Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Blood ; 143(20): 2089-2098, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38271661

ABSTRACT

ABSTRACT: von Willebrand factor (VWF) is an essential contributor to microvascular thrombosis. Physiological cleavage by ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13) limits its prothrombotic properties, explaining why ADAMTS13 deficiency leads to attacks of microthrombosis in patients with thrombotic thrombocytopenic purpura (TTP). We previously reported that plasminogen activation takes place during TTP attacks in these patients. Furthermore, stimulation of plasminogen activation attenuates pathogenesis in preclinical TTP models in vivo. This suggests that plasmin is an endogenous regulator of VWF thrombogenicity, in particular when ADAMTS13 falls short to prevent microvascular occlusions. VWF cleavage by plasmin is biochemically distinct from cleavage by ADAMTS13. We hypothesized that plasmin-cleaved VWF (cVWF) holds value as a biomarker of microvascular thrombosis. Here, we describe the development of a variable domain of heavy-chain-only antibody (VHH)-based bioassay that can distinguish cVWF from intact and ADAMTS13-cleaved VWF in plasma. We validate this assay by tracking cVWF release during degradation of microthombi in vitro. We demonstrate that endogenous cVWF formation takes place in patients with TTP during acute attacks of thrombotic microangiopathy but not in those in remission. Finally, we show that therapeutic plasminogen activation in a mouse model of TTP amplifies cVWF formation, which is accompanied by VWF clearance. Our combined findings indicate that cVWF is released from microthrombi in the context of microvascular occlusion.


Subject(s)
ADAMTS13 Protein , Biomarkers , Fibrinolysin , Purpura, Thrombotic Thrombocytopenic , von Willebrand Factor , von Willebrand Factor/metabolism , Humans , Biomarkers/blood , Biomarkers/metabolism , ADAMTS13 Protein/metabolism , ADAMTS13 Protein/blood , Animals , Mice , Fibrinolysin/metabolism , Purpura, Thrombotic Thrombocytopenic/metabolism , Purpura, Thrombotic Thrombocytopenic/blood , Purpura, Thrombotic Thrombocytopenic/diagnosis , Thrombosis/metabolism , Thrombosis/blood , Thrombosis/pathology , Thrombotic Microangiopathies/metabolism , Thrombotic Microangiopathies/blood , Female
2.
Int J Mol Sci ; 24(8)2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37108326

ABSTRACT

Procoagulant platelets are associated with an increased risk for thrombosis. Procoagulant platelet formation is mediated via Cyclophilin D (CypD) mediated opening of the mitochondrial permeability transition pore. Inhibiting CypD activity could therefore be an interesting approach to limiting thrombosis. In this study, we investigated the potential of two novel, non-immunosuppressive, non-peptidic small-molecule cyclophilin inhibitors (SMCypIs) to limit thrombosis in vitro, in comparison with the cyclophilin inhibitor and immunosuppressant Cyclosporin A (CsA). Both cyclophilin inhibitors significantly decreased procoagulant platelet formation upon dual-agonist stimulation, shown by a decreased phosphatidylserine (PS) exposure, as well as a reduction in the loss of mitochondrial membrane potential. Furthermore, the SMCypIs potently reduced procoagulant platelet-dependent clotting time, as well as fibrin formation under flow, comparable to CsA. No effect was observed on agonist-induced platelet activation measured by P-selectin expression, as well as CypA-mediated integrin αIIbß3 activation. Importantly, whereas CsA increased Adenosine 5'-diphosphate (ADP)-induced platelet aggregation, this was unaffected in the presence of the SMCypIs. We here demonstrate specific cyclophilin inhibition does not affect normal platelet function, while a clear reduction in procoagulant platelets is observed. Reducing platelet procoagulant activity by inhibiting cyclophilins with SMCypIs forms a promising strategy to limit thrombosis.


Subject(s)
Cyclophilins , Thrombosis , Mice , Animals , Humans , Cyclophilins/metabolism , Mice, Knockout , Blood Platelets/metabolism , Platelet Activation , Thrombosis/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
3.
J Photochem Photobiol B ; 234: 112500, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35816857

ABSTRACT

BACKGROUND AND AIM: A photosensitizer (PS) delivery and comprehensive tumor targeting platform was developed that is centered on the photosensitization of key pharmacological targets in solid tumors (cancer cells, tumor vascular endothelium, and cellular and non-cellular components of the tumor microenvironment) before photodynamic therapy (PDT). Interstitially targeted liposomes (ITLs) encapsulating zinc phthalocyanine (ZnPC) and aluminum phthalocyanine (AlPC) were formulated for passive targeting of the tumor microenvironment. In previous work it was established that the PEGylated ITLs were taken up by cultured cholangiocarcinoma cells. The aim of this study was to verify previous results in cancer cells and to determine whether the ITLs can also be used to photosensitize cells in the tumor microenvironment and vasculature. Following positive results, rudimentary in vitro and in vivo experiments were performed with ZnPC-ITLs and AlPC-ITLs as well as their water-soluble tetrasulfonated derivatives (ZnPCS4 and AlPCS4) to assemble a research dossier and bring this platform closer to clinical transition. METHODS: Flow cytometry and confocal microscopy were employed to determine ITL uptake and PS distribution in cholangiocarcinoma (SK-ChA-1) cells, endothelial cells (HUVECs), fibroblasts (NIH-3T3), and macrophages (RAW 264.7). Uptake of ITLs by endothelial cells was verified under flow conditions in a flow chamber. Dark toxicity and PDT efficacy were determined by cell viability assays, while the mode of cell death and cell cycle arrest were assayed by flow cytometry. In vivo systemic toxicity was assessed in zebrafish and chicken embryos, whereas skin phototoxicity was determined in BALB/c nude mice. A PDT efficacy pilot was conducted in BALB/c nude mice bearing human triple-negative breast cancer (MDA-MB-231) xenografts. RESULTS: The key findings were that (1) photodynamically active PSs (i.e., all except ZnPCS4) were able to effectively photosensitize cancer cells and non-cancerous cells; (2) following PDT, photodynamically active PSs were highly toxic-to-potent as per anti-cancer compound classification; (3) the photodynamically active PSs did not elicit notable systemic toxicity in zebrafish and chicken embryos; (4) ITL-delivered ZnPC and ZnPCS4 were associated with skin phototoxicity, while the aluminum-containing PSs did not exert detectable skin phototoxicity; and (5) ITL-delivered ZnPC and AlPC were equally effective in their tumor-killing capacity in human tumor breast cancer xenografts and superior to other non-phthalocyanine PSs when appraised on a per mole administered dose basis. CONCLUSIONS: AlPC(S4) are the safest and most effective PSs to integrate into the comprehensive tumor targeting and PS delivery platform. Pending further in vivo validation, these third-generation PSs may be used for multi-compartmental tumor photosensitization.


Subject(s)
Cholangiocarcinoma , Organometallic Compounds , Photochemotherapy , Animals , Cell Line, Tumor , Chick Embryo , Endothelial Cells , Humans , Liposomes , Mice , Mice, Nude , Organometallic Compounds/pharmacology , Organometallic Compounds/therapeutic use , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Tumor Microenvironment , Zebrafish
4.
J Thromb Haemost ; 20(5): 1213-1222, 2022 05.
Article in English | MEDLINE | ID: mdl-35170225

ABSTRACT

BACKGROUND: Thrombomodulin on endothelial cells can form a complex with thrombin. This complex has both anticoagulant properties, by activating protein C, and clot-protective properties, by activating thrombin-activatable fibrinolysis inhibitor (TAFI). Activated TAFI (TAFIa) inhibits plasmin-mediated fibrinolysis. OBJECTIVES: TAFIa inhibition is considered a potential antithrombotic strategy. So far, this goal has been pursued by developing compounds that directly inhibit TAFIa. In contrast, we here describe variable domain of heavy-chain-only antibody (VhH) clone 1 that inhibits TAFI activation by targeting human thrombomodulin. METHODS: Two llamas (Lama Glama) were immunized, and phage display was used to select VhH anti-thrombomodulin (TM) clone 1. Affinity was determined with surface plasmon resonance and binding to native TM was confirmed with flow cytometry. Clone 1 was functionally assessed by competition, clot lysis, and thrombin generation assays. Last, the effect of clone 1 on tPA-mediated fibrinolysis in human whole blood was investigated in a microfluidic fibrinolysis model. RESULTS: VhH anti-TM clone 1 bound recombinant TM with a binding affinity of 1.7 ± 0.4 nM and showed binding to native TM. Clone 1 competed with thrombin for binding to TM and attenuated TAFI activation in clot lysis assays and protein C activation in thrombin generation experiments. In a microfluidic fibrinolysis model, inhibition of TM with clone 1 fully prevented TAFI activation. DISCUSSION: We have developed VhH anti-TM clone 1, which inhibits TAFI activation and enhances tPA-mediated fibrinolysis under flow. Different from agents that directly target TAFIa, our strategy should preserve direct TAFI activation via thrombin.


Subject(s)
Carboxypeptidase B2 , Carboxypeptidase B2/metabolism , Clone Cells/metabolism , Endothelial Cells/metabolism , Fibrinolysis , Humans , Protein C/metabolism , Thrombin/metabolism , Thrombomodulin/chemistry
5.
Blood ; 139(4): 597-607, 2022 01 27.
Article in English | MEDLINE | ID: mdl-34752601

ABSTRACT

Thrombotic microangiopathies are hallmarked by attacks of disseminated microvascular thrombosis. In thrombotic thrombocytopenic purpura (TTP), this is caused by a rise in thrombogenic ultra-large von Willebrand factor (VWF) multimers because of ADAMTS13 deficiency. We previously reported that systemic plasminogen activation is therapeutic in a TTP mouse model. In contrast to its natural activators (ie, tissue plasminogen activator and urokinase plasminogen activator [uPA]), plasminogen can directly bind to VWF. For optimal efficacy and safety, we aimed to focus and accelerate plasminogen activation at sites of microvascular occlusion. We here describe the development and characterization of Microlyse, a fusion protein consisting of a high-affinity VHH targeting the CT/CK domain of VWF and the protease domain of uPA, for localized plasminogen activation on microthrombi. Microlyse triggers targeted destruction of platelet-VWF complexes by plasmin on activated endothelial cells and in agglutination studies. At equal molar concentrations, Microlyse degrades microthrombi sevenfold more rapidly than blockade of platelet-VWF interactions with a bivalent humanized VHH (caplacizumab*). Finally, Microlyse attenuates thrombocytopenia and tissue damage (reflected by increased plasma lactate dehydrogenase activity, as well as PAI-1 and fibrinogen levels) more efficiently than caplacizumab* in an ADAMTS13-/- mouse model of TTP, without affecting hemostasis in a tail-clip bleeding model. These findings show that targeted thrombolysis of VWF by Microlyse is an effective strategy for the treatment of TTP and might hold value for other forms of VWF-driven thrombotic disease.


Subject(s)
Fibrinolytic Agents/therapeutic use , Thrombotic Microangiopathies/drug therapy , von Willebrand Factor/metabolism , Animals , Female , Humans , Male , Mice, Inbred C57BL , Purpura, Thrombotic Thrombocytopenic/drug therapy , Purpura, Thrombotic Thrombocytopenic/metabolism , Recombinant Fusion Proteins/therapeutic use , Thrombotic Microangiopathies/metabolism
6.
J Thromb Haemost ; 20(2): 353-365, 2022 02.
Article in English | MEDLINE | ID: mdl-34653316

ABSTRACT

BACKGROUND: Occlusive thrombi are not homogeneous in composition. The core of a thrombus is rich in activated platelets and fibrin while the outer shell contains resting platelets. This core is inaccessible to plasma proteins. We produced a fusion protein (targeted SERPIN-TaSER), consisting of a function-blocking VH H against glycoprotein Ibα (GPIbα) and a thrombin-inhibiting serine protease inhibitor (SERPIN; α1-antitrypsin 355 AIAR358 ) to interfere with platelet-driven thrombin formation. AIM: To evaluate the antithrombotic properties of TaSER. METHODS: Besides TaSER, we generated three analogous control variants with either a wild-type antitrypsin subunit, a non-targeting control VH H, or their combination. We investigated TaSER and controls in protease activity assays, (platelet-dependent) thrombin generation assays, and by western blotting. The effects of TaSER on platelet activation and von Willebrand factor (VWF) binding were studied by fluorescence-activated cell sorting, in agglutination studies, and in ATP secretion experiments. We studied the influence of TaSER in whole blood (1) on platelet adhesion on VWF, (2) aggregate formation on collagen, and (3) thrombus formation (after recalcification) on collagen and tissue factor. RESULTS: TaSER binds platelets and inhibits thrombin activity on the platelet surface. It blocks VWF binding and disassembles platelet agglutinates. TaSER delays tissue factor-triggered thrombin generation and ATP secretion in platelet-rich plasma in a targeted manner. In flow studies, TaSER interferes with platelet adhesion and aggregate formation due to GPIbα blockade and limits thrombus formation due to targeted inhibition of platelet-dependent thrombin activity. CONCLUSION: The synergy between the individual properties of TaSER makes it a highly effective antithrombotic agent with possible clinical implications.


Subject(s)
Fibrinolytic Agents , Serpins , Blood Platelets/metabolism , Fibrinolytic Agents/pharmacology , Humans , Platelet Adhesiveness , Serpins/metabolism , von Willebrand Factor/metabolism
7.
Front Med (Lausanne) ; 8: 650129, 2021.
Article in English | MEDLINE | ID: mdl-33968958

ABSTRACT

Background: Coronavirus disease of 2019 (COVID-19) is associated with a prothrombotic state and a high incidence of thrombotic event(s) (TE). Objectives: To study platelet reactivity in hospitalized COVID-19 patients and determine a possible association with the clinical outcomes thrombosis and all-cause mortality. Methods: Seventy nine hospitalized COVID-19 patients were enrolled in this retrospective cohort study and provided blood samples in which platelet reactivity in response to stimulation with ADP and TRAP-6 was determined using flow cytometry. Clinical outcomes included thrombotic events, and all-cause mortality. Results: The incidence of TE in this study was 28% and all-cause mortality 16%. Patients that developed a TE were younger than patients that did not develop a TE [median age of 55 vs. 70 years; adjusted odds ratio (AOR) = 0.96 per 1 year of age, 95% confidence interval (CI) 0.92-1.00; p = 0.041]. Furthermore, patients using preexisting thromboprophylaxis were less likely to develop a thrombotic complication than patients that were not (18 vs. 54%; AOR = 0.19, 95% CI 0.04-0.84; p = 0.029). Conversely, having asthma strongly increased the risk on TE development (AOR = 6.2, 95% CI 1.15-33.7; p = 0.034). No significant differences in baseline P-selectin expression or platelet reactivity were observed between the COVID-19 positive patients (n = 79) and COVID-19 negative hospitalized control patients (n = 21), nor between COVID-19 positive survivors or non-survivors. However, patients showed decreased platelet reactivity in response to TRAP-6 following TE development. Conclusion: We observed an association between the use of preexisting thromboprophylaxis and a decreased risk of TE during COVID-19. This suggests that these therapies are beneficial for coping with COVID-19 associated hypercoagulability. This highlights the importance of patient therapy adherence. We observed lowered platelet reactivity after the development of TE, which might be attributed to platelet desensitization during thromboinflammation.

8.
Int J Mol Sci ; 21(17)2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32824972

ABSTRACT

Extracellular vesicles (EV) secreted in the tumour microenvironment (TME) are emerging as major antagonists of anticancer therapies by orchestrating the therapeutic outcome through altering the behaviour of recipient cells. Recent evidence suggested that chemotherapeutic drugs could be responsible for the EV-mediated tumour-stroma crosstalk associated with cancer cell drug resistance. Here, we investigated the capacity of tumour EV (TEV) secreted by normoxic and hypoxic (1% oxygen) C26 cancer cells after doxorubicin (DOX) treatment to alter the response of naïve C26 cells and RAW 264.7 macrophages to DOX. We observed that C26 cells were less responsive to DOX treatment under normoxia compared to hypoxia, and a minimally cytotoxic DOX concentration that mounted distinct effects on cell viability was selected for TEV harvesting. Homotypic and heterotypic pretreatment of naïve hypoxic cancer and macrophage-like cells with normoxic DOX-elicited TEV rendered these cells slightly less responsive to DOX treatment. The observed effects were associated with strong hypoxia-inducible factor 1-alpha (HIF-1α) induction and B-cell lymphoma-extra-large anti-apoptotic protein (Bcl-xL)-mediated anti-apoptotic response in normoxic DOX-treated TEV donor cells, being also tightly connected to the DOX-TEV-mediated HIF-1α induction, as well as Bcl-xL levels increasing in recipient cells. Altogether, our results could open new perspectives for investigating the role of chemotherapy-elicited TEV in the colorectal cancer TME and their modulatory actions on promoting drug resistance.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Colonic Neoplasms/metabolism , Doxorubicin/toxicity , Drug Resistance, Neoplasm , Extracellular Vesicles/metabolism , Tumor Hypoxia , Animals , Cell Line, Tumor , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , RAW 264.7 Cells , Stromal Cells/drug effects , Stromal Cells/metabolism , bcl-X Protein/metabolism
9.
Nanomaterials (Basel) ; 10(7)2020 Jul 10.
Article in English | MEDLINE | ID: mdl-32664364

ABSTRACT

Since αvß3 integrin is a key component of angiogenesis in health and disease, Arg-Gly-Asp (RGD) peptide-functionalized nanocarriers have been investigated as vehicles for targeted delivery of drugs to the αvß3 integrin-overexpressing neovasculature of tumors. In this work, PEGylated nanoparticles (NPs) based on poly(lactic-co-glycolic acid) (PLGA) functionalized with cyclic-RGD (cRGD), were evaluated as nanocarriers for the targeting of angiogenic endothelium. For this purpose, NPs (~300 nm) functionalized with cRGD with different surface densities were prepared by maleimide-thiol chemistry and their interactions with human umbilical vein endothelial cells (HUVECs) were evaluated under different conditions using flow cytometry and microscopy. The cell association of cRGD-NPs under static conditions was time-, concentration- and cRGD density-dependent. The interactions between HUVECs and cRGD-NPs dispersed in cell culture medium under flow conditions were also time- and cRGD density-dependent. When washed red blood cells (RBCs) were added to the medium, a 3 to 8-fold increase in NPs association to HUVECs was observed. Moreover, experiments conducted under flow in the presence of RBC at physiologic hematocrit and shear rate, are a step forward in the prediction of in vivo cell-particle association. This approach has the potential to assist development and high-throughput screening of new endothelium-targeted nanocarriers.

10.
Blood Adv ; 4(13): 2967-2978, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32609845

ABSTRACT

Thrombocytopenia is a common complication of influenza virus infection, and its severity predicts the clinical outcome of critically ill patients. The underlying cause(s) remain incompletely understood. In this study, in patients with an influenza A/H1N1 virus infection, viral load and platelet count correlated inversely during the acute infection phase. We confirmed this finding in a ferret model of influenza virus infection. In these animals, platelet count decreased with the degree of virus pathogenicity varying from 0% in animals infected with the influenza A/H3N2 virus, to 22% in those with the pandemic influenza A/H1N1 virus, up to 62% in animals with a highly pathogenic A/H5N1 virus infection. This thrombocytopenia is associated with virus-containing platelets that circulate in the blood. Uptake of influenza virus particles by platelets requires binding to sialoglycans and results in the removal of sialic acids by the virus neuraminidase, a trigger for hepatic clearance of platelets. We propose the clearance of influenza virus by platelets as a paradigm. These insights clarify the pathophysiology of influenza virus infection and show how severe respiratory infections, including COVID-19, may propagate thrombocytopenia and/or thromboembolic complications.


Subject(s)
Blood Platelets/virology , Influenza A virus/pathogenicity , Influenza, Human/complications , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Thrombocytopenia/etiology , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Disease Models, Animal , Ferrets , Host-Pathogen Interactions , Humans , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H3N2 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/physiology , Influenza A virus/physiology , Influenza, Human/metabolism , Influenza, Human/pathology , Influenza, Human/virology , Orthomyxoviridae Infections/complications , Orthomyxoviridae Infections/metabolism , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Thrombocytopenia/metabolism , Thrombocytopenia/pathology , Thrombocytopenia/virology , Virus Internalization
11.
Sci Rep ; 9(1): 18316, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31797980

ABSTRACT

Heparin is a widely used anticoagulant which inhibits factor Xa and thrombin through potentiation of antithrombin. We recently identified that the nucleic acid stain SYTOX reacts with platelet polyphosphate due to molecular similarities, some of which are shared by heparin. We attempted to study heparin in flowing blood by live-cell fluorescence microscopy, using SYTOX for heparin visualisation. Immunostaining was performed with monoclonal antibodies directed against various heparin-binding proteins. In addition, we studied modulation of heparin activity in coagulation assays, as well its effects on fibrin formation under flow in recalcified whole blood. We found that SYTOX-positive polymers appear in heparinised blood under flow. These polymers typically associate with platelet aggregates and their length (reversibly) increases with shear rate. Immunostaining revealed that of the heparin-binding proteins assessed, they only contain histones. In coagulation assays and flow studies on fibrin formation, we found that addition of exogenous histones reverses the anticoagulant effects of heparin. Furthermore, the polymers do not appear in the presence of DNase I, heparinase I/III, or the heparin antidote protamine. These findings suggest that heparin forms polymeric complexes with cell-free DNA in whole blood through a currently unidentified mechanism.


Subject(s)
Anticoagulants/blood , Cell-Free Nucleic Acids/blood , Heparin/blood , Histones/blood , Blood Platelets/metabolism , Humans , Organic Chemicals , Polymers/metabolism , Shear Strength
12.
Angiogenesis ; 21(2): 325-334, 2018 05.
Article in English | MEDLINE | ID: mdl-29532289

ABSTRACT

INTRODUCTION: At the clinical introduction of antiangiogenic agents as anticancer agents, no major toxicities were expected as merely just endothelial cells (ECs) in tumors would be affected. However, several (serious) toxicities became apparent, of which underlying mechanisms are largely unknown. We investigated to what extent sunitinib (multitargeted antiangiogenic tyrosine kinase inhibitor (TKI)), sorafenib (TKI) and bevacizumab [specific antibody against vascular endothelial growth factor (VEGF)] may impair platelet function, which might explain treatment-related bleedings. MATERIALS AND METHODS: In vitro, the influence of sunitinib, sorafenib, and bevacizumab on platelet aggregation, P-selectin expression and fibrinogen binding, platelet-EC interaction, and tyrosine phosphorylation of c-Src was studied by optical aggregation, flow cytometry, real-time perfusion, and western blotting. Ex vivo, platelet aggregation was analyzed in 25 patients upon sunitinib or bevacizumab treatment. Concentrations of sunitinib, VEGF, and platelet and EC activation markers were measured by LC-MS/MS and ELISA. RESULTS: In vitro, sunitinib and sorafenib significantly inhibited platelet aggregation (20 µM sunitinib: 71.3%, p < 0.001; 25 µM sorafenib: 55.8%, p = 0.042). Sorafenib and sunitinib significantly inhibited P-selectin expression on platelets. Exposure to both TKIs resulted in a reduced tyrosine phosphorylation of c-Src. Ex vivo, within 24 h sunitinib impaired platelet aggregation (83.0%, p = 0.001, N = 8). Plasma concentrations of sunitinib, VEGF, and platelet/EC activation markers were not correlated with disturbed aggregation. In contrast, bevacizumab only significantly impaired platelet aggregation in vitro at high concentrations, but not ex vivo. CONCLUSION: Sunitinib significantly inhibits platelet aggregation in patients already after 24 h of first administration, whereas bevacizumab had no effect on aggregation. These findings may explain the clinically observed bleedings during treatment with antiangiogenic TKIs.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Bevacizumab/pharmacology , Blood Platelets/metabolism , Platelet Aggregation/drug effects , Sorafenib/pharmacology , Sunitinib/pharmacology , CSK Tyrosine-Protein Kinase , Endothelial Cells/metabolism , Female , Humans , Male , P-Selectin/metabolism , src-Family Kinases/metabolism
13.
Eur J Pharmacol ; 816: 129-137, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-28899695

ABSTRACT

Atherosclerosis is the major underlying pathology of cardiovascular disease (CVD). The risk for CVD is increased in women with a history of preeclampsia. Multiple studies have indicated that accelerated atherosclerosis underlies this increased CVD risk. Furthermore, it has been suggested that endothelial dysfunction and inflammation play an important role in the increased CVD risk of women with preeclampsia. Rupture or erosion of atherosclerotic plaques can induce the formation of thrombi that underlie the onset of acute clinical CVD such as myocardial infarction and stroke. In relatively young women, cardiovascular events are mainly due to plaque erosions. Eroded plaques have a distinct morphology compared to ruptured plaques, but have been understudied as a substrate for CVD. The currently available evidence points towards lesions with features of stability such as high collagen content and smooth muscle cells and with distinct mechanisms that further promote the pro-thrombotic environment such as Toll Like Receptor (TLR) signaling and endothelial apoptosis. These suggested mechanisms, that point to endothelial dysfunction and intimal thickening, may also play a role in preeclampsia. Pregnancy is considered a stress test for the cardiovascular system with preeclampsia as an additional pathological substrate for earlier manifestation of vascular disease. This review provides a summary of the possible common mechanisms involved in preeclampsia and accelerated atherosclerosis in young females and highlights plaque erosion as a likely substrate for CVD events in women with a history of preeclampsia.


Subject(s)
Coronary Artery Disease/complications , Endothelium, Vascular/pathology , Plaque, Atherosclerotic/complications , Pre-Eclampsia/pathology , Female , Humans , Pregnancy , Risk
14.
J Vis Exp ; (125)2017 07 10.
Article in English | MEDLINE | ID: mdl-28715386

ABSTRACT

Blood platelets are essential players in hemostasis, the formation of thrombi to seal vascular breaches. They are also involved in thrombosis, the formation of thrombi that occlude the vasculature and injure organs, with life-threatening consequences. This motivates scientific research on platelet function and the development of methods to track cell-biological processes as they occur under flow conditions. A variety of flow models are available for the study of platelet adhesion and aggregation, two key phenomena in platelet biology. This work describes a method to study real-time platelet degranulation under flow during activation. The method makes use of a flow chamber coupled to a syringe-pump setup that is placed under a wide-field, inverted, LED-based fluorescence microscope. The setup described here allows for the simultaneous excitation of multiple fluorophores that are delivered by fluorescently labeled antibodies or fluorescent dyes. After live-cell imaging experiments, the cover glasses can be further processed and analyzed using static microscopy (i.e., confocal microscopy or scanning electron microscopy).


Subject(s)
Blood Platelets/metabolism , Hemostasis/physiology , Microscopy, Fluorescence/methods , Platelet Adhesiveness/physiology , Blood Platelets/cytology , Humans , Thrombosis/blood
15.
Blood ; 129(12): 1707-1717, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28049643

ABSTRACT

Polyphosphate is an inorganic polymer that can potentiate several interactions in the blood coagulation system. Blood platelets contain polyphosphate, and the secretion of platelet-derived polyphosphate has been associated with increased thrombus formation and activation of coagulation factor XII. However, the small polymer size of secreted platelet polyphosphate limits its capacity to activate factor XII in vitro. Thus, the mechanism by which platelet polyphosphate contributes to thrombus formation remains unclear. Using live-cell imaging, confocal and electron microscopy, we show that activated platelets retain polyphosphate on their cell surface. The apparent polymer size of membrane-associated polyphosphate largely exceeds that of secreted polyphosphate. Ultracentrifugation fractionation experiments revealed that membrane-associated platelet polyphosphate is condensed into insoluble spherical nanoparticles with divalent metal ions. In contrast to soluble polyphosphate, membrane-associated polyphosphate nanoparticles potently activate factor XII. Our findings identify membrane-associated polyphosphate in a nanoparticle state on the surface of activated platelets. We propose that these polyphosphate nanoparticles mechanistically link the procoagulant activity of platelets with the activation of coagulation factor XII.


Subject(s)
Blood Coagulation/drug effects , Blood Platelets/metabolism , Polyphosphates/metabolism , Blood Platelets/chemistry , Blood Platelets/ultrastructure , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Factor XII/metabolism , Humans , Nanoparticles/chemistry , Polyphosphates/pharmacology
16.
Methods Mol Biol ; 1545: 205-213, 2017.
Article in English | MEDLINE | ID: mdl-27943217

ABSTRACT

In the last few years it has become clear that, in addition to soluble molecules such as growth factors and cytokines, cells use extracellular vesicles (EVs) for intercellular communication. For example, EVs derived from cancer cells interact with endothelial cells, thereby affecting angiogenesis and metastasis, two essential processes in tumor progression. In most experiments, the interaction of EVs with target cells is investigated under static conditions. However the use of dynamic flow conditions is considered more relevant, especially when studying EV uptake by endothelial cells. Here, we describe the use of a perfusion system to investigate the interaction of (tumor) EVs with endothelial cells under dynamic flow conditions.


Subject(s)
Cell Communication , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Cell Fractionation/methods , Cell Line , Culture Media, Conditioned , Humans , Microfluidics/instrumentation , Microfluidics/methods
17.
Innovations (Phila) ; 10(3): 195-201, 2015.
Article in English | MEDLINE | ID: mdl-26181583

ABSTRACT

OBJECTIVE: Heart valve and vascular prosthesis implantation is a common procedure for patients with heart valve stenosis or regurgitation and dilated or obstructive vascular disease. Drawbacks of conventional valve prostheses are the requirement for anticoagulant drugs, moderate durability, and suboptimal resistance to fatigue and tear. Dyneema Purity fibers are made from ultra-high-molecular-weight polyethylene filaments and are very thin, flexible, and fatigue and abrasion resistant and have high strength. Therefore, prostheses made from Dyneema Purity fibers might be attractive for use in the minimally invasive treatment of valvular- and vascular diseases. The aim of this study was to test the hemocompatibility of Dyneema Purity fibers in contact with blood. METHODS: Real-time platelet adhesion in human blood of 3 volunteers was quantified after 5 minutes of perfusion on single filaments (Ø 15 µm) of Dyneema Purity and polyester fibers. Plasma thrombin generation was measured by fluoroscopy for patches of Dyneema Purity fibers and for 5 commonly used polyester and expanded polytetrafluoroethylene cardiovascular prostheses. RESULTS: Platelet adhesion per 1 mm was 6 ± 1.4 on Dyneema Purity filaments and 15 ± 3.4 on polyester filaments (P = 0.02). Total formed thrombin and the time to peak of its maximum were noninferior for patches of Dyneema Purity fibers compared with the reference materials. CONCLUSIONS: Dyneema Purity fibers are noninferior in adhesion and coagulation activation compared with commonly used cardiovascular prostheses.


Subject(s)
Blood Vessel Prosthesis , Heart Valve Prosthesis , Materials Testing/methods , Blood Physiological Phenomena , Blood Platelets/drug effects , Blood Vessel Prosthesis Implantation/methods , Fluoroscopy/methods , Heart Valves/pathology , Heart Valves/surgery , Humans , In Vitro Techniques/methods , Platelet Adhesiveness/drug effects , Polyethylenes , Polytetrafluoroethylene , Vascular Diseases/pathology , Vascular Diseases/surgery
19.
Cardiovasc Res ; 103(1): 140-6, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24776597

ABSTRACT

AIMS: Platelets are a natural source of growth factors, cytokines and chemokines, that regulate angiogenesis and inflammation. It has been suggested that differential release of pro- and anti-angiogenic growth factors from platelet α-granules by protease-activated receptors (PAR) 1 and 4 may be important for the regulation of angiogenesis. We aimed to compare the releasates of unstimulated platelets with PAR-1- and PAR-4-stimulated platelets. METHODS AND RESULTS: The release of ß-thromboglobulin, platelet factor (PF)-4, thrombospondin, platelet-derived growth factor (PDGF)-A/B, regulated and normal T-cell expressed and secreted (RANTES/CCL5), endostatin, CXCL12, and vascular endothelial growth factor (VEGF) was measured with enzyme-linked immunosorbent assay (ELISA). Mass spectrometry (MS)-based quantitative proteomics identified 93 proteins from platelets stimulated with PAR-1 and PAR-4. A strong correlation between the factors released after either stimulus was observed (Spearman's r 0.94, P < 0.001). Analysis with ELISA showed that stimulation with PAR-1 or PAR-4 lead to non-differential release of ß-thromboglobulin, PF-4, thrombospondin, PDGF-A/B, RANTES/CCL5, endostatin, CXCL12, and VEGF. Release of thrombospondin was slightly lower after PAR-1 stimulation (7.2 µg/mL), compared with PAR-4 induced release (9.8 µg/mL; P < 0.05). CONCLUSIONS: Both ELISA on established α-granule proteins and MS-based quantitative proteomics showed that the most abundant α-granule proteins are released in similar quantities from platelets after stimulation with either PAR-1 or PAR-4. Our findings provide evidence against the hypothesis that PAR-1 and PAR-4 stimulation of platelets trigger differential release of alpha-granule, but further studies are needed to draw conclusions for physiological conditions.


Subject(s)
Apoptosis Regulatory Proteins/blood , Blood Platelets/physiology , Receptor, PAR-1/blood , Chemokine CXCL12/blood , Enzyme-Linked Immunosorbent Assay , Fibrinogen/metabolism , Humans , Mass Spectrometry , Neovascularization, Physiologic , P-Selectin/blood , Platelet Activation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Proteomics , Secretory Vesicles/physiology , Vascular Endothelial Growth Factor A/blood , beta-Thromboglobulin/metabolism
20.
Circulation ; 129(12): 1320-31, 2014 Mar 25.
Article in English | MEDLINE | ID: mdl-24449821

ABSTRACT

BACKGROUND: Von Willebrand factor (VWF) multimer size is controlled through continuous proteolysis by ADAMTS13 (a disintegrin and metalloproteinase with a thrombospondin type I motif, member 13). This prevents spontaneous platelet agglutination and microvascular obstructions. ADAMTS13 deficiency is associated with thrombotic thrombocytopenic purpura, in which life-threatening episodes of microangiopathy damage kidneys, heart, and brain. Enigmatically, a complete ADAMTS13 deficiency does not lead to continuous microangiopathy. We hypothesized that plasmin, the key enzyme of the fibrinolytic system, serves as a physiological backup enzyme for ADAMTS13 in the degradation of pathological platelet-VWF complexes. METHODS AND RESULTS: Using real-time microscopy, we determined that plasmin rapidly degrades platelet-VWF complexes on endothelial cells in absence of ADAMTS13, after activation by urokinase-type plasminogen activator or the thrombolytic agent streptokinase. Similarly, plasmin degrades platelet-VWF complexes in platelet agglutination studies. Plasminogen directly binds to VWF and its A1 domain in a lysine-dependent manner, as determined by enzyme-linked immunosorbent assay. Plasma levels of plasmin-α2-antiplasmin complexes increase with the extent of thrombocytopenia in patients with acute episodes of thrombotic thrombocytopenic purpura, independent of ADAMTS13 activity. This indicates that plasminogen activation takes place during microangiopathy. Finally, we show that the thrombolytic agent streptokinase has therapeutic value for Adamts13(-/-) mice in a model of thrombotic thrombocytopenic purpura. CONCLUSIONS: We propose that plasminogen activation on endothelial cells acts as a natural backup for ADAMTS13 to degrade obstructive platelet-VWF complexes. Our findings indicate that thrombolytic agents may have therapeutic value in the treatment of microangiopathies and may be useful to bypass inhibitory antibodies against ADAMTS13 that cause thrombotic thrombocytopenic purpura.


Subject(s)
ADAM Proteins/metabolism , Fibrinolysin/metabolism , Fibrinolysis/physiology , Thrombotic Microangiopathies/metabolism , von Willebrand Factor/metabolism , ADAM Proteins/genetics , ADAM Proteins/immunology , ADAMTS13 Protein , Animals , Autoantibodies/metabolism , Blood Platelets/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Weight , Plasminogen/metabolism , Purpura, Thrombotic Thrombocytopenic/metabolism , Streptokinase/metabolism , Streptokinase/pharmacology , Thrombotic Microangiopathies/drug therapy , von Willebrand Factor/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...