Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters

Affiliation country
Publication year range
1.
Blood ; 139(17): 2691-2705, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35313337

ABSTRACT

The prevention and treatment of arterial thrombosis continue to be clinically challenging, and understanding the relevant molecular mechanisms in detail may facilitate the quest to identify novel targets and therapeutic approaches that improve protection from ischemic and bleeding events. The chemokine CXCL12 augments collagen-induced platelet aggregation by activating its receptor CXCR4. Here we show that inhibition of CXCR4 attenuates platelet aggregation induced by collagen or human plaque homogenate under static and arterial flow conditions by antagonizing the action of platelet-secreted CXCL12. We further show that platelet-specific CXCL12 deficiency in mice limits arterial thrombosis by affecting thrombus growth and stability without increasing tail bleeding time. Accordingly, neointimal lesion formation after carotid artery injury was attenuated in these mice. Mechanistically, CXCL12 activated via CXCR4 a signaling cascade involving Bruton's tyrosine kinase (Btk) that led to integrin αIIbß3 activation, platelet aggregation, and granule release. The heterodimeric interaction between CXCL12 and CCL5 can inhibit CXCL12-mediated effects as mimicked by CCL5-derived peptides such as [VREY]4. An improved variant of this peptide, i[VREY]4, binds to CXCL12 in a complex with CXCR4 on the surface of activated platelets, thereby inhibiting Btk activation and preventing platelet CXCL12-dependent arterial thrombosis. In contrast to standard antiplatelet therapies such as aspirin or P2Y12 inhibition, i[VREY]4 reduced CXCL12-induced platelet aggregation and yet did not prolong in vitro bleeding time. We provide evidence that platelet-derived CXCL12 is involved in arterial thrombosis and can be specifically targeted by peptides that harbor potential therapeutic value against atherothrombosis.


Subject(s)
Blood Platelets , Thrombosis , Agammaglobulinaemia Tyrosine Kinase/metabolism , Animals , Blood Platelets/metabolism , Chemokine CXCL12/metabolism , Collagen/metabolism , Mice , Platelet Activation , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thrombosis/metabolism
2.
Circ Res ; 126(4): 486-500, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31859592

ABSTRACT

RATIONALE: A reduced rate of myocardial infarction has been reported in patients with atrial fibrillation treated with FXa (factor Xa) inhibitors including rivaroxaban compared with vitamin K antagonists. At the same time, low-dose rivaroxaban has been shown to reduce mortality and atherothrombotic events in patients with coronary artery disease. Yet, the mechanisms underlying this reduction remain unknown. OBJECTIVE: In this study, we hypothesized that rivaroxaban's antithrombotic potential is linked to a hitherto unknown rivaroxaban effect that impacts on platelet reactivity and arterial thrombosis. METHODS AND RESULTS: In this study, we identified FXa as potent, direct agonist of the PAR-1 (protease-activated receptor 1), leading to platelet activation and thrombus formation, which can be inhibited by rivaroxaban. We found that rivaroxaban reduced arterial thrombus stability in a mouse model of arterial thrombosis using intravital microscopy. For in vitro studies, atrial fibrillation patients on permanent rivaroxaban treatment for stroke prevention, respective controls, and patients with new-onset atrial fibrillation before and after first intake of rivaroxaban (time series analysis) were recruited. Platelet aggregation responses, as well as thrombus formation under arterial flow conditions on collagen and atherosclerotic plaque material, were attenuated by rivaroxaban. We show that rivaroxaban's antiplatelet effect is plasma dependent but independent of thrombin and rivaroxaban's anticoagulatory capacity. CONCLUSIONS: Here, we identified FXa as potent platelet agonist that acts through PAR-1. Therefore, rivaroxaban exerts an antiplatelet effect that together with its well-known potent anticoagulatory capacity might lead to reduced frequency of atherothrombotic events and improved outcome in patients.


Subject(s)
Arteries/metabolism , Blood Platelets/drug effects , Factor Xa/pharmacology , Receptor, PAR-1/agonists , Rivaroxaban/pharmacology , Thrombosis/prevention & control , Animals , Arteries/pathology , Blood Platelets/metabolism , Factor Xa Inhibitors/pharmacology , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacology , Humans , Mice, Inbred C57BL , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Receptor, PAR-1/metabolism , Rivaroxaban/administration & dosage , Thrombosis/metabolism
3.
Blood ; 131(24): 2605-2616, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29559479

ABSTRACT

Interaction of von Willebrand factor (VWF) with platelet glycoprotein Ib (GPIb) and interaction of collagen with GPVI are essential for thrombus formation on ruptured or eroded atherosclerotic plaques (atherothrombosis). GPIb and GPVI signal through Bruton tyrosine kinase (Btk), which can be blocked irreversibly by oral application of ibrutinib, an established therapy for chronic lymphocytic leukemia (CLL) with long-term safety. We found that ibrutinib and the novel Btk inhibitors acalabrutinib and ONO/GS-4059 block GPVI-dependent static platelet aggregation in blood exposed to human plaque homogenate and collagen but not to ADP or arachidonic acid. Moreover, Btk inhibitors prevented platelet thrombus formation on human atherosclerotic plaque homogenate and plaque tissue sections from arterially flowing blood, whereas integrin α2ß1 and VWF-dependent platelet adhesion to collagen, which is important for physiologic hemostasis, was not affected. This plaque-selective platelet inhibition was also observed in CLL patients taking 450 mg of ibrutinib and in volunteers after much lower and intermittent dosing of the drug. We conclude that Btk inhibitors, by targeting GPIb and GPVI signal transduction, suppress platelet thrombus accretion from flowing blood on atherosclerotic plaque but spare hemostatic platelet function. Btk inhibitors hold promise as the first culprit lesion-focused oral antiplatelet drugs and are effective at low doses.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Benzamides/therapeutic use , Imidazoles/therapeutic use , Plaque, Atherosclerotic/complications , Platelet Aggregation Inhibitors/therapeutic use , Pyrazines/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Thrombosis/etiology , Thrombosis/prevention & control , Adenine/analogs & derivatives , Administration, Oral , Adult , Agammaglobulinaemia Tyrosine Kinase/metabolism , Aged , Benzamides/administration & dosage , Humans , Imidazoles/administration & dosage , Male , Middle Aged , Piperidines , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/therapeutic use , Pyrazines/administration & dosage , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Thrombosis/metabolism , Thrombosis/pathology
5.
Cardiovasc Res ; 119(1): 155-166, 2023 03 17.
Article in English | MEDLINE | ID: mdl-35238350

ABSTRACT

AIMS: Atherosclerosis is a chronic inflammatory disease of the arteries leading to the formation of atheromatous plaques. Human mesenchymal stem cells (hMSCs) are recruited from the circulation into plaques where in response to their environment they adopt a phenotype with immunomodulatory properties. However, the mechanisms underlying hMSC function in these processes are unclear. Recently, we described that miRNA let-7f controls hMSC invasion guided by inflammatory cytokines and chemokines. Here, we investigated the role of let-7f in hMSC tropism to human atheromas and the effects of the plaque microenvironment on cell fate and release of soluble factors. METHODS AND RESULTS: Incubation of hMSCs with LL-37, an antimicrobial peptide abundantly found in plaques, increased biosynthesis of let-7f and N-formyl peptide receptor 2 (FPR2), enabling chemotactic invasion of the cells towards LL-37, as determined by qRT-PCR, flow cytometry, and cell invasion assay analysis. In an Apoe-/- mouse model of atherosclerosis, circulating hMSCs preferentially adhered to athero-prone endothelium. This property was facilitated by elevated levels of let-7f in the hMSCs, as assayed by ex vivo artery perfusion and two-photon laser scanning microscopy. Exposure of hMSCs to homogenized human atheromatous plaque material considerably induced the production of various cytokines, chemokines, matrix metalloproteinases, and tissue inhibitors of metalloproteinases, as studied by PCR array and western blot analysis. Moreover, exposure to human plaque extracts elicited differentiation of hMSCs into cells of the myogenic lineage, suggesting a potentially plaque-stabilizing effect. CONCLUSIONS: Our findings indicate that let-7f promotes hMSC tropism towards atheromas through the LL-37/FPR2 axis and demonstrate that hMSCs upon contact with human plaque environment develop a potentially athero-protective signature impacting the pathophysiology of atherosclerosis.


Subject(s)
Atherosclerosis , Mesenchymal Stem Cells , MicroRNAs , Plaque, Atherosclerotic , Mice , Animals , Humans , MicroRNAs/genetics , Atherosclerosis/genetics , Cytokines , Immunologic Factors
6.
J Thromb Thrombolysis ; 32(2): 158-66, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21424266

ABSTRACT

Atherosclerosis has an important inflammatory component. Macrophages accumulating in atherosclerotic arteries produce prostaglandin E(2) (PGE(2)), a main inflammatory mediator. Platelets express inhibitory receptors (EP(2), EP(4)) and a stimulatory receptor (EP(3)) for this prostanoid. Recently, it has been reported in ApoE(-/-) mice that PGE(2) accumulating in inflammatory atherosclerotic lesions might contribute to atherothrombosis after plaque rupture by activating platelet EP(3), and EP(3) blockade has been proposed to be a promising new approach in anti-thrombotic therapy. The aim of our investigation was to study the role of PGE(2) in human atherosclerotic plaques on human platelet function and thrombus formation. Plaque PGE(2) might either activate or inhibit platelets depending on stimulation of either EP(3) or EP(4), respectively. We found that the two EP(3)-antagonists AE5-599 (300 nM) and AE3-240 (300 nM) specifically and completely inhibited the synergistic effect of the EP(3)-agonist sulprostone on U46619-induced platelet aggregation in blood. However, these two EP(3)-antagonists neither inhibited atherosclerotic plaque-induced platelet aggregation, GPIIb/IIIa exposure, dense and alpha granule secretion in blood nor reduced plaque-induced platelet thrombus formation under arterial flow. The EP(4)-antagonist AE3-208 (1-3 µM) potentiated in combination with PGE(2) (1 µM) ADP-induced aggregation, demonstrating that PGE(2) enhances platelet aggregation when the inhibitory EP(4)-receptor is inactivated. However, plaque-induced platelet aggregation was not augmented after platelet pre-treatment with AE3-208, indicating that plaque PGE(2) does not stimulate the EP(4)-receptor. We found that PGE(2) was present in plaques only at very low levels (15 pg PGE(2)/mg plaque). We conclude that PGE(2) in human atherosclerotic lesions does not modulate (i.e. stimulate or inhibit) atherothrombosis in blood after plaque rupture.


Subject(s)
Blood Platelets/metabolism , Carotid Stenosis/metabolism , Dinoprostone/metabolism , Plaque, Atherosclerotic/metabolism , Platelet Aggregation , Receptors, Prostaglandin E, EP3 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Thrombosis/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Abortifacient Agents, Nonsteroidal/pharmacology , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Blood Platelets/pathology , Carotid Stenosis/genetics , Carotid Stenosis/pathology , Dinoprostone/analogs & derivatives , Dinoprostone/pharmacology , Female , Humans , Male , Mice , Mice, Knockout , Naphthalenes , Phenylbutyrates , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Receptors, Prostaglandin E, EP3 Subtype/agonists , Receptors, Prostaglandin E, EP3 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP3 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/agonists , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/genetics , Rupture, Spontaneous , Thrombosis/genetics , Vasoconstrictor Agents/pharmacology
7.
Front Cardiovasc Med ; 8: 749022, 2021.
Article in English | MEDLINE | ID: mdl-34631841

ABSTRACT

Background: Bruton tyrosine kinase inhibitors (BTKi) are used in B-cell malignancies and in development against various autoimmune diseases. Since Btk is also involved in specific pathways of platelet activation, BTKi might be considered to target platelet GPVI/GPIb-mediated atherothrombosis and platelet FcγRIIA-dependent immune disorders. However, BTKi treatment of patients with B-cell malignancies is frequently associated with mild bleeding events caused possibly by off-target inhibition of Tec. Here, we compared the platelet effects of two novel BTKi that exhibit a high (remibrutinib) or low (rilzabrutinib) selectivity for Btk over Tec. Methods and Results: Remibrutinib and rilzabrutinib were pre-incubated with anticoagulated blood. Platelet aggregation and in vitro bleeding time (closure time) were studied by multiple electrode aggregometry (MEA) and platelet-function analyzer-200 (PFA-200), respectively. Both BTKi inhibited atherosclerotic plaque-stimulated GPVI-mediated platelet aggregation, remibrutinib being more potent (IC50 = 0.03 µM) than rilzabrutinib (IC50 = 0.16 µM). Concentrations of remibrutinib (0.1 µM) and rilzabrutinib (0.5 µM), >80% inhibitory for plaque-induced aggregation, also significantly suppressed (>90%) the Btk-dependent pathways of platelet aggregation upon GPVI, von Willebrand factor/GPIb and FcγRIIA activation stimulated by low collagen concentrations, ristocetin and antibody cross-linking, respectively. Both BTKi did not inhibit aggregation stimulated by ADP, TRAP-6 or arachidonic acid. Remibrutinib (0.1 µM) only slightly prolonged closure time and significantly less than rilzabrutinib (0.5 µM). Conclusion: Remibrutinib and rilzabrutinib inhibit Btk-dependent pathways of platelet aggregation upon GPVI, VWF/GPIb, and FcγRIIA activation. Remibrutinib being more potent and showing a better profile of inhibition of Btk-dependent platelet activation vs. hemostatic impairment than rilzabrutinib may be considered for further development as an antiplatelet drug.

8.
J Transl Med ; 8: 128, 2010 Dec 06.
Article in English | MEDLINE | ID: mdl-21134286

ABSTRACT

BACKGROUND: Platelet activation requires rapid remodeling of the actin cytoskeleton which is regulated by small GTP-binding proteins. By using the Rac1-specific inhibitor NSC23766, we have recently found that Rac1 is a central component of a signaling pathway that regulates dephosphorylation and activation of the actin-dynamising protein cofilin, dense and α-granule secretion, and subsequent aggregation of thrombin-stimulated washed platelets. OBJECTIVES: To study whether NSC23766 inhibits stimulus-induced platelet secretion and aggregation in blood. METHODS: Human platelet aggregation and ATP-secretion were measured in hirudin-anticoagulated blood and platelet-rich plasma (PRP) by using multiple electrode aggregometry and the Lumi-aggregometer. Platelet P-selectin expression was quantified by flow cytometry. RESULTS: NSC23766 (300 µM) inhibited TRAP-, collagen-, atherosclerotic plaque-, and ADP-induced platelet aggregation in blood by 95.1%, 93.4%, 92.6%, and 70%, respectively. The IC50 values for inhibition of TRAP-, collagen-, and atherosclerotic plaque-, were 50 ± 18 µM, 64 ± 35 µM, and 50 ± 30 µM NSC23766 (mean ± SD, n = 3-7), respectively. In blood containing RGDS to block integrin αIIbß3-mediated platelet aggregation, NSC23766 (300 µM) completely inhibited P-selectin expression and reduced ATP-secretion after TRAP and collagen stimulation by 73% and 85%, respectively. In ADP-stimulated PRP, NSC23766 almost completely inhibited P-selectin expression, in contrast to aspirin, which was ineffective. Moreover, NSC23766 (300 µM) decreased plaque-stimulated platelet adhesion/aggregate formation under arterial flow conditions (1500s-1) by 72%. CONCLUSIONS: Rac1-mediated signaling plays a central role in secretion-dependent platelet aggregation in blood stimulated by a wide array of platelet agonists including atherosclerotic plaque. By specifically inhibiting platelet secretion, the pharmacological targeting of Rac1 could be an interesting approach in the development of future antiplatelet drugs.


Subject(s)
Adenosine Triphosphate/metabolism , Plaque, Atherosclerotic/pathology , Platelet Aggregation/drug effects , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Acid Phosphatase/metabolism , Adenosine Diphosphate/pharmacology , Aminoquinolines/pharmacology , Blood Platelets/drug effects , Blood Platelets/enzymology , Collagen/pharmacology , Humans , Isoenzymes/metabolism , P-Selectin/metabolism , Platelet-Rich Plasma/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Tartrate-Resistant Acid Phosphatase , Thrombosis/pathology , Thrombosis/physiopathology
9.
J Vasc Surg ; 48(5): 1217-22, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18692349

ABSTRACT

BACKGROUND: Endothelial progenitor cells (EPC) contribute to vascular regeneration. Since surgical injury and burns induce a pro-inflammatory and proangiogenic response, we investigated the effect of vascular injury with minimal surgical trauma after endarterectomy on the number of circulating EPC and systemic inflammatory changes. METHODS AND RESULTS: Forty-five patients with peripheral arterial occlusive disease were included in the study. Venous blood samples were taken before and 1 day after endarterectomy and plaque material was obtained. Ten patients with minor surgery served as controls. Circulating CD133+CD34+, VEGFR-2+CD34+ progenitor cells and surface expression of CD11b on circulating neutrophils were analysed using flow cytometry. EPCs were characterized in a culture assay as double-positive for DiI-LDL uptake and lectin binding. Cytokine concentrations of interleukin (IL)-6, IL-8, TNF-alpha, IL-1ss, IL-10, IL-12, SDF-1, G-CSF, and VEGF were measured in plasma and tissue samples. After endarterectomy a significant decrease in circulating EPC, CD133+CD34+, and VEGFR-2+CD34+ cells was observed. This was associated with a specific pattern of changes in circulating cytokine levels after endarterectomy with a decrease in IL-1 beta and IL-12, an increase in IL-6 and G-CSF plasma concentrations, and surface expression of CD11b on circulating neutrophils. In contrast, after minor surgery an increase in circulating CD133+CD34+ cells, IL-6, IL-8, and IL-10 was found. Interestingly there was a negative association between levels of local IL-6 within the plaque and only the preoperative levels of circulating CD133+C34+. CONCLUSION: Endarterectomy induces changes in circulating cytokines and a decline in circulating progenitor cells, which may be due to recruitment of progenitor cells to the injured vessels. This is supported by the negative association between plaque inflammation and circulating progenitor cells before endarterectomy.


Subject(s)
Arterial Occlusive Diseases/pathology , Atherosclerosis/pathology , Endarterectomy , Endothelial Cells/pathology , Peripheral Vascular Diseases/pathology , Stem Cells/pathology , AC133 Antigen , Aged , Antigens, CD/blood , Antigens, CD34/blood , Arterial Occlusive Diseases/immunology , Arterial Occlusive Diseases/metabolism , Arterial Occlusive Diseases/surgery , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/surgery , CD11b Antigen/blood , Cells, Cultured , Constriction, Pathologic , Cytokines/blood , Endothelial Cells/chemistry , Endothelial Cells/immunology , Female , Glycoproteins/blood , Humans , Male , Peptides/blood , Peripheral Vascular Diseases/immunology , Peripheral Vascular Diseases/metabolism , Peripheral Vascular Diseases/surgery , Stem Cells/chemistry , Stem Cells/immunology , Time Factors , Vascular Endothelial Growth Factor Receptor-2/blood
10.
Arterioscler Thromb Vasc Biol ; 27(4): 769-75, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17255532

ABSTRACT

OBJECTIVE: The serine protease MT-SP1/matriptase plays an important role in cell migration and matrix degradation. Hepatocyte growth factor (HGF), urokinase-type plasminogen activator (uPA), and protease-activated receptor 2 (PAR-2) have been identified as in vitro substrates of MT-SP1/matriptase. Because PAR-2 is expressed in endothelial cells and contributes to inflammatory processes, we sought to investigate the effects of MT-SP1/matriptase on endothelial cytokine expression and analyzed MT-SP1/matriptase expression in vascular cells and atherosclerotic lesions. METHODS AND RESULTS: In endothelial cells, recombinant MT-SP1/matriptase dose-dependently induced interleukin (IL)-8 and IL-6 mRNA and protein expression dependent on its proteolytic activity. MT-SP1/matriptase time-dependently induced phosphorylation of p38 MAPK and p42/44 MAPK. Inhibitor experiments revealed that p38 MAPK and PKCalpha were necessary for IL-8 induction. PAR-2 downregulation abolished and PAR-2 overexpression augmented MT-SP1/matriptase-induced IL-8 expression as evidence for PAR-2 signaling. In human atherectomies, MT-SP1/matriptase was expressed in blood cells adherent to the endothelium. Concordantly, basal MT-SP1/matriptase expression was detected in isolated monocytes. Coincubation of monocytes and endothelial cells resulted in an increased IL-8 release, which was reduced after downregulation of endothelial PAR-2 and monocytic MT-SP1/matriptase. CONCLUSION: MT-SP1/matriptase induces release of proinflammatory cytokines in endothelial cells through activation of PAR-2. MT-SP1/matriptase is expressed in monocytes, thus, interaction of monocytic MT-SP1/matriptase with endothelial PAR-2 may contribute to atherosclerosis.


Subject(s)
Endothelial Cells/metabolism , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Receptor, PAR-2/metabolism , Serine Endopeptidases/pharmacology , Atherosclerosis/enzymology , Atherosclerosis/etiology , Catalysis , Cells, Cultured , Cytokines/biosynthesis , Endothelial Cells/drug effects , Enzyme Activation , Humans , Interleukin-6/genetics , Interleukin-8/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/metabolism , NF-kappa B/metabolism , Protein Kinase C-alpha/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Serine Endopeptidases/blood , Serine Endopeptidases/isolation & purification , Serine Endopeptidases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Thromb Haemost ; 118(2): 351-361, 2018 02.
Article in English | MEDLINE | ID: mdl-29378359

ABSTRACT

Platelet glycoprotein VI (GPVI) acts as a decisive collagen receptor in atherothrombosis. Besides collagen, injured atherosclerotic plaques expose tissue factor (TF) that triggers fibrin formation. Two recent studies reported that platelet GPVI also functions as fibrin receptor, which would importantly widen the mode of action of GPVI-targeted antithrombotic drugs. We studied the binding of two GPVI fusion proteins to fibrin under static and arterial flow conditions. Fibrin was prepared from purified fibrinogen or generated more physiologically from endogenous fibrinogen by coagulating plasma with thrombin. Fibrin formation was also triggered by exposing TF-coated surfaces or human atherosclerotic plaque slices to arterially flowing blood. By binding studies and advanced optical imaging, we found that recombinant dimeric GPVI-Fc fusion proteins with Fc from either IgG1 (GPVI-Fc1) or IgG2 (GPVI-Fc2) bound to collagen fibres, but neither to fibrin prepared from purified fibrinogen obtained from three suppliers, nor to physiological fibrin formed by thrombin in plasma or triggered by exposing TF or atherosclerotic plaque slices to arterially flowing blood. Our findings do not support a role of dimeric platelet GPVI as receptor for fibrin. This is important for the understanding of plaque-triggered platelet thrombus formation and is clinically relevant for future GPVI-targeting therapies with recombinant GPVI-Fc and anti-GPVI antibodies.


Subject(s)
Collagen/metabolism , Fibrin/metabolism , Fibrinogen/metabolism , Platelet Membrane Glycoproteins/metabolism , Thrombin/metabolism , Atherosclerosis/metabolism , Blood Coagulation , Blood Platelets/metabolism , Humans , Microscopy, Fluorescence , Plaque, Atherosclerotic/metabolism , Platelet Activation , Platelet Adhesiveness , Platelet Aggregation , Protein Binding , Protein Multimerization , Recombinant Proteins
12.
Thromb Haemost ; 97(3): 435-43, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17334511

ABSTRACT

Anti-platelet drugs are used to prevent intra-arterial thrombus formation after rupture of atherosclerotic plaques. Until now, the inhibitory effect of present and future anti-platelet drugs such as aspirin, ADP receptor P2Y(1)/P2Y(12) antagonists and glycoprotein (GP) Ibalpha inhibitors on the interaction of platelets with human plaques is not known. To study those effects we obtained human atherosclerotic plaques by surgical endarterectomy. Plaques induced maximal platelet aggregation in hirudinized platelet-rich plasma (PRP) and blood that was effectively inhibited by aspirin, the P2Y(1) antagonist MRS2179 and the P2Y(12) antagonist AR-C69931MX, but not by GPIbalpha blockade with the mAB 6B4. Inhibition of platelet aggregation by MRS2179 was 74 +/- 37% and 68 +/- 20%, by AR-C69931MX 94 +/- 7% and 80 +/- 6%, and by aspirin 88 +/- 19% and 64 +/- 28%, in PRP and blood, respectively (mean +/- SD; n = 6-12 with plaques from 6 patients). The combination of both ADP receptor antagonists completely inhibited plaque-induced platelet aggregation in hirudinized PRP and blood. Under arterial flow conditions (1,500s(-1)), blockade of platelet GPIbalpha resulted in a strong decrease of plaque-stimulated platelet adhesion/aggregate formation of 77 +/- 5% (mean +/- SD; n = 4). Furthermore, MRS2179, AR-C69931MX and their combination reduced plaque-dependent platelet aggregate formation by 35 +/- 14%, 32 +/- 13% and 58 +/- 12% (mean +/- SD; n = 5), respectively. Aspirin was without significant effect. In conclusion, a GPIbalpha-blocking antibody, as well as P2Y(1) and P2Y(12) receptor antagonists, alone or in combination, reduce in contrast to aspirin human plaque-induced platelet thrombus formation under arterial flow. Although these new anti-platelet agents inhibit platelet thrombus formation after plaque rupture, more efficient platelet blockers are required.


Subject(s)
Aspirin/pharmacology , Atherosclerosis/complications , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIb-IX Complex/antagonists & inhibitors , Purinergic P2 Receptor Antagonists , Thrombosis/prevention & control , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Antibodies, Monoclonal/pharmacology , Aspirin/therapeutic use , Atherosclerosis/blood , Blood Coagulation Tests/methods , Blood Platelets/metabolism , Carotid Stenosis/complications , Dose-Response Relationship, Drug , Hemorheology , Humans , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Platelet Glycoprotein GPIb-IX Complex/immunology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Receptors, Purinergic P2/metabolism , Receptors, Purinergic P2Y1 , Receptors, Purinergic P2Y12 , Thrombosis/blood , Thrombosis/etiology , Thrombosis/metabolism , Time Factors , von Willebrand Factor/metabolism
13.
Thromb Haemost ; 117(8): 1651-1659, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28569920

ABSTRACT

The efficiency of current dual antiplatelet therapy might be further improved by its combination with a glycoprotein (GP) VI-targeting strategy without increasing bleeding. GPVI-Fc, a recombinant dimeric fusion protein binding to plaque collagen and concealing binding sites for platelet GPVI, acts as a lesion-focused antiplatelet drug, and does not increase bleeding in vivo. We investigated, whether GPVI-Fc added in vitro on top of acetylsalicylic acid (ASA), the P2Y12 antagonist ticagrelor, and the fibrinogen receptor antagonist abciximab alone or in combination would increase inhibition of platelet activation by atherosclerotic plaque. Under static conditions, GPVI-Fc inhibited plaque-induced platelet aggregation by 53 %, and increased platelet inhibition by ASA (51 %) and ticagrelor (64 %) to 66 % and 80 %, respectively. Under arterial flow, GPVI-Fc inhibited plaque-induced platelet aggregation by 57 %, and significantly increased platelet inhibition by ASA (28 %) and ticagrelor (47 %) to about 81 % each. The triple combination of GPVI-Fc, ASA and ticagrelor achieved almost complete inhibition of plaque-induced platelet aggregation (93 %). GPVI-Fc alone or in combination with ASA or ticagrelor did not increase closure time measured by the platelet function analyzer (PFA)-200. GPVI-Fc added on top of abciximab, a clinically used anti-fibrinogen receptor antibody which blocks platelet aggregation, strongly inhibited total (81 %) and stable (89 %) platelet adhesion. We conclude that GPVI-Fc added on top of single or dual antiplatelet therapy with ASA and/or a P2Y12 antagonist is likely to improve anti-atherothrombotic protection without increasing bleeding risk. In contrast, the strong inhibition of platelet adhesion by GPVI-Fc in combination with GPIIb/IIIa inhibitors could be harmful.


Subject(s)
Adenosine/analogs & derivatives , Antibodies, Monoclonal/pharmacology , Aspirin/pharmacology , Blood Coagulation/drug effects , Blood Platelets/drug effects , Glycoproteins/pharmacology , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Plaque, Atherosclerotic , Platelet Aggregation Inhibitors/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Thrombosis/prevention & control , Abciximab , Adenosine/pharmacology , Adenosine/toxicity , Antibodies, Monoclonal/toxicity , Aspirin/toxicity , Blood Platelets/metabolism , Drug Therapy, Combination , Glycoproteins/toxicity , Hemorrhage/chemically induced , Humans , Immunoglobulin Fab Fragments/toxicity , Immunoglobulin Fc Fragments/toxicity , Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/toxicity , Purinergic P2Y Receptor Antagonists/toxicity , Thrombosis/blood , Thrombosis/pathology , Ticagrelor , Time Factors
14.
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
15.
FASEB J ; 19(8): 898-909, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15923400

ABSTRACT

Lipid-rich atherosclerotic plaques are vulnerable, and their rupture can cause the formation of a platelet- and fibrin-rich thrombus leading to myocardial infarction and ischemic stroke. Although the role of plaque-based tissue factor as stimulator of blood coagulation has been recognized, it is not known whether plaques can cause thrombus formation through direct activation of platelets. We isolated lipid-rich atheromatous plaques from 60 patients with carotid stenosis and identified morphologically diverse collagen type I- and type III-positive structures in the plaques that directly stimulated adhesion, dense granule secretion, and aggregation of platelets in buffer, plasma, and blood. This material also elicited platelet-monocyte aggregation and platelet-dependent blood coagulation. Plaques exposed to flowing blood at arterial wall shear rate induced platelets to adhere to and spread on the collagenous structures, triggering subsequent thrombus formation. Plaque-induced platelet thrombus formation was observed in fully anticoagulated blood (i.e., in the absence of tissue factor-mediated coagulation). Mice platelets lacking glycoprotein VI (GPVI) were unable to adhere to atheromatous plaque or form thrombi. Human platelet thrombus formation onto plaques in flowing blood was completely blocked by GPVI inhibition with the antibody 10B12 but not affected by integrin alpha2beta1 inhibition with 6F1 mAb. Moreover, the initial platelet response, shape change, induced by plaque was blocked by GPVI inhibition but not with alpha2beta1 antagonists (6F1 mAb or GFOGER-GPP peptide). Pretreatment of plaques with collagenase or anti-collagen type I and anti-collagen type III antibodies abolished plaque-induced platelet activation. Our results indicate that morphologically diverse collagen type I- and collagen type III-containing structures in lipid-rich atherosclerotic plaques stimulate thrombus formation by activating platelet GPVI. This platelet collagen receptor, essential for plaque-induced thrombus formation, presents a promising new anti-thrombotic target for the prevention of ischemic cardiovascular diseases.


Subject(s)
Atherosclerosis/complications , Platelet Membrane Glycoproteins/physiology , Thrombosis/etiology , Animals , Atherosclerosis/pathology , Blood Coagulation , Blood Platelets/physiology , Carotid Stenosis/blood , Collagen Type I/analysis , Collagen Type III/analysis , Humans , Integrin alpha2beta1/physiology , Lipids/analysis , Mice , Microscopy, Fluorescence , Monocytes/physiology , Platelet Activation , Platelet Adhesiveness , Platelet Aggregation , Platelet Membrane Glycoproteins/antagonists & inhibitors
16.
Sci Transl Med ; 8(367): 367ra168, 2016 11 30.
Article in English | MEDLINE | ID: mdl-27903864

ABSTRACT

In patients with atrial fibrillation, oral anticoagulation with oral thrombin inhibitors (OTIs), in contrast to vitamin K antagonists (VKAs), associates with a modest increase in acute coronary syndromes (ACSs). Whether this observation is causatively linked to OTI treatment and, if so, whether OTI action is the result of a lower antithrombotic efficacy of OTI compared to VKA or reflects a yet undefined prothrombotic activity of OTI remain unclear. We analyzed platelet function in patients receiving OTI or dose-adapted VKA under static and flow conditions. In vivo, we studied arterial thrombosis in OTI-, VKA-, and vehicle-treated mice using carotid ligation and wire injury models. Further, we examined thrombus formation on human atherosclerotic plaque homogenates under arterial shear to address the relevance to human pathology. Under static conditions, aggregation in the presence of ristocetin was increased in OTI-treated blood, whereas platelet reactivity and aggregation to other agonists were only marginally affected. Under flow conditions, firm platelet adhesion and thrombus formation on von Willebrand factor, collagen, and human atherosclerotic plaque were increased in the presence of OTI in comparison to VKA. OTI treatment was associated with increased thrombus formation in injured carotid arteries of mice. Inhibition or ablation of GPIbα-thrombin interactions abolished the effect of OTI on thrombus formation, suggesting a mechanistic role of the platelet receptor GPIbα and its thrombin-binding site. The effect of OTI was also abrogated in the presence of aspirin. In summary, OTI treatment has prothrombotic activity that might contribute to the increase in ACS observed clinically in patients.


Subject(s)
Platelet Adhesiveness/drug effects , Platelet Aggregation/drug effects , Thrombin/antagonists & inhibitors , Thrombosis/pathology , Acute Coronary Syndrome/pathology , Administration, Oral , Animals , Anticoagulants/pharmacology , Arteries/pathology , Aspirin/pharmacology , Atherosclerosis/pathology , Binding Sites , Blood Platelets/drug effects , Fibrinolytic Agents/pharmacology , Humans , Mice , Platelet Aggregation Inhibitors/pharmacology , Randomized Controlled Trials as Topic , Vitamin K/antagonists & inhibitors
17.
JACC Basic Transl Sci ; 1(3): 131-142., 2016 04.
Article in English | MEDLINE | ID: mdl-27766315

ABSTRACT

To enhance the antithrombotic properties of recombinant glycoprotein VI fragment crystallizable (GPVI-Fc), the authors incubated GPVI-Fc with anti-human Fc antibodies to cross-link the Fc tails of GPVI-Fc. Cross-linking potentiated the inhibition of human plaque- and collagen-induced platelet aggregation by GPVI-Fc under static and flow conditions without increasing bleeding time in vitro. Cross-linking with anti-human-Fc Fab2 was even superior to anti-human-Fc immunoglobulin G (IgG). Advanced optical imaging revealed a continuous sheath-like coverage of collagen fibers by cross-linked GPVI-Fc complexes. Cross-linking of GPVI into oligomeric complexes provides a new, highly effective, and probably safe antithrombotic treatment as it suppresses platelet GPVI-plaque interaction selectively at the site of acute atherothrombosis.

18.
Circulation ; 108(6): 741-7, 2003 Aug 12.
Article in English | MEDLINE | ID: mdl-12885756

ABSTRACT

BACKGROUND: Lysophosphatidic acid (LPA) is a platelet-activating component of mildly oxidized LDL (mox-LDL) and lipids isolated from human atherosclerotic plaques. Specific antagonists of platelet LPA receptors could be useful inhibitors of thrombus formation in patients with cardiovascular disease. METHODS AND RESULTS: Short-chain analogs of phosphatidic acid (PA) were examined for their effect on two initial platelet responses, platelet shape change and Ca2+ mobilization. Dioctylglycerol pyrophosphate [DGPP(8:0)] and dioctylphosphatidic acid [PA(8:0)], recently described selective antagonists of the LPA1 and LPA3 receptors, inhibited platelet activation evoked by LPA but not by other platelet stimuli. DGPP(8:0) was more potent than PA(8:0). DGPP(8:0) also inhibited platelet shape change induced by mox-LDL and lipid extracts from human atherosclerotic plaques. Notably, we demonstrate for the first time that the lipid-rich core isolated from soft plaques was able to directly induce shape change. This effect was completely abrogated by prior incubation of platelets with DGPP(8:0). Moreover, coapplication of the lipid-rich core or LPA together with subthreshold concentrations of ADP or epinephrine synergistically induced platelet aggregation; this effect was inhibited by DGPP(8:0). Analysis by liquid chromatography-mass spectrometry revealed the presence of LPA alkyl- and acyl-molecular species with high platelet-activating potency (16:0-alkyl-LPA, 20:4-acyl-LPA). CONCLUSIONS: LPA molecules present in the core region of atherosclerotic plaques trigger rapid platelet activation through the stimulation of LPA1 and LPA3 receptors. Antagonists of platelet LPA receptors might provide a new strategy to prevent thrombus formation in patients with cardiovascular diseases.


Subject(s)
Arteriosclerosis/physiopathology , Lipids/pharmacology , Organophosphorus Compounds/pharmacology , Phosphatidic Acids/pharmacology , Platelet Activation/drug effects , Receptors, Cell Surface/antagonists & inhibitors , Receptors, G-Protein-Coupled , Adenosine Diphosphate/antagonists & inhibitors , Adenosine Diphosphate/pharmacology , Arteriosclerosis/pathology , Biological Assay , Blood Platelets/cytology , Blood Platelets/drug effects , Blood Platelets/metabolism , Cell Separation , Cell Size/drug effects , Chromatography, Liquid , Dose-Response Relationship, Drug , Drug Synergism , Epinephrine/antagonists & inhibitors , Epinephrine/pharmacology , Humans , Lipids/isolation & purification , Lysophospholipids/antagonists & inhibitors , Lysophospholipids/pharmacology , Mass Spectrometry , Platelet Aggregation/drug effects , Receptors, Lysophosphatidic Acid
19.
Dtsch Med Wochenschr ; 140(8): 593-602, 2015 Apr.
Article in German | MEDLINE | ID: mdl-25945909

ABSTRACT

The diabetic foot syndrome (DFS) is a complication of diabetes mellitus, implying a serious impairment in quality of life for patients in advanced stages of the disease. Early detection of risks and stage-appropriate intervention are essential to increase the chances of foot salvage. The pathophysiological conditions for the formation of a DFS and treatment guidelines are currently underestimated. Up to 80 % of amputations are preventable if appropriate therapeutic steps were initiated on time in patients with DFS as part of a multidisciplinary approach. By proper prevention, the number of patients with DFS as well as the risk of recurrent ulcers can be reduced. This CME article gives an overview of the pathogenesis, diagnosis, therapy and prevention of DFS.


Subject(s)
Amputation, Surgical/methods , Anti-Bacterial Agents/therapeutic use , Compression Bandages , Debridement/methods , Diabetic Foot/diagnosis , Diabetic Foot/therapy , Exercise Therapy/methods , Aged , Combined Modality Therapy/methods , Diabetic Foot/etiology , Female , Humans , Syndrome
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL