Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Thromb Haemost ; 120(6): 977-993, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32492725

RESUMO

Glycoprotein VI (GPVI), a platelet collagen receptor, is crucial in mediating atherothrombosis. Besides collagen, injured plaques expose tissue factor (TF) that triggers fibrin formation. Previous studies reported that GPVI also is a platelet receptor for fibrinogen and fibrin. We studied the effect of anti-GPVI antibodies and inhibitors of GPVI signaling kinases (Syk and Btk) on platelet adhesion and aggregate formation onto immobilized fibrinogen and different types of fibrin under arterial flow conditions. Fibrin was prepared from isolated fibrinogen ("pure fibrin"), recombinant fibrinogen ("recombinant fibrin"), or generated more physiologically from endogenous fibrinogen in plasma ("plasma fibrin") or by exposing TF-coated surfaces to flowing blood ("blood fibrin"). Inhibition of GPVI and Syk did not inhibit platelet adhesion and aggregate formation onto fibrinogen. In contrast anti-GPVI antibodies, inhibitors of Syk and Btk and the anti-GPIb antibody 6B4 inhibited platelet aggregate formation onto pure and recombinant fibrin. However, inhibition of GPVI and GPVI signaling did not significantly reduce platelet coverage of plasma fibrin and blood fibrin. Plasma fibrin contained many proteins incorporated during clot formation. Advanced optical imaging revealed plasma fibrin as a spongiform cushion with thicker, knotty, and long fibers and little activation of adhering platelets. Albumin intercalated in plasma fibrin fibers left only little space for platelet attachment. Pure fibrin was different showing a dense mesh of thin fibers with strongly activated platelets. We conclude that fibrin formed in plasma and blood contains plasma proteins shielding GPVI-activating epitopes. Our findings do not support a role of GPVI for platelet activation by physiologic fibrin.


Assuntos
Plaquetas/metabolismo , Fibrina/metabolismo , Glicoproteínas da Membrana de Plaquetas/fisiologia , Receptores de Peptídeos/metabolismo , Tirosina Quinase da Agamaglobulinemia/sangue , Tirosina Quinase da Agamaglobulinemia/fisiologia , Ativação Enzimática , Fibrinogênio/metabolismo , Hemorreologia , Humanos , Microscopia Confocal/métodos , Plasma , Adesividade Plaquetária , Agregação Plaquetária , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Glicoproteínas da Membrana de Plaquetas/imunologia , Ligação Proteica , Proteínas Recombinantes/metabolismo , Quinase Syk/antagonistas & inibidores , Quinase Syk/sangue , Quinase Syk/fisiologia , Tromboplastina/metabolismo
2.
Thromb Haemost ; 119(3): 397-406, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30685871

RESUMO

Ibrutinib and acalabrutinib are approved for B cell malignancies and novel Bruton's tyrosine kinase (Btk) inhibitors undergo clinical testing also in B cell-driven autoimmune disorders. Btk in platelets mediates platelet activation via glycoprotein (GP) VI, which is crucial for atherosclerotic plaque-induced platelet thrombus formation. This can be selectively inhibited by Btk inhibitors. Since patients on second-generation Btk inhibitors apparently show less bleeding than patients on ibrutinib, we compared the effects of ibrutinib and four novel irreversible Btk inhibitors on GPVI-dependent platelet aggregation in blood and in vitro bleeding time. Low concentrations of collagen which induced the same low degree of GPVI-mediated platelet aggregation as atherosclerotic plaque material were applied. IC50 values for collagen (0.2-0.5 µg/mL)-induced platelet aggregation after 15-minute pre-incubation were: ibrutinib 0.12 µM, BGB-3111 0.51 µM, acalabrutinib 1.21 µM, ONO/GS-4059 1.20 µM and evobrutinib 5.84 µM. Peak venous plasma concentrations of ibrutinib (0.5 µM), acalabrutinib (2 µM) and ONO/GS-4059 (2 µM) measured after anti-proliferative dosage inhibited collagen-induced platelet aggregation, but did not increase PFA-200 closure time on collagen/epinephrine. Closure times were moderately increased by 2- to 2.5-fold higher concentrations of these inhibitors, but not by BGB-3111 (1 µM) and evobrutinib (10 µM). Prolonging platelet drug exposure to 60 minutes lowered IC50 values of any Btk inhibitor for GPVI-mediated aggregation by several fold, and 5- to 10-fold below anti-proliferative therapeutic drug plasma levels. In conclusion, low blood concentrations of ibrutinib and the novel Btk inhibitors suffice for GPVI selective platelet inhibition relevant for atherothrombosis but do not impair primary haemostasis.


Assuntos
Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Benzamidas/farmacologia , Plaquetas/efeitos dos fármacos , Imidazóis/farmacologia , Piperidinas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Pirazinas/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Adenina/análogos & derivados , Tirosina Quinase da Agamaglobulinemia/sangue , Benzamidas/toxicidade , Plaquetas/metabolismo , Relação Dose-Resposta a Droga , Hemorragia/sangue , Hemorragia/induzido quimicamente , Hemostasia/efeitos dos fármacos , Humanos , Imidazóis/toxicidade , Concentração Inibidora 50 , Piperidinas/toxicidade , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/toxicidade , Glicoproteínas da Membrana de Plaquetas/metabolismo , Pirazinas/toxicidade , Pirazóis/toxicidade , Pirimidinas/toxicidade
3.
Blood ; 131(24): 2605-2616, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29559479

RESUMO

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.


Assuntos
Tirosina Quinase da Agamaglobulinemia/antagonistas & inibidores , Benzamidas/uso terapêutico , Imidazóis/uso terapêutico , Placa Aterosclerótica/complicações , Inibidores da Agregação Plaquetária/uso terapêutico , Pirazinas/uso terapêutico , Pirazóis/uso terapêutico , Pirimidinas/uso terapêutico , Trombose/etiologia , Trombose/prevenção & controle , Adenina/análogos & derivados , Administração Oral , Adulto , Tirosina Quinase da Agamaglobulinemia/metabolismo , Idoso , Benzamidas/administração & dosagem , Humanos , Imidazóis/administração & dosagem , Masculino , Pessoa de Meia-Idade , Piperidinas , Placa Aterosclerótica/tratamento farmacológico , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/uso terapêutico , Pirazinas/administração & dosagem , Pirazóis/administração & dosagem , Pirimidinas/administração & dosagem , Trombose/metabolismo , Trombose/patologia
4.
Thromb Haemost ; 118(2): 351-361, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29378359

RESUMO

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.


Assuntos
Colágeno/metabolismo , Fibrina/metabolismo , Fibrinogênio/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Trombina/metabolismo , Aterosclerose/metabolismo , Coagulação Sanguínea , Plaquetas/metabolismo , Humanos , Microscopia de Fluorescência , Placa Aterosclerótica/metabolismo , Ativação Plaquetária , Adesividade Plaquetária , Agregação Plaquetária , Ligação Proteica , Multimerização Proteica , Proteínas Recombinantes
5.
J Am Heart Assoc ; 6(8)2017 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-28751543

RESUMO

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.


Assuntos
Antígenos CD/farmacologia , Apirase/farmacologia , Lesões das Artérias Carótidas/tratamento farmacológico , Fibrinolíticos/farmacologia , Glicoproteínas/farmacologia , Fragmentos Fc das Imunoglobulinas/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/farmacologia , Trombose/prevenção & controle , Animais , Antígenos CD/toxicidade , Apirase/farmacocinética , Apirase/toxicidade , Doenças das Artérias Carótidas/sangue , Doenças das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/induzido quimicamente , Lesões das Artérias Carótidas/patologia , Cloretos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Compostos Férricos , Fibrinolíticos/farmacocinética , Fibrinolíticos/toxicidade , Glicoproteínas/farmacocinética , Glicoproteínas/toxicidade , Hemorragia/induzido quimicamente , Humanos , Fragmentos Fc das Imunoglobulinas/toxicidade , Masculino , Camundongos Endogâmicos C57BL , Placa Aterosclerótica , Inibidores da Agregação Plaquetária/farmacocinética , Inibidores da Agregação Plaquetária/toxicidade , Glicoproteínas da Membrana de Plaquetas/farmacocinética , Glicoproteínas da Membrana de Plaquetas/toxicidade , Proteínas Recombinantes de Fusão/farmacologia , Trombose/sangue , Trombose/induzido quimicamente , Trombose/patologia
6.
Thromb Haemost ; 117(8): 1651-1659, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28569920

RESUMO

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.


Assuntos
Adenosina/análogos & derivados , Anticorpos Monoclonais/farmacologia , Aspirina/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Glicoproteínas/farmacologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Fragmentos Fc das Imunoglobulinas/farmacologia , Placa Aterosclerótica , Inibidores da Agregação Plaquetária/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Trombose/prevenção & controle , Abciximab , Adenosina/farmacologia , Adenosina/toxicidade , Anticorpos Monoclonais/toxicidade , Aspirina/toxicidade , Plaquetas/metabolismo , Quimioterapia Combinada , Glicoproteínas/toxicidade , Hemorragia/induzido quimicamente , Humanos , Fragmentos Fab das Imunoglobulinas/toxicidade , Fragmentos Fc das Imunoglobulinas/toxicidade , Adesividade Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/toxicidade , Antagonistas do Receptor Purinérgico P2Y/toxicidade , Trombose/sangue , Trombose/patologia , Ticagrelor , Fatores de Tempo
7.
Thromb Haemost ; 117(7): 1249-1257, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28597906

RESUMO

Anti-platelet drugs reduce arterial thrombosis after plaque rupture and erosion, prevent stent thrombosis and are used to prevent and treat myocardial infarction and ischaemic stroke. Some of them may also be helpful in treating less frequent diseases such as thrombotic thrombocytopenic purpura. The present concise review aims to cover current and future developments of anti-platelet drugs interfering with the interaction of von Willebrand factor (VWF) with glycoprotein (GP) Ibα, and directed against GPVI, GPIIb/IIIa (integrin αIIbß3), the thrombin receptor PAR-1, and the ADP receptor P2Y12. The high expectations of having novel antiplatelet drugs which selectively inhibit arterial thrombosis without interfering with normal haemostasis could possibly be met in the near future.


Assuntos
Inibidores da Agregação Plaquetária/uso terapêutico , Glicoproteínas da Membrana de Plaquetas/antagonistas & inibidores , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Humanos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Receptor PAR-1/antagonistas & inibidores , Trombina/antagonistas & inibidores , Trombose/sangue , Trombose/prevenção & controle , Fator de von Willebrand/antagonistas & inibidores
8.
Sci Transl Med ; 8(367): 367ra168, 2016 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-27903864

RESUMO

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.


Assuntos
Adesividade Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Trombina/antagonistas & inibidores , Trombose/patologia , Síndrome Coronariana Aguda/patologia , Administração Oral , Animais , Anticoagulantes/farmacologia , Artérias/patologia , Aspirina/farmacologia , Aterosclerose/patologia , Sítios de Ligação , Plaquetas/efeitos dos fármacos , Fibrinolíticos/farmacologia , Humanos , Camundongos , Inibidores da Agregação Plaquetária/farmacologia , Ensaios Clínicos Controlados Aleatórios como Assunto , Vitamina K/antagonistas & inibidores
9.
JACC Basic Transl Sci ; 1(3): 131-142., 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27766315

RESUMO

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.

10.
Circulation ; 134(16): 1176-1188, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27660294

RESUMO

BACKGROUND: Therapeutic targeting of arterial leukocyte recruitment in the context of atherosclerosis has been disappointing in clinical studies. Reasons for such failures include the lack of knowledge of arterial-specific recruitment patterns. Here we establish the importance of the cathepsin G (CatG) in the context of arterial myeloid cell recruitment. METHODS: Intravital microscopy of the carotid artery, the jugular vein, and cremasteric arterioles and venules in Apoe-/-and CatG-deficient mice (Apoe-/-Ctsg-/-) was used to study site-specific myeloid cell behavior after high-fat diet feeding or tumor necrosis factor stimulation. Atherosclerosis development was assessed in aortic root sections after 4 weeks of high-fat diet, whereas lung inflammation was assessed after inhalation of lipopolysaccharide. Endothelial deposition of CatG and CCL5 was quantified in whole-mount preparations using 2-photon and confocal microscopy. RESULTS: Our observations elucidated a crucial role for CatG during arterial leukocyte adhesion, an effect not found during venular adhesion. Consequently, CatG deficiency attenuates atherosclerosis but not acute lung inflammation. Mechanistically, CatG is immobilized on arterial endothelium where it activates leukocytes to firmly adhere engaging integrin clustering, a process of crucial importance to achieve effective adherence under high-shear flow. Therapeutic neutralization of CatG specifically abrogated arterial leukocyte adhesion without affecting myeloid cell adhesion in the microcirculation. Repetitive application of CatG-neutralizing antibodies permitted inhibition of atherogenesis in mice. CONCLUSIONS: Taken together, these findings present evidence of an arterial-specific recruitment pattern centered on CatG-instructed adhesion strengthening. The inhibition of this process could provide a novel strategy for treatment of arterial inflammation with limited side effects.


Assuntos
Artérias , Catepsina G/metabolismo , Quimiotaxia , Células Mieloides/metabolismo , Vênulas , Animais , Aterosclerose/tratamento farmacológico , Aterosclerose/etiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Biomarcadores , Catepsina G/antagonistas & inibidores , Catepsina G/genética , Adesão Celular/genética , Quimiocina CCL5/genética , Quimiocina CCL5/metabolismo , Quimiotaxia/genética , Quimiotaxia/imunologia , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Humanos , Integrinas/metabolismo , Migração e Rolagem de Leucócitos , Camundongos , Camundongos Knockout , Microcirculação , Células Mieloides/imunologia , Ligação Proteica , Resistência ao Cisalhamento
11.
J Am Coll Cardiol ; 65(22): 2404-15, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26046734

RESUMO

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.


Assuntos
Velocidade do Fluxo Sanguíneo/fisiologia , Artérias Carótidas/fisiopatologia , Fragmentos Fab das Imunoglobulinas/farmacologia , Placa Aterosclerótica/tratamento farmacológico , Ativação Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/farmacologia , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Estenose das Carótidas/tratamento farmacológico , Estenose das Carótidas/etiologia , Estenose das Carótidas/fisiopatologia , Humanos , Fatores Imunológicos/farmacologia , Placa Aterosclerótica/complicações , Placa Aterosclerótica/diagnóstico , Ratos
12.
Biochem Pharmacol ; 86(2): 279-85, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23665353

RESUMO

Two synthetic LXR agonists were recently reported to inhibit collagen-induced platelet aggregation and thrombus formation in mice. We therefore studied whether also natural LXR agonists inhibit human platelet activation and whether they can be fluorescence-labelled preserving their bioactivity for LXR-related functional imaging. The natural LXR agonist 22(R)-OH-cholesterol - but not its stereoisomer 22(S)-OH-cholesterol - inhibited collagen induced platelet shape change and aggregation similar to synthetic LXR agonists in a concentration- and time-dependent manner. First exposure to 22(S)-OH-cholesterol prevented the subsequent inhibition of platelets by 22(R)-OH-cholesterol but not vice versa. 22(R)- and 22(S)-OH-cholesterol could be fluorescence-labelled as 22(R)- and 22(S)-OH-cholesteryl-3-dodecanoic-3-BODIPY esters with high yield and purity using the Steglich acylation. Labelled 22(R)- and 22(S)-OH-cholesterol esters retained the stereo specific bioactivity of their parent compounds, were metabolically stable and not cytotoxic at LXR agonistic concentrations. Live staining with labelled 22(R)- or 22(S)-OH-cholesterol esters demonstrated stereo specific inhibition of platelet spreading and chiral handling by macrophages that reflect LXR activation. The rapid inhibition of platelet reactivity to collagen by natural and pharmacologic LXR agonists offers a mechanism that could attenuate platelet activation by denuded plaques that expose collagen and LXR agonistic oxysterols. Stable fluorescence labelled 22(R)- and 22(S)-OH-cholesterol analogues with preserved stereo specific bioactivity and staining characteristics provide valuable tools for LXR-related functional imaging in pathophysiologic studies, for binding assays and for LXR-targeted drug development.


Assuntos
Hidroxicolesteróis/farmacologia , Macrófagos/efeitos dos fármacos , Receptores Nucleares Órfãos/agonistas , Ativação Plaquetária/efeitos dos fármacos , Acilação , Corantes Fluorescentes , Humanos , Cinética , Receptores X do Fígado , Macrófagos/metabolismo , Microscopia de Fluorescência , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...