearly linear, indicating very low affinity or nonspecific binding. In clots formed in the presence of platelets, clot-bound platelet-derived proteins were integrin αIIbß3, present at high levels, and GPVI. CONCLUSIONS: We conclude that dimeric GPVI is the receptor for fibrin, exhibiting a similar KD to those obtained for its binding to fibrinogen D-fragment and D-dimer, suggesting that fibrin(ogen)'s GPVI-binding site becomes exposed after fibrin formation or cleavage to fragment D. Analysis of platelets bound to fibrin clots indicates that platelet GPVI binds to fibrin fibers comprising the clot.


Subject(s)
Fibrin , Fibrinogen , Blood Platelets , Collagen , Humans , Platelet Membrane Glycoproteins , Receptors, Collagen
5.
Res Pract Thromb Haemost ; 4(2): 285-297, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32110760

ABSTRACT

OBJECTIVE: Platelet surface receptors are also present subcellularly in organelle membranes and can be expressed on the surface upon platelet activation. However, some receptors were reported to be decreased after activation. We analyzed the mechanism of activation-dependent expression for different receptors. METHODS: Flow cytometry using platelet-rich plasma or washed platelets was used to analyze receptor-expression changes after platelet activation by glycoprotein (GP) VI-specific agonists, crosslinked collagen-related peptide (CRP-XL) and convulxin (Cvx), and thrombin. Platelets prelabeled with fluorescent antibody specific for a receptor were allowed to adhere on immobilized collagen or fibrinogen and post-stained with antibody against the same receptor labeled with another fluorophore, allowing us to differentiate preexisting receptors from newly expressed receptors. RESULTS: Surface expression of αIIbß3 increased in CRP-XL-, Cvx-, or thrombin-stimulated platelets, but GPIb decreased due to shedding and internalization. Both total and dimeric GPVI increased in thrombin-induced platelets, but decreased in platelets stimulated by Cvx, as a result of internalization. The larger platelets showed a greater increase in surface receptor (α2ß1, αIIbß3, GPVI, GPIb) expression upon activation compared to the smaller ones. Pre- and postlabeling with antibody specific for the same receptor, but conjugated with different fluorophores, allowed us to differentiate the receptors expressed on the surface of resting platelets from receptors newly exposed to the surface upon platelet activation. CONCLUSIONS: Increased receptor expressions after activation are mainly manifested in the larger platelets. On platelets adhered on fibrinogen, the newly expressed receptors, especially GPVI, are localized in the lamellipodia of the spread platelets.

6.
Data Brief ; 23: 103784, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31372431

ABSTRACT

This data article is associated with the manuscript "GPVI surface expression and signalling pathway activation are increased in platelets from obese patients: elucidating potential anti-atherothrombotic targets in obesity" [1]. The study refers to a combination of different approaches in order to identify platelet-derived biomarkers in obesity. A total of 34 obese patients and their lean-matched controls were included in the study. We carried out a proteomic and functional (aggregation assays) analysis to find alterations in platelet-derived signalling pathways. After that, biochemical and mechanistic (flow cytometry assays) approaches were done in order to confirm a hyperactivation of the GPVI-related signalling pathway.

7.
Atherosclerosis ; 281: 62-70, 2019 02.
Article in English | MEDLINE | ID: mdl-30658193

ABSTRACT

BACKGROUND AND AIMS: Platelets play a fundamental role in the increased atherothrombotic risk related to central obesity since they show hyperactivation and lower sensitivity to antiplatelet therapy in obese patients. The main goal of this study was to identify platelet biomarkers related to the risk of atherothrombosis in obese patients, confirm platelet activation levels in these patients, and identify altered activation pathways. METHODS: Platelets were obtained from cohorts of obese patients and age- and sex-matched lean controls. Biochemical and proteome analyses were done by two-dimensional differential in-gel electrophoresis (2D-DIGE), mass spectrometry, and immunoblotting. Functional and mechanistic studies were conducted with aggregation assays and flow cytometry. RESULTS: We confirmed an up-regulation of αIIb and fibrinogen isoforms in platelets from obese patients. A complementary platelet aggregation approach showed platelets from obese patients are hyper-reactive in response to collagen and collagen-related peptide (CRP), revealing the collagen receptor Glycoprotein VI (GPVI) signalling as one of the altered pathways. We also found the active form of Src (pTyr418) is up-regulated in platelets from obese individuals, which links proteomics to aggregation data. Moreover, we showed that CRP-activated platelets present higher levels of tyrosine phosphorylated PLCγ2 in obese patients, confirming alterations in GPVI signalling. In line with the above, flow cytometry studies show higher surface expression levels of total GPVI and GPVI-dimer in obese platelets, both correlating with BMI. CONCLUSIONS: Our results suggest a higher activation state of SFKs-mediated signalling pathways in platelets from obese patients, with a primary involvement of GPVI signalling.


Subject(s)
Blood Platelets/metabolism , Obesity/blood , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , Adolescent , Adult , Body Mass Index , Case-Control Studies , Female , Humans , Male , Obesity/diagnosis , Phospholipase C gamma/blood , Phosphorylation , Platelet Aggregation , Signal Transduction , Up-Regulation , Young Adult
8.
Sci Rep ; 8(1): 16677, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30420683

ABSTRACT

Citalopram, a selective serotonin reuptake inhibitor (SSRI), inhibits platelet function in vitro. We have previously shown that this action is independent of citalopram's ability to block serotonin uptake by the serotonin transporter and must therefore be mediated via distinct pharmacological mechanisms. We now report evidence for two novel and putative mechanisms of citalopram-induced platelet inhibition. Firstly, in platelets, citalopram blocked U46619-induced Rap1 activation and subsequent platelet aggregation, but failed to inhibit U46619-induced increases in cytosolic Ca2+. Similarly, in neutrophils, citalopram inhibited Rap1 activation and downstream functions but failed to block PAF-induced Ca2+ mobilisation. In a cell-free system, citalopram also reduced CalDAG-GEFI-mediated nucleotide exchange on Rap1B. Secondly, the binding of anti-GPVI antibodies to resting platelets was inhibited by citalopram. Furthermore, citalopram-induced inhibition of GPVI-mediated platelet aggregation was instantaneous, reversible and displayed competitive characteristics, suggesting that these effects were not caused by a reduction in GPVI surface expression, but by simple competitive binding. In conclusion, we propose two novel, putative and distinct inhibitory mechanisms of action for citalopram: (1) inhibition of CalDAG-GEFI/Rap1 signalling, and (2) competitive antagonism of GPVI in platelets. These findings may aid in the development of novel inhibitors of CalDAG-GEFI/Rap1-dependent nucleotide exchange and novel GPVI antagonists.


Subject(s)
Citalopram/pharmacology , Neutrophils/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Calcium/metabolism , Cytosol/metabolism , Humans , L-Lactate Dehydrogenase/metabolism , Models, Biological , Neutrophils/cytology , Platelet Membrane Glycoproteins/metabolism
9.
Int J Hematol ; 96(6): 733-42, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23143686

ABSTRACT

Bernard-Soulier syndrome (BSS) is an inherited bleeding disorder caused by a defect in the platelet glycoprotein (GP) Ib/IX complex. The GPIX W127X mutation is the most common genetic defect in Japanese patients with BSS, which is often misdiagnosed as immune thrombocytopenic purpura, presumably due to residual expression of GPIbα. Neither the mechanism by which this mutation leads to a mild bleeding diathesis, nor whether functional GPIbα is expressed on platelet surfaces is known. We investigated GPIbα expression and function in platelets with a GPIX W127X mutation (GPIXW127X). GPIbα complexed with GPIbß by disulfide bonding was expressed on GPIXW127X platelets and stable CHO-K1 cells lacking GPIX but expressing GPIbα and GPIbß. Expression of GPIbα/ß on GPIXW127X platelets was sufficient to support adhesion to immobilized von Willebrand factor and type III collagen and ristocetin-induced platelet agglutination. A residual amount of functional GPIbα/ß heteromer expressed on GPIXW127X platelets partially compensates for the absence of the GPIb/IX complex. This may account for the mild bleeding phenotype of the BSS variant characterized by a non-sense mutation in GPIX.


Subject(s)
Bernard-Soulier Syndrome/genetics , Blood Platelets/metabolism , Codon, Nonsense , Membrane Glycoproteins/biosynthesis , Platelet Glycoprotein GPIb-IX Complex/genetics , Point Mutation , Adult , Animals , Antibodies, Monoclonal/immunology , Bernard-Soulier Syndrome/diagnosis , CHO Cells , Collagen Type III/metabolism , Cricetinae , Cystine/chemistry , Diagnostic Errors , Female , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Phenotype , Platelet Adhesiveness , Platelet Glycoprotein GPIb-IX Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/physiology , Pregnancy , Pregnancy Complications, Hematologic/diagnosis , Pregnancy Complications, Hematologic/genetics , Purpura, Thrombocytopenic, Idiopathic/diagnosis , Recombinant Fusion Proteins/physiology , Ristocetin/pharmacology , Transfection , von Willebrand Factor/metabolism
10.
J Biol Chem ; 287(35): 30000-13, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22773837

ABSTRACT

The platelet collagen receptor glycoprotein VI (GPVI) has been suggested to function as a dimer, with increased affinity for collagen. Dissociation constants (K(d)) obtained by measuring recombinant GPVI binding to collagenous substrates showed that GPVI dimers bind with high affinity to tandem GPO (Gly-Pro-Hyp) sequences in collagen, whereas the markedly lower affinity of the monomer for all substrates implies that it is not the collagen-binding form of GPVI. Dimer binding required a high density of immobilized triple-helical (GPO)(10)-containing peptide, suggesting that the dimer binds multiple, discrete peptide helices. Differential inhibition of dimer binding by dimer-specific antibodies, m-Fab-F and 204-11 Fab, suggests that m-Fab-F binds at the collagen-binding site of the dimer, and 204-11 Fab binds to a discrete site. Flow cytometric quantitation indicated that GPVI dimers account for ~29% of total GPVI in resting platelets, whereas activation by either collagen-related peptide or thrombin increases the number of dimers to ~39 and ~44%, respectively. m-Fab-F inhibits both GPVI-dependent static platelet adhesion to collagen and thrombus formation on collagen under low and high shear, indicating that pre-existing dimeric GPVI is required for the initial interaction with collagen because affinity of the monomer is too low to support binding and that interaction through the dimer is essential for platelet activation. These GPVI dimers in resting circulating platelets will enable them to bind injury-exposed subendothelial collagen to initiate platelet activation. The GPVI-specific agonist collagen-related peptide or thrombin further increases the number of dimers, thereby providing a feedback mechanism for reinforcing binding to collagen and platelet activation.


Subject(s)
Collagen/pharmacology , Peptides/pharmacology , Platelet Activation/drug effects , Platelet Membrane Glycoproteins/agonists , Platelet Membrane Glycoproteins/metabolism , Protein Multimerization/drug effects , Blood Flow Velocity , Humans , Immunoglobulin Fab Fragments , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Secondary , Thrombin/pharmacology
11.
Platelets ; 21(2): 101-11, 2010.
Article in English | MEDLINE | ID: mdl-20063990

ABSTRACT

Platelets in flowing blood at high-shear stress are recruited to exposed subendothelial collagen of injured vessels by GPIb-von Willebrand factor (vWf) and integrin alpha(2)beta(1) (alpha(2)beta(1))-collagen interactions. Platelet adhesion to type I collagen depends mainly on the alpha(2)beta(1)-collagen interaction and that to type III collagen depends on the GPIb-vWf interaction due to vWf's weak affinity for type I collagen. Contributions of these two interactions would differ depending on expressions of alpha(2)beta(1), vWf, or GPIb. We quantitated platelet adhesion to low- and high-density collagen under high-shear flow conditions in the presence of anti-alpha(2)beta(1) (Gi9) and anti-GPIb (NNKY5-5) antibodies to determine if their inhibitory effects were correlated with the amounts of alpha(2)beta(1), GPIb and vWf. Gi9 inhibition of adhesion to type I collagen was decreased in platelets with more integrin alpha(2)beta(1). Gi9 and NNKY5-5 are more inhibitory against adhesion to low-density type III and I, respectively. Higher alpha(2)beta(1) expression decreases adhesion to low-density type III and increases Gi9 inhibition of adhesion to high-density type III, suggesting crosstalk between the alpha(2)beta(1)-collagen and GPIb-vWf interactions in adhesion to type III. Integrin alpha(2)beta(1)-collagen and GPIb-vWf interactions both contribute to platelet adhesion to collagen under high-shear flow. In adhesion under high-shear stress, the two interactions would compensate for each other, when there is a deficiency in one or the other. The alpha(2)beta(1)-collagen interaction was also suggested to have an inhibitory effect on platelet adhesion to type III collagen, through a yet undefined mechanism.


Subject(s)
Blood Platelets/physiology , Collagen Type III/metabolism , Collagen Type I/metabolism , Integrin alpha2beta1/metabolism , Platelet Adhesiveness/physiology , Platelet Glycoprotein GPIb-IX Complex/metabolism , von Willebrand Factor/metabolism , Adolescent , Animals , Humans , Platelet Function Tests , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Shear Strength/physiology , Stress, Mechanical , Young Adult , von Willebrand Diseases/blood
12.
Platelets ; 20(3): 143-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19437330

ABSTRACT

Recent experimental evidence demonstrates that the platelet-specific collagen receptor, glycoprotein (GP)VI is essentially all uncleaved on normal circulating platelets, but is shed from the platelet surface in a metalloproteinase-dependent manner in response to GPVI ligands (including collagen), anti-GPVI antibodies or activation at the platelet Fc receptor, FcgammaRIIa. This raises the question of whether shed ectodomain fragment in plasma could be a useful biomarker of thrombotic risk and/or autoimmune thrombocytopenia. In this study, we developed a sandwich enzyme-linked immunosorbent assay (ELISA) for measuring soluble GPVI in human plasma, using rabbit anti-GPVI polyclonal antibody in the solid-phase, murine anti-GPVI monoclonal antibody (1A12) in the fluid-phase and horseradish peroxidase (HRP)-coupled anti-mouse antibody and enhanced chemiluminescence (ECL) for detection. The ELISA was optimized for sensitivity, reproducibility, inter- and intra-assay precision, addition and recovery and detected GPVI in plasma with a lower detection limit of approximately 1 ng/mL. Effects of different anti-coagulants (trisodium citrate, acid-citrate-dextrose or EDTA) were negligible. In ten healthy donors, soluble plasma GPVI levels were 18.9 +/- 4.1 ng/mL. Treating normal platelet-rich plasma with a GPVI ligand (collagen-related peptide, CRP), calmodulin inhibitor W7 (that induces GPVI shedding without platelet activation) or N-ethylmaleimide (that directly activates platelet sheddases), under conditions previously shown to induce GPVI shedding, also increased plasma GPVI levels by up to approximately 7-fold, compared to previously reported autoimmune (anti-GPVI) patient plasma where soluble GPVI was approximately 10-fold higher than normal. Characterization of this sensitive ELISA should facilitate analysis of functional/diagnostic role(s) for soluble GPVI in human plasma associated with thrombotic/immune dysfunction.


Subject(s)
Blood Platelets/metabolism , CD36 Antigens/blood , Animals , Antibodies , Anticoagulants/pharmacology , Blood Platelets/drug effects , CD36 Antigens/immunology , Calmodulin/antagonists & inhibitors , Carrier Proteins/pharmacology , Enzyme Activators/pharmacology , Enzyme-Linked Immunosorbent Assay , Ethylmaleimide/pharmacology , Humans , In Vitro Techniques , Mice , Peptides/pharmacology , Platelet Activation , Platelet-Rich Plasma , Rabbits , Reproducibility of Results , Sensitivity and Specificity , Sulfonamides/pharmacology
13.
Platelets ; 20(2): 75-82, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19235048

ABSTRACT

Adhesion of circulating platelets to the blood vessel wall initiates thrombus formation in haemostasis and thrombotic disease. The platelet collagen receptor, glycoprotein (GP) VI, is critical for thrombus formation at arterial shear rates and is a potential therapeutic target for anti-thrombotic drugs. In this study, we evaluate eight newly-derived, purified murine anti-human GPVI monoclonal antibodies (mAbs) for their effect on GPVI-dependent platelet aggregation and GPVI ectodomain shedding. All mAbs were raised against the ligand-binding GPVI ectodomain encompassing two immunoglobulin domains (residues 21-234, excluding the signal sequence) and recognized full-length GPVI in human platelet lysates by western blotting. The majority of antibodies induced aggregation in both human platelet-rich plasma (PRP) and washed platelets independently of the Fc receptor, FcgammaRIIa (not inhibited by the blocking anti-FcgammaRIIa mAb, IV.3), whereas one mAb (11A7) neither induced aggregation nor inhibited aggregation in response to GPVI ligands, collagen, and collagen-related peptide (CRP). In contrast, Fab fragments of mAb 12A5 strongly blocked collagen- and CRP-, but not convulxin-induced aggregation. In addition, it is shown for the first time in vitro that anti-GPVI mAbs can induce metalloproteinase-dependent ectodomain shedding of human GPVI, generating an approximately 10-kDa remnant that remained platelet-associated and an approximately 55-kDa soluble fragment. In conclusion, this analysis of anti-GPVI mAbs provides useful tools for studying the functional role of platelet GPVI.


Subject(s)
Antibodies, Monoclonal/metabolism , Blood Platelets/metabolism , Platelet Aggregation/physiology , Platelet Membrane Glycoproteins/metabolism , Receptors, IgG/metabolism , Animals , Humans , Immunoglobulin Fab Fragments/metabolism , Mice
14.
Kurume Med J ; 56(3-4): 61-9, 2009.
Article in English | MEDLINE | ID: mdl-20505283

ABSTRACT

Megakaryocytes (MKs) and platelet-like particles (PLPs) have generally been obtained by culturing embryonic stem (ES) cells over feeder cells. However, using feeder cells need many labor-consuming processes and the MK and PLP fractions obtained are often contaminated by such cells and their fragments. Here we describe our new culture system for differentiating mouse ES cells to MKs and PLPs without using feeder cells. ES cells are differentiated to cells with MK-like morphology and properties, including proplatelet formation, high ploidy (>8N), and CD41 expression. The culture medium contained PLPs expressing platelet glycoproteins, CD41 and GPIb. Integrin alpha(IIb)beta(3) of PLPs can be activated by thrombin. Addition of the metalloproteinase inhibitor TAPI-2 to the culture increased the surface expression of GPIbalpha and augmented the adhesion of PLPs to immobilized von Willebrand factor through decreasing the shedding of GPIbalpha. Thus our mouse ES cells culture system is a suitable and efficient method for obtaining MKs and functional PLPs that obviates the need for feeder cells.


Subject(s)
Embryonic Stem Cells/cytology , Megakaryocytes/cytology , Animals , Cell Differentiation , Dipeptides/pharmacology , Hydroxamic Acids/pharmacology , Membrane Glycoproteins/blood , Mice , Mice, Inbred C57BL , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Platelet Glycoprotein GPIb-IX Complex , Thrombopoiesis
15.
Adv Exp Med Biol ; 640: 53-63, 2008.
Article in English | MEDLINE | ID: mdl-19065783

ABSTRACT

Glycoprotein VI (GPVI) is a membrane glycoprotein unique to platelets and has been identified as a physiological receptor for collagen. Damage to a vessel wall exposes the subendothelial component collagen to platelets in the blood flow. Interaction of platelets with collagen via the receptor GPVI results in platelet activation and adhesion--the processes that are essential for thrombus formation. On the platelet surface, GPVI is present as a complex with the homodimeric Fc receptor y-chain (FcRgamma with a possible stoichiometry of two GPVI molecules and one FcRgamma dimer). When collagen binds to GPVI, a platelet activation cascade is initiated by tyrosine phosphorylation of the immunoreceptor tyrosine-based activation motif of FcRgamma and this phosphorylation induces the formation of a large complex composed from many signal-transducing proteins. In flow adhesion experiments that closely approximate physiological conditions, GPVI is essential for the formation of large platelet aggregates on collagen. However, GPVI-deficient patients or mice do not show any severe bleeding tendency. This suggests that a GPVI inhibitor would be able to inhibit thrombus formation but still not cause a significant bleeding tendency. Such an inhibitor would show promise as an anti-thrombotic agent for clinical use.


Subject(s)
Platelet Membrane Glycoproteins/metabolism , Animals , Collagen/chemistry , Collagen/metabolism , Humans , Platelet Membrane Glycoproteins/chemistry , Signal Transduction
16.
Int J Oncol ; 33(6): 1271-80, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19020761

ABSTRACT

Transcription factor binding sites are short DNA sequences that interact with transcription factors and the proper control of gene expression appears to require the mechanisms including the regulation through the genome context around the transcription factor binding sites. The MYC proteins are central regulators of cell growth. Many genes have been reported to be regulated by MYC through E-box sites. However, the characters of E-box that Myc selects to function are not clear and identification of additional genes controlled by MYC will provide information to completely understand the functions of MYC. Here we report that MYC directly induces TAF4b expression. We mapped the transcription start site and characterized functional promoter elements for MYC response in the TAF4b promoter. There are several E-box sequences near the transcription start site, including canonical (CACGTG) and non-canonical (CGCGTG) ones. We found that c-MYC induces TAF4b expression through one of the non-canonical E-box sites, which is in a highly conserved region of TAF4b promoters in mammals, suggesting the importance of the genome context around the target E-box. When the non-canonical E-box in the TAF4b promoter was mutated to a canonical one, MYC functioned on both E-boxes, while another E-box-binding transcription factor, USF, did so on only the canonical E-box. These results suggest that in addition to the context where the target E-box exists, a sequence within an E-box is involved in the mechanisms by which specific E-box sites are selected by Myc.


Subject(s)
E-Box Elements , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/metabolism , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Transcriptional Activation , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Conserved Sequence , Genes, Reporter , HL-60 Cells , Humans , Molecular Sequence Data , Mutation , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/metabolism , Rats , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Recombinant Fusion Proteins/metabolism , Sequence Homology, Nucleic Acid , TATA-Binding Protein Associated Factors/metabolism , Time Factors , Transcription Factor TFIID/metabolism , Transfection , Upstream Stimulatory Factors/metabolism
17.
Platelets ; 19(1): 32-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18231936

ABSTRACT

Of the two physiologically important platelet collagen receptors, glycoprotein (GP) VI is the receptor responsible for platelet activation. However, its reactivities towards different types of vascular collagen have not been directly and quantitatively analysed with collagen preparations of defined composition, although the other major platelet collagen receptor integrin alpha(2)beta(1) was shown to react with collagen types I-VI and VIII under either static or flow conditions. We analysed the collagen type specificity of GPVI binding to identify the physiological contribution of the various vascular collagens and how platelet reactivity towards the various collagens may be affected by fibril size. We used two methods to analyse the binding of recombinant GPVI (GPVI-Fc(2)) to different types of bovine collagen: binding to collagen microparticles in suspension and binding to immobilized collagen. GPVI-Fc(2) bound to type I-III collagens that can form large fibrils, but not to type V that only forms small fibrils. The apparent GPVI binding to types IV and V could be ascribed to type I collagen that was a contaminant in each of these preparations. Kinetic analyses of the binding data showed that type III collagen fibrils have both a higher Kd and Bmax than types I and II. Flow adhesion studies demonstrated that type III collagen supports the formation of larger platelet aggregates than type I. Our present results suggest that the physiological importance of type III collagen is to induce thrombus formation. Furthermore, these studies indicate that GPVI mainly binds to collagen types that can form large collagen fibrils.


Subject(s)
Blood Platelets/metabolism , Fibrillar Collagens/metabolism , Platelet Adhesiveness/physiology , Platelet Membrane Glycoproteins/metabolism , Animals , Blood Platelets/chemistry , Cattle , Fibrillar Collagens/chemistry , Humans , Integrin alpha2beta1/chemistry , Integrin alpha2beta1/metabolism , Kinetics , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Protein Binding/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Substrate Specificity/physiology
18.
Thromb Haemost ; 98(6): 1285-90, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18064326

ABSTRACT

Glycoprotein (GP)VI, that binds collagen, together with GPIb-IX-V which binds vonWillebrand factor, forms an adheso-signalling complex on platelets that initiates thrombus formation in haemostasis and thrombosis. In this study, we show that two snake venom metalloproteinases, crotarhagin and alborhagin, induce ectodomain shedding of GPVI by a mechanism that involves activation of endogenous platelet metalloproteinases. Alborhagin is a viper venom metalloproteinase from Trimeresurus albolabris, while crotarhagin is a previously undescribed toxin from the rattlesnake Crotalus horridus horridus ( approximately 60-kDa non-reduced and reduced). Like alborhagin, crotarhagin induces aggregation in human platelet-rich plasma (maximal activity, approximately 0.3 microg/ml). Aggregation of washed platelets was inhibited by soluble GPVI ectodomain expressed as an Fc-fusion protein, confirming crotarhagin targeted GPVI. Treating washed platelets with crotarhagin or alborhagin resulted in time-dependent loss of surface GPVI and the appearance of an approximately 55-kDa soluble GPVI fragment in supernatants. Crotarhagin also induced shedding in GPVItransfected RBL-2H3 cells. Crotarhagin-induced shedding was metalloproteinase-dependent (inhibited by EDTA), but also blocked by inhibitors of GPVI signalling (Src kinase inhibitors, PP1 or PP2, or Syk inhibitor, piceatannol), indicating shedding required GPVI-dependent platelet activation. Together, the data suggest that the rattlesnake metalloproteinase, crotarhagin, and the viper toxin alborhagin, induce GPVI shedding by a mechanism involving activation of endogenous platelet metalloproteinases rather than direct cleavage of GPVI.


Subject(s)
Blood Platelets/drug effects , Crotalid Venoms/toxicity , Metalloendopeptidases/toxicity , Metalloproteases/toxicity , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/drug effects , Receptors, Collagen/drug effects , Viper Venoms/toxicity , Animals , Blood Platelets/enzymology , Blood Platelets/metabolism , Cell Line , Dose-Response Relationship, Drug , Edetic Acid/pharmacology , Enzyme Activation , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Peptide Fragments/metabolism , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/metabolism , Protease Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Receptors, Collagen/chemistry , Receptors, Collagen/genetics , Receptors, Collagen/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction/drug effects , Stilbenes/pharmacology , Syk Kinase , Time Factors , Transfection , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
19.
Thromb Res ; 118(4): 509-21, 2006.
Article in English | MEDLINE | ID: mdl-16380154

ABSTRACT

The abundant Rap1 in platelets becomes activated when these cells are stimulated by various agonists, but its function has remained unknown. In view of this, we developed an assay to quantitatively measure activated Rap1 and used it to determine relationships between Rap1 activation and several platelet functions: integrin alpha2beta1 activation, tyrosine phosphorylation, and the release reaction. We looked at how these processes are affected by the protein kinase C inhibitor BIMI, tyrosine kinase inhibitor PP2, PI 3-kinase inhibitor wortmannin, and ADP scavenger apyrase. In CRP (collagen related peptide)-activated platelets, all the inhibitors severely inhibited Rap1 activation, but had little effect on integrin alpha2beta1 activation, indicating that the integrin activation mechanism is different from the Rap1 activation mechanism, at least in GPVI-dependent activation. With p85alpha-null mouse platelets, we demonstrated that Rap1 activation involves PI 3-kinase p85alpha-dependent tyrosine phosphorylation. All the inhibitors similarly decreased Rap1 activation and the serotonin release reaction, and the inhibition of Rap1 activation was not due to the lack of released ADP. Our results indicate that platelet Rap1 activation is closely related to the release reaction and not to integrin alpha2beta1 activation in GPVI-mediated platelet activation.


Subject(s)
Blood Platelets/metabolism , Platelet Membrane Glycoproteins/metabolism , rap1 GTP-Binding Proteins/metabolism , Androstadienes/pharmacology , Apyrase/pharmacology , Collagen/metabolism , Humans , Integrin alpha2beta1/antagonists & inhibitors , Integrin alpha2beta1/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Tyrosine/metabolism , Wortmannin , rap1 GTP-Binding Proteins/antagonists & inhibitors
20.
Blood ; 107(4): 1405-12, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16219796

ABSTRACT

The extracellular matrix protein, laminin, supports platelet adhesion through binding to integrin alpha6beta1 In the present study, we demonstrate that human laminin, purified from placenta, also stimulates formation of filopodia and lamellipodia in human and mouse platelets through a pathway that is dependent on alpha6beta1 and the collagen receptor GPVI. The integrin alpha6beta1 is essential for adhesion to laminin, as demonstrated using an alpha6-blocking antibody, whereas GPVI is dispensable for this response, as shown using "knockout" mouse platelets. On the other hand, lamellipodia formation on laminin is completely inhibited in the absence of GPVI, although filopodia formation remains and is presumably mediated via alpha6beta1 Lamellipodia and filopodia formation are inhibited in Syk-deficient platelets, demonstrating a key role for the kinase in signaling downstream of GPVI and integrin alpha6beta1 GPVI was confirmed as a receptor for laminin using surface plasmon resonance spectroscopy and by demonstration of lamellipodia formation on laminin in the presence of collagenase. These results identify GPVI as a novel receptor for laminin and support a model in which integrin alpha6beta1 brings laminin to GPVI, which in turn mediates lamellipodia formation. We speculate that laminin contributes to platelet spreading in vivo through a direct interaction with GPVI.


Subject(s)
Blood Platelets/cytology , Blood Platelets/physiology , Integrin alpha6beta1/physiology , Laminin/pharmacology , Platelet Membrane Glycoproteins/metabolism , Animals , Blood Platelets/drug effects , Female , Humans , Laminin/isolation & purification , Mice , Placenta , Pregnancy , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL