Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
Bioorg Chem ; 121: 105661, 2022 04.
Article in English | MEDLINE | ID: mdl-35182883

ABSTRACT

Cardiovascular diseases are the primary reason for deaths in the world. However, antiplatelet drugs in the market have limitations in use because of the risk of increased bleeding and other side effects that impair primary homeostasis. Therefore, safe and effective antithrombotic drugs are needed for the treatment of plaque formation in blood vessels. Glycoprotein VI (GPVI) is a platelet major collagen receptor and a target for potent and safe antithrombotic therapy. We designed this study based on the molecular interaction pattern of previously published GPVI receptor antagonists within the reported binding site. We selected sixteen hit compounds from a large chemical database that contains>6 million in-stock compounds by following a combined virtual screening. Then, we evaluated their inhibitory effects on platelet aggregation induced by GPVI receptor agonists (collagen, collagen related peptide (CRP), convulxin) and the most potent platelet agonist, thrombin, in vitro by using washed human platelets. IC50 values of compounds 1 and 2 are, respectively, 0.35 µM and 1.01 µM for collagen, 0.80 µM and 1.92 µM for CRP, 195.2 and 7.24 µM for convulxin and 81.38 and 51.74 µM for thrombin. We identified compounds 1 and 2 as the most promising antiplatelet agents out of sixteen compounds. Additionally, compounds 1 and 2 may serve as promising starting points and shed light on the design of new, potent and selective GPVI receptor antagonists.


Subject(s)
Platelet Aggregation Inhibitors , Platelet Membrane Glycoproteins , Blood Platelets , Collagen/metabolism , Collagen/pharmacology , Humans , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/pharmacology , Thrombin/metabolism
2.
Int J Obes (Lond) ; 46(1): 68-76, 2022 01.
Article in English | MEDLINE | ID: mdl-34493775

ABSTRACT

BACKGROUND/OBJECTIVES: Platelet-activating factor receptor (PAFR) activation controls adipose tissue (AT) expansion in animal models. Our objective was twofold: (i) to check whether PAFR signaling is involved in human obesity and (ii) investigate the PAF pathway role in hematopoietic or non-hematopoietic cells to control adipocyte size. MATERIALS/SUBJECTS AND METHODS: Clinical parameters and adipose tissue gene expression were evaluated in subjects with obesity. Bone marrow (BM) transplantation from wild-type (WT) or PAFR-/- mice was performed to obtain chimeric PAFR-deficient mice predominantly in hematopoietic or non-hematopoietic-derived cells. A high carbohydrate diet (HC) was used to induce AT remodeling and evaluate in which cell compartment PAFR signaling modulates it. Also, 3T3-L1 cells were treated with PAF to evaluate fat accumulation and the expression of genes related to it. RESULTS: PAFR expression in omental AT from humans with obesity was negatively correlated to different corpulence parameters and more expressed in the stromal vascular fraction than adipocytes. Total PAFR-/- increased adiposity compared with WT independent of diet-induced obesity. Differently, WT mice receiving PAFR-/--BM exhibited similar adiposity gain as WT chimeras. PAFR-/- mice receiving WT-BM showed comparable augmentation in adiposity as total PAFR-/- mice, demonstrating that PAFR signaling modulates adipose tissue expansion through non-hematopoietic cells. Indeed, the PAF treatment in 3T3-L1 adipocytes reduced fat accumulation and expression of adipogenic genes. CONCLUSIONS: Therefore, decreased PAFR signaling may favor an AT accumulation in humans and animal models. Importantly, PAFR signaling, mainly in non-hematopoietic cells, especially in adipocytes, appears to play a significant role in regulating diet-induced AT expansion.


Subject(s)
Adipose Tissue/physiopathology , Obesity/complications , Platelet Membrane Glycoproteins/pharmacology , Adipose Tissue/metabolism , Adult , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Obesity/physiopathology , Paris , Receptors, G-Protein-Coupled , Signal Transduction/physiology
3.
Platelets ; 32(6): 724-732, 2021 Aug 18.
Article in English | MEDLINE | ID: mdl-33634725

ABSTRACT

GPVI is a critical signaling receptor responsible for collagen-induced platelet activation and a promising anti-thrombotic target in conditions such as coronary artery thrombosis, ischemic stroke, and atherothrombosis. This is due to the ability to block GPVI while having minimal effects on hemostasis, making it a more attractive target over current dual-antiplatelet therapy (DAPT) with acetyl salicylic acid and P2Y12 inhibitors where bleeding can be a problem. Our current understanding of how the structure of GPVI relates to function is inadequate and recent studies contradict each other. In this article, we summarize the structure-function relationships underlying the activation of GPVI by its major ligands, including collagen, fibrin(ogen), snake venom toxins and charged exogenous ligands such as diesel exhaust particles. We argue that contrary to popular belief dimerization of GPVI is not required for binding to collagen but serves to facilitate binding through increased avidity, and that GPVI is expressed as a mixture of monomers and dimers on resting platelets, with binding of multivalent ligands inducing higher order clustering.


Subject(s)
Platelet Membrane Glycoproteins/pharmacology , Humans , Structure-Activity Relationship
4.
Nanomedicine ; 29: 102274, 2020 10.
Article in English | MEDLINE | ID: mdl-32712174

ABSTRACT

Thrombus formation at athero-thrombotic sites is initiated by the exposure of collagen followed by platelet adhesion mediated by the platelet-specific collagen receptor glycoprotein VI (GPVI). Here, dimeric GPVI was used as a targeting motif to functionalize polymeric nanoparticle-based drug carriers and to show that with proper design, such GPVI-coated nanoparticles (GPNs) can efficiently and specifically target arterial injury sites while withstanding physiological flow. In a microfluidic model, under physiological shear levels (1-40 dyne/cm2), 200 nm and 2 µm GPNs exhibited a >60 and >10-fold increase in binding to collagen compared to control particles, respectively. In vitro experiments in an arterial stenosis injury model, subjected to physiological pulsatile flow, showed shear-enhanced adhesion of 200 nm GPNs at the stenosis region which was confirmed in vivo in a mice ligation carotid injury model using intravital microscopy. Altogether, our results illustrate how engineering tools can be harnessed to design nano-carriers that efficiently target cardiovascular disease sites.


Subject(s)
Atherosclerosis/drug therapy , Carotid Artery Injuries/drug therapy , Nanoparticles/chemistry , Platelet Membrane Glycoproteins/pharmacology , Animals , Atherosclerosis/pathology , Blood Platelets/drug effects , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Artery Injuries/pathology , Disease Models, Animal , Humans , Mice , Platelet Activation/drug effects , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/chemistry
5.
Blood Adv ; 3(7): 1154-1166, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30967391

ABSTRACT

Diacylglycerol kinases (DGKs) are a family of enzymes that convert diacylglycerol (DAG) into phosphatidic acid (PA). The ζ isoform of DGK (DGKζ) has been reported to inhibit T-cell responsiveness by downregulating intracellular levels of DAG. However, its role in platelet function remains undefined. In this study, we show that DGKζ was expressed at significant levels in both platelets and megakaryocytes and that DGKζ-knockout (DGKζ-KO) mouse platelets were hyperreactive to glycoprotein VI (GPVI) agonists, as assessed by aggregation, spreading, granule secretion, and activation of relevant signal transduction molecules. In contrast, they were less responsive to thrombin. Platelets from DGKζ-KO mice accumulated faster on collagen-coated microfluidic surfaces under conditions of arterial shear and stopped blood flow faster after ferric chloride-induced carotid artery injury. Other measures of hemostasis, as measured by tail bleeding time and rotational thromboelastometry analysis, were normal. Interestingly, DGKζ deficiency led to increased GPVI expression on the platelet and megakaryocyte surfaces without affecting the expression of other platelet surface receptors. These results implicate DGKζ as a novel negative regulator of GPVI-mediated platelet activation that plays an important role in regulating thrombus formation in vivo.


Subject(s)
Diacylglycerol Kinase/pharmacology , Platelet Activation/drug effects , Platelet Membrane Glycoproteins/pharmacology , Animals , Blood Platelets/metabolism , Diacylglycerol Kinase/deficiency , Diacylglycerol Kinase/genetics , Diacylglycerol Kinase/metabolism , Hemostasis , Humans , Megakaryocytes/metabolism , Mice , Mice, Knockout , Platelet Membrane Glycoproteins/drug effects , Thrombosis/etiology
7.
J Am Heart Assoc ; 6(8)2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28751543

ABSTRACT

BACKGROUND: GPVI (Glycoprotein VI) is the essential platelet collagen receptor in atherothrombosis. Dimeric GPVI-Fc (Revacept) binds to GPVI binding sites on plaque collagen. As expected, it did not increase bleeding in clinical studies. GPVI-Fc is a potent inhibitor of atherosclerotic plaque-induced platelet aggregation at high shear flow, but its inhibition at low shear flow is limited. We sought to increase the platelet inhibitory potential by fusing GPVI-Fc to the ectonucleotidase CD39 (fusion protein GPVI-CD39), which inhibits local ADP accumulation at vascular plaques, and thus to create a lesion-directed dual antiplatelet therapy that is expected to lack systemic bleeding risks. METHODS AND RESULTS: GPVI-CD39 effectively stimulated local ADP degradation and, compared with GPVI-Fc alone, led to significantly increased inhibition of ADP-, collagen-, and human plaque-induced platelet aggregation in Multiplate aggregometry and plaque-induced platelet thrombus formation under arterial flow conditions. GPVI-CD39 did not increase bleeding time in an in vitro assay simulating primary hemostasis. In a mouse model of ferric chloride-induced arterial thrombosis, GPVI-CD39 effectively delayed vascular thrombosis but did not increase tail bleeding time in vivo. CONCLUSIONS: GPVI-CD39 is a novel approach to increase local antithrombotic activity at sites of atherosclerotic plaque rupture or injury. It enhances GPVI-Fc-mediated platelet inhibition and presents a potentially effective and safe molecule for the treatment of acute atherothrombotic events, with a favorable risk-benefit ratio.


Subject(s)
Antigens, CD/pharmacology , Apyrase/pharmacology , Carotid Artery Injuries/drug therapy , Fibrinolytic Agents/pharmacology , Glycoproteins/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/pharmacology , Thrombosis/prevention & control , Animals , Antigens, CD/toxicity , Apyrase/pharmacokinetics , Apyrase/toxicity , Carotid Artery Diseases/blood , Carotid Artery Diseases/pathology , Carotid Artery Injuries/blood , Carotid Artery Injuries/chemically induced , Carotid Artery Injuries/pathology , Chlorides , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Ferric Compounds , Fibrinolytic Agents/pharmacokinetics , Fibrinolytic Agents/toxicity , Glycoproteins/pharmacokinetics , Glycoproteins/toxicity , Hemorrhage/chemically induced , Humans , Immunoglobulin Fc Fragments/toxicity , Male , Mice, Inbred C57BL , Plaque, Atherosclerotic , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Aggregation Inhibitors/toxicity , Platelet Membrane Glycoproteins/pharmacokinetics , Platelet Membrane Glycoproteins/toxicity , Recombinant Fusion Proteins/pharmacology , Thrombosis/blood , Thrombosis/chemically induced , Thrombosis/pathology
8.
J Am Coll Cardiol ; 65(22): 2404-15, 2015 Jun 09.
Article in English | MEDLINE | ID: mdl-26046734

ABSTRACT

BACKGROUND: Glycoprotein VI (GPVI) is the essential platelet collagen receptor in atherothrombosis, but its inhibition causes only a mild bleeding tendency. Thus, targeting this receptor has selective antithrombotic potential. OBJECTIVES: This study sought to compare compounds interfering with platelet GPVI-atherosclerotic plaque interaction to improve current antiatherothrombotic therapy. METHODS: Human atherosclerotic plaque-induced platelet aggregation was measured in anticoagulated blood under static and arterial flow conditions (550/s, 1,100/s, and 1,500/s). Inhibition by dimeric GPVI fragment crystallizable region of IgG (Fc) masking GPVI binding sites on collagen was compared with that of 3 anti-GPVI antibodies: BLO8-1, a human domain antibody; 5C4, a fragment antigen-binding (Fab fragment) of monoclonal rat immunoglobulin G; and m-Fab-F, a human recombinant sFab against GPVI dimers. RESULTS: GPVI-Fc reduced plaque-triggered platelet aggregation in static blood by 51%, BLO8-1 by 88%, and 5C4 by 93%. Under arterial flow conditions, BLO8-1 and 5C4 almost completely inhibited platelet aggregation while preserving platelet adhesion on plaque. Inhibition by GPVI-Fc, even at high concentrations, was less marked but increased with shear rate. Advanced optical imaging revealed rapid persistent GPVI-Fc binding to collagen under low and high shear flow, upstream and downstream of plaque fragments. At low shear particularly, platelets adhered in plaque flow niches to GPVI-Fc-free segments of collagen fibers and recruited other platelets onto aggregates via ADP and TxA2 release. CONCLUSIONS: Anti-GPVI antibodies inhibit atherosclerotic plaque-induced platelet aggregation under static and flow conditions more effectively than GPVI-Fc. However, potent platelet inhibition by GPVI-Fc at a higher shear rate (1,500/s) suggests localized antithrombotic efficacy at denuded or fissured stenotic high-risk lesions without systemic bleeding. The compound-specific differences have relevance for clinical trials targeting GPVI-collagen interaction combined with established antiplatelet therapies in patients with spontaneous plaque rupture or intervention-associated plaque injury.


Subject(s)
Blood Flow Velocity/physiology , Carotid Arteries/physiopathology , Immunoglobulin Fab Fragments/pharmacology , Plaque, Atherosclerotic/drug therapy , Platelet Activation/drug effects , Platelet Membrane Glycoproteins/pharmacology , Animals , Blood Flow Velocity/drug effects , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Stenosis/drug therapy , Carotid Stenosis/etiology , Carotid Stenosis/physiopathology , Humans , Immunologic Factors/pharmacology , Plaque, Atherosclerotic/complications , Plaque, Atherosclerotic/diagnosis , Rats
9.
Circulation ; 125(5): 685-96, 2012 Feb 07.
Article in English | MEDLINE | ID: mdl-22223428

ABSTRACT

BACKGROUND: CXCR4-positive bone marrow cells (BMCs) are critically involved in cardiac repair mechanisms contributing to preserved cardiac function. Stromal cell-derived factor-1 (SDF-1) is the most prominent BMC homing factor known to augment BMC engraftment, which is a limiting step of stem cell-based therapy. After myocardial infarction, SDF-1 expression is rapidly upregulated and promotes myocardial repair. METHODS AND RESULTS: We have established a bifunctional protein consisting of an SDF-1 domain and a glycoprotein VI (GPVI) domain with high binding affinity to the SDF-1 receptor CXCR4 and extracellular matrix proteins that become exposed after tissue injury. SDF1-GPVI triggers chemotaxis of CXCR4-positive cells, preserves cell survival, enhances endothelial differentiation of BMCs in vitro, and reveals proangiogenic effects in ovo. In a mouse model of myocardial infarction, administration of the bifunctional protein leads to enhanced recruitment of BMCs, increases capillary density, reduces infarct size, and preserves cardiac function. CONCLUSIONS: These results indicate that administration of SDF1-GPVI may be a promising strategy to treat myocardial infarction to promote myocardial repair and to preserve cardiac function.


Subject(s)
Cell- and Tissue-Based Therapy , Chemokine CXCL12/pharmacology , Heart/drug effects , Heart/physiopathology , Ischemic Attack, Transient/therapy , Myocardial Infarction/therapy , Platelet Membrane Glycoproteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Survival/drug effects , Chemokine CXCL12/metabolism , Chemokine CXCL12/therapeutic use , Collagen/metabolism , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/physiopathology , Mice , Mice, Inbred C57BL , Models, Animal , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Neovascularization, Physiologic/drug effects , Platelet Membrane Glycoproteins/metabolism , Platelet Membrane Glycoproteins/therapeutic use , Protein Binding , Receptors, CXCR4/metabolism , Treatment Outcome
10.
Platelets ; 23(1): 1-6, 2012.
Article in English | MEDLINE | ID: mdl-21671729

ABSTRACT

Vascular lesion formation and collagen presentation are key events leading to the development of vulnerable plaques. Glycoprotein VI (GPVI) significantly contributes to plaque-associated collagen binding and thrombus formation. The aim of this study was to image endothelial injury using fluorescence-labelled GPVI-Fc (Fc, fragment crystallized), a soluble form of GPVI that was generated by cloning and fusing GPVI to an Fc-domain, in an ex-vivo rabbit model. This study serves as a proof-of-principle study to demonstrate that GPVI-Fc is a useful tool for detecting endothelial damage. The carotid and femoral arteries and the aorta abdominalis were isolated from rabbits and perfused with phosphate buffered saline (PBS) to remove all blood, and a catheter was placed into the vessels in situ. Endothelial damage was achieved by pulling an inflated balloon approximately 1 inch through the vessels, while control vessels were not balloon-treated. After balloon deflation, the catheter was removed. Fluorescence-labelled GPVI-Fc (50 µg/mL) was injected into the injured and control intact vessels, and the opened vessels were sealed by clamps. After incubation, the vessels were rinsed with PBS, and optical imaging was performed to measure GPVI-Fc binding to injured endothelium. The optical data corresponding to the mean detected optical signal of the regions of interest were corrected by subtracting the mean data of the background fluorescence (arbitrary units). After denudation, fluorescence was enhanced in injured femoral and carotid arteries when compared to intact femoral (41.1 ± 17.5 vs. 14.6 ± 6.5; P = 0.021) and carotid (30.2 ± 7.6 vs. 7.9 ± 3.9; P = 0.005) arteries. This preclinical GPVI-Fc-based vascular lesion imaging approach may be the first step towards a method that allows identification of vascular lesions in vivo.


Subject(s)
Carotid Artery Injuries/pathology , Endothelium, Vascular/pathology , Femoral Artery/pathology , Fluorescein Angiography/methods , Immunoglobulin Fc Fragments/pharmacology , Platelet Membrane Glycoproteins/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Carotid Arteries/pathology , Endothelium, Vascular/injuries , Female , Femoral Artery/injuries , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Platelet Membrane Glycoproteins/chemistry , Platelet Membrane Glycoproteins/genetics , Rabbits , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
11.
J Biol Chem ; 285(30): 23410-9, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20479008

ABSTRACT

Arterial thrombosis, a major cause of myocardial infarction and stroke, is initiated by activation of blood platelets by subendothelial collagen. The protein kinase C (PKC) family centrally regulates platelet activation, and it is becoming clear that the individual PKC isoforms play distinct roles, some of which oppose each other. Here, for the first time, we address all four of the major platelet-expressed PKC isoforms, determining their comparative roles in regulating platelet adhesion to collagen and their subsequent activation under physiological flow conditions. Using mouse gene knock-out and pharmacological approaches in human platelets, we show that collagen-dependent alpha-granule secretion and thrombus formation are mediated by the conventional PKC isoforms, PKCalpha and PKCbeta, whereas the novel isoform, PKC, negatively regulates these events. PKCdelta also negatively regulates thrombus formation but not alpha-granule secretion. In addition, we demonstrate for the first time that individual PKC isoforms differentially regulate platelet calcium signaling and exposure of phosphatidylserine under flow. Although platelet deficient in PKCalpha or PKCbeta showed reduced calcium signaling and phosphatidylserine exposure, these responses were enhanced in the absence of PKC. In summary therefore, this direct comparison between individual subtypes of PKC, by standardized methodology under flow conditions, reveals that the four major PKCs expressed in platelets play distinct non-redundant roles, where conventional PKCs promote and novel PKCs inhibit thrombus formation on collagen.


Subject(s)
Blood Platelets/enzymology , Collagen/pharmacology , Protein Kinase C/metabolism , Thrombosis/blood , Thrombosis/enzymology , Animals , Anticoagulants/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Platelets/physiology , Calcium Signaling/drug effects , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/deficiency , Isoenzymes/metabolism , Mice , Platelet Activation/drug effects , Platelet Membrane Glycoproteins/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/deficiency , Thrombosis/chemically induced , Thrombosis/physiopathology
12.
Thromb Res ; 123(2): 236-43, 2008.
Article in English | MEDLINE | ID: mdl-18452977

ABSTRACT

BACKGROUND: Drug eluting stents have reduced the incidence of restenosis after percutaneous coronary interventions significantly, but cause concern about long term safety. Local drug delivery using special application catheters is an alternative approach for intracoronary pharmacotherapy. Besides the fact, that no problematic coating as drug carrier has to be used, a local delivery independent of the stent itself by using catheter techniques offers further advantages - such as the possibility to treat the whole vessel wall, stent edges and adjacent vessel segments and not only the area close to the stent struts. METHODS AND RESULTS: We have developed a new local catheter-based delivery system for local intracoronary pharmacotherapy. An antithrombotic as well as an antiproliferative therapy concept for prevention of restenosis are presented in the manuscript. Our data show that local drug delivery of platelet glycoprotein VI and paclitaxel were effective in the reduction of thrombus formation and neointima formation in experimental animal models. CONCLUSIONS: A combination of early antithrombotic and antiatherosclerotic mechanisms may be a realistic and effective approach to minimize postinterventional thromboischemic events and neointima formation. These results may contribute to an advanced and even combined local intracoronary pharmacotherapy in near future, independent of stent coatings.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Catheterization/instrumentation , Cell Proliferation/drug effects , Coronary Restenosis/prevention & control , Drug Delivery Systems/instrumentation , Fibrinolytic Agents/administration & dosage , Paclitaxel/administration & dosage , Platelet Membrane Glycoproteins/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Coronary Vessels/drug effects , Coronary Vessels/pathology , Fibrinolytic Agents/pharmacology , Paclitaxel/pharmacology , Platelet Membrane Glycoproteins/pharmacology , Solubility , Swine , Time Factors , Tunica Intima/drug effects , Tunica Intima/pathology
13.
Thromb Haemost ; 95(5): 763-6, 2006 May.
Article in English | MEDLINE | ID: mdl-16676064

ABSTRACT

Platelet-mediated thrombus formation at the site of vascular injury is a major trigger for thrombo-ischemic complications after coronary interventions. The platelet collagen receptor glycoprotein VI (GPVI) plays a critical role in the initiation of arterial thrombus formation. Endothelial denudation of the right carotid artery in rabbits was induced through balloon injury. Subsequently, local delivery of soluble, dimeric fusion protein of GPVI (GPVI-Fc) (n = 7) or control Fc (n = 7) at the site of vascular injury was performed with a modified double-balloon drug-delivery catheter. Thrombus area within the injured carotid artery was quantified using a computer-assisted image analysis and was used as index of thrombus formation. The extent of thrombus formation was significantly reduced in GPVI-Fc- compared with control Fc-treated carotid arteries (relative thrombus area, GPVI-Fc vs. Fc: 9.3 +/- 4.2 vs. 2.3 +/- 1.7, p < 0.001). Local delivery of soluble GPVI resulted in reduced thrombus formation after catheter-induced vascular injury. These data suggest a selective pharmacological modulation of GPVI-collagen interactions to be important for controlling onset and progression of pathological arterial thrombosis, predominantly or even exclusively at sites of injured carotid arteries in the absence of systemic platelet therapy.


Subject(s)
Platelet Membrane Glycoproteins/administration & dosage , Thrombosis/prevention & control , Animals , Carotid Artery Injuries , Catheterization , Drug Delivery Systems , Endothelium, Vascular/injuries , Platelet Membrane Glycoproteins/pharmacology , Rabbits , Solubility , Thrombosis/drug therapy
14.
Blood ; 105(4): 1492-9, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15507524

ABSTRACT

Glycoprotein VI (GPVI) is an essential platelet collagen receptor; therefore, the inhibition of GPVI-collagen interactions may be an attractive antithrombotic strategy. We have previously shown that targeting of GPVI with antibodies leads to the depletion of the receptor and to long-term antithrombotic protection in mice. An alternative agent to interfere with GPVI-collagen interactions might be soluble GPVI acting as a competitive inhibitor, thereby averting undesired effects on platelets. To test this, we expressed soluble dimeric human GPVI, comprising the extracellular domain of the receptor fused to the human immunoglobulin Fc domain (GPVI-Fc), and compared its antithrombotic potential with that of anti-GPVI antibodies in mice. In contrast to a recent report, we found by intravital fluorescence microscopy and ultrasonic flow measurements that GPVI-Fc had no effect on platelet adhesion and thrombus formation at the injured arterial wall, whereas anti-GPVI antibodies profoundly inhibited these processes. Similar results were obtained with a fusion protein comprising the extracellular domain of mouse GPVI and human IgG-Fc. This indicates that direct targeting of GPVI provides significantly stronger protection against arterial thrombosis than soluble GPVI dimer.


Subject(s)
Fibrinolytic Agents/pharmacology , Isoantibodies/pharmacology , Platelet Membrane Glycoproteins/immunology , Platelet Membrane Glycoproteins/pharmacology , Animals , Binding Sites, Antibody , Binding, Competitive/physiology , Carotid Artery Injuries/blood , Carotid Artery Injuries/immunology , Carotid Artery Injuries/therapy , Collagen/metabolism , Collagen/pharmacology , Dimerization , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/therapeutic use , Humans , Immunoglobulin Fc Fragments/metabolism , Injections, Intravenous , Isoantibodies/metabolism , Isoantibodies/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Adhesiveness/physiology , Platelet Aggregation/physiology , Platelet Aggregation Inhibitors/metabolism , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation Inhibitors/therapeutic use , Platelet Membrane Glycoproteins/genetics , Platelet Membrane Glycoproteins/therapeutic use , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Solubility , Thrombosis/blood , Thrombosis/immunology , Thrombosis/prevention & control
15.
J Thromb Haemost ; 1(11): 2419-26, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14629478

ABSTRACT

BACKGROUND: The adapter protein c-Cbl has emerged as having a potential role in negative regulation of immune receptor signaling. The major platelet-signaling receptor for collagen, glycoprotein VI (GpVI), is associated with the Fc receptor (FcR) gamma-chain, and signals through a similar pathway to immune receptors. c-Cbl is tyrosine-phosphorylated in response to stimulation of GpVI, whereas phosphorylation of c-Cbl in thrombin-activated platelets is dependent on fibrinogen binding to the integrin GpIIb/IIIa. OBJECTIVE: To investigate the role of c-Cbl in platelet signaling. METHODS: Murine platelets lacking functional c-Cbl or Src family kinases were analyzed. RESULTS: Phosphorylation of c-Cbl through GpVI is reduced in murine platelets deficient in the Src-family kinases Fyn and Lyn, demonstrating that they lie upstream of c-Cbl phosphorylation. Phosphorylation of several proteins of the GpVI-signaling pathway, including the FcR gamma-chain, Syk and phospholipase Cgamma2 (PLCgamma2), is increased in the absence of c-Cbl. In line with this, aggregation is potentiated in response to the GpVI-specific collagen-related peptide (CRP) after a slight delay. A delay in potentiation is also seen in response to stimulation by thrombin. CONCLUSIONS: These observations demonstrate that c-Cbl negatively regulates platelet responses to GpVI agonists and to thrombin, with the latter effect possibly being mediated downstream of GpIIb/IIIa. c-Cbl may play a physiological role in helping to prevent unwanted platelet activation in vivo.


Subject(s)
Platelet Activation , Platelet Membrane Glycoproteins/pharmacology , Proto-Oncogene Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Adaptor Proteins, Vesicular Transport/physiology , Animals , Blood Platelets , Down-Regulation , Humans , Mice , Mice, Inbred Strains , Phosphorylation , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Membrane Glycoproteins/agonists , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins c-cbl , Signal Transduction , Thrombin/pharmacology , Ubiquitin-Protein Ligases/deficiency
16.
Toxicon ; 42(2): 183-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12906889

ABSTRACT

Recent data suggest that diarrhea caused by Vibrio cholerae involves a pro-inflammatory mediators release, such as cytokines, prostaglandin and nitric oxide. The aim of this study was to investigate the role of mast cells and their mediators in the intestinal secretion induced by cholera toxin. We examined the dose responses, time course and role of mast cells and pro-inflammatory mediators in cholera toxin intestinal secretory response, in vivo. Cholera toxin caused a dose-dependent secretion, in ligated small intestine loops, at 18 h. Rats treated with 48/80 compound or ketotifen had a significant decrease in the intestinal secretory response. Cholera toxin secretion was significantly reduced by an unspecific histamine/serotonin receptor antagonist, histamine receptor antagonist, phospholipase A2 and cyclooxygenase inhibitors, platelet-activating factor (PAF) receptor antagonists and TNF-alpha synthesis blockers. On the other hand, pretreatment with a specific serotonin receptor antagonist and lipoxygenase inhibitors failed to block this effect. Analysis of the intestinal fluid from rats injected with cholera toxin, revealed that cholera toxin induces the release of IL-1beta and TNF-alpha into fluid. The data suggest that, at least in part, mast cells are involved in cholera toxin-induced secretion, as well as point to the importance of histamine, prostaglandins, PAF, IL-1beta and TNF-alpha in this process.


Subject(s)
Cholera Toxin/toxicity , Intestine, Small/drug effects , Intestine, Small/metabolism , Mast Cells/metabolism , Receptors, G-Protein-Coupled , Animals , Cholera Toxin/antagonists & inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Histamine Antagonists/pharmacology , Inflammation/metabolism , Interleukin-1/metabolism , Phospholipases A/antagonists & inhibitors , Phospholipases A/pharmacology , Phospholipases A2 , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/pharmacology , Rats , Receptors, Cell Surface/antagonists & inhibitors , Serotonin Antagonists/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Vibrio cholerae
17.
J Biol Chem ; 278(39): 37520-9, 2003 Sep 26.
Article in English | MEDLINE | ID: mdl-12832411

ABSTRACT

The interaction of fibrinogen with the integrin alphaIIbbeta3 plays a crucial role in platelet adhesion and platelet activation leading to the generation of intracellular signals that nucleate the reorganization of the cytoskeleton. Presently, we have only a limited understanding of the signaling cascades and effector proteins through which changes in the cytoskeletal architecture are mediated. The present study identifies phospholipase Cgamma2 (PLCgamma2) as an important target of the Src-dependent signaling cascade regulated by alphaIIbbeta3. Real time phasecontrast microscopy is used to show that formation of filopodia and lamellapodia in murine platelets on a fibrinogen surface is dramatically inhibited in the absence of PLCgamma2. Significantly, the formation of these structures is mediated by Ca2+ elevation and activation of protein kinase C, both directly regulated by PLC activity. With the involvement of Syk, SLP-76, and Btk, alphaIIbbeta3-induced PLCgamma2 activation partly overlaps with the pathway used by the collagen receptor glycoprotein VI. Important differences, however, exist between the two signaling cascades in that activation of PLCgamma2 by alphaIIbbeta3 is unaltered in murine platelets, which lack the FcR gamma-chain or the adaptor LAT, but is abolished in the presence of cytochalasin D. Therefore, PLCgamma2 plays not only a crucial role in activation of alphaIIbbeta3 by collagen receptors but also in alphaIIbbeta3-mediated responses.


Subject(s)
Adaptor Proteins, Signal Transducing , Blood Platelets/physiology , Membrane Proteins , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Type C Phospholipases/physiology , Animals , Calcium/metabolism , Carrier Proteins/physiology , Cytochalasin D/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Mice , Mice, Inbred C57BL , Phospholipase C gamma , Phosphoproteins/physiology , Phosphorylation , Platelet Membrane Glycoproteins/pharmacology , Protein-Tyrosine Kinases/physiology
18.
Blood ; 102(4): 1367-73, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12730118

ABSTRACT

Collagen stimulates platelet activation through a tyrosine kinase-based pathway downstream of the glycoprotein VI (GPVI)-Fc receptor (FcR) gamma-chain complex. Genetic ablation of FcR gamma-chain results in a complete inhibition of aggregation to collagen. In contrast, a steady increase in light transmission is induced by collagen in phospholipase Cgamma2-deficient (PLCgamma2-/-) platelets in a Born aggregometer, indicating a weak level of activation. This increase is inhibited partially in the presence of an alpha2beta1-blocking antibody or an alphaIIbbeta3 antagonist and completely by a combination of the 2 inhibitors. It is also abolished by the Src kinase inhibitor PP1 and reduced in the presence of the phosphatidylinositol (PI) 3-kinase inhibitor wortmannin. The GPVI-specific agonists convulxin and collagen-related peptide (CRP) also stimulate weak aggregation in PLCgamma2-/- platelets, which is inhibited by wortmannin and PP1. Collagen and CRP stimulate tyrosine phosphorylation of PLCgamma1 at its regulatory site, Tyr 783, in murine but not in human platelets through a Src kinase-dependent pathway. Adhesion of PLCgamma2-/- platelets to a collagen monolayer is severely reduced at a shear rate of 800 s-1, relative to controls, whereas it is abolished in FcR gamma-chain-/- platelets. These results provide strong evidence that engagement of GPVI stimulates limited integrin activation in PLCgamma2-/- platelets via PLCgamma1 and PI3-kinase.


Subject(s)
Blood Platelets/enzymology , Integrin alpha2beta1/metabolism , Lectins, C-Type , Phosphatidylinositol 3-Kinases/blood , Platelet Activation/physiology , Platelet Membrane Glycoproteins/agonists , Type C Phospholipases/blood , Animals , Blood Platelets/drug effects , Collagen/metabolism , Collagen/pharmacology , Crotalid Venoms/pharmacology , Enzyme Inhibitors/pharmacology , Fibrinogen/metabolism , Humans , Integrin alpha2beta1/antagonists & inhibitors , Mice , Phosphoinositide-3 Kinase Inhibitors , Phospholipase C gamma , Phosphorylation , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors , Platelet Membrane Glycoproteins/pharmacology , Receptors, Fc/metabolism , Type C Phospholipases/deficiency , Tyrosine/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
19.
Blood ; 101(4): 1392-9, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12393506

ABSTRACT

Protein kinase D (PKD, also known as PKCmu) is closely related to the protein kinase C superfamily but is differentially regulated and has a distinct catalytic domain that shares homology with Ca(2+)-dependent protein kinases. PKD is highly expressed in hematopoietic cells and undergoes rapid and sustained activation upon stimulation of immune receptors. PKD is regulated through phosphorylation by protein kinase C (PKC). In the present study, we show that PKD is expressed in human platelets and that it is rapidly activated by receptors coupled to heterotrimeric G-proteins or tyrosine kinases. Activation of PKD is mediated downstream of PKC. Strong agonists such as convulxin, which acts on GPVI, and thrombin cause sustained activation of PKC and PKD, whereas the thromboxane mimetic U46619 gives rise to transient activation of PKC and PKD. Activation of PKD by submaximal concentrations of phospholipase C-coupled receptor agonists is potentiated by G(i)-coupled receptors (eg, adenosine diphosphate and epinephrine). This study shows that PKD is rapidly activated by a wide variety of platelet agonists through a PKC-dependent pathway. Activation of PKD enables phosphorylation of a distinct set of substrates to those targeted by PKC in platelets.


Subject(s)
Blood Platelets/enzymology , Lectins, C-Type , Protein Kinase C/blood , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Adenosine Diphosphate/pharmacology , Blotting, Western , Crotalid Venoms/pharmacology , Drug Synergism , Enzyme Activation/drug effects , Epinephrine/pharmacology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Heterotrimeric GTP-Binding Proteins/physiology , Humans , Immunosorbent Techniques , Phosphorylation , Platelet Membrane Glycoproteins/pharmacology , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Thrombin/pharmacology
20.
Neurol Neurochir Pol ; 36(4): 801-8, 2002.
Article in Polish | MEDLINE | ID: mdl-12418143

ABSTRACT

This review describes the role of platelet activating factor (PAF) in the central nervous system injury. Cerebral ischaemia, traumatic injury of central nervous system, metabolic, toxic and degenerative neuropathy, and also the increase in Ca2+ concentration in the cell, are strong stimulators of PAF synthesis and its release from cell membranes. Neurons, glial and microglial cells, monocyte cell populations, macrophages and endothelial cells of blood vessels are the targets of platelet activating factor. The release of PAF leads to ischaemia of nervous tissue, acute traumatic or nontraumatic injuries, degenerative and metabolic nervous system disorders in adults. The use of PAF receptor antagonists prevents partially cell injury in central nervous system and leukocyte adhesion to endothelial cells.


Subject(s)
Central Nervous System Diseases/metabolism , Central Nervous System Diseases/physiopathology , Neurotransmitter Agents/metabolism , Platelet Activating Factor/metabolism , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, Cell Surface/antagonists & inhibitors , Receptors, G-Protein-Coupled , Humans , Platelet Membrane Glycoproteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL