Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur Heart J ; 43(6): 504-514, 2022 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-34529782

RESUMO

AIMS: Platelets participate in atherogenesis with mechanisms not yet fully clarified. Vascular wall MMP-2 is involved in the arterial remodelling accompanying atherosclerosis. Platelets contain and release MMP-2 but no informations are available on its role in atherosclerotic lesion formation. METHODS AND RESULTS: We generated double knockout mice lacking the LDL receptor and MMP-2 only in circulating blood cells showing that they develop significantly lesser femoral intima thickening after photochemical-induced arterial damage and atherosclerotic lesions in the aorta, measured by the en face method, after 4 months of atherogenic diet. Moreover, repeated transfusions of autologous-activated platelets in LDLR-/- mice on atherogenic diet significantly enhanced the extension of aortic atherosclerotic lesions while transfusion of activated platelets from MMP-2-/- mice did not. In vitro coincubation studies showed that platelet-derived MMP-2 plays a pivotal role in the development and progression of atherosclerosis through a complex cross-talk between activated platelets, monocyte/macrophages, and endothelial cells. Translational studies in patients with CAD and chronic HIV infection showed that platelet surface expression of MMP-2 highly significantly correlated with the degree of carotid artery stenosis. CONCLUSION: We show a previously unknown mechanism of the pathway through which platelets expressing MMP-2 trigger the initial phases of atherosclerosis and provide a mechanism showing that they activate endothelial PAR-1 triggering endothelial p38MAPK signalling and the expression of adhesion molecules. The development of drugs blocking selectively platelet MMP-2 or its expression may represent a new approach to the prevention of atherosclerosis.


Assuntos
Aterosclerose , Infecções por HIV , Animais , Aterosclerose/metabolismo , Plaquetas/metabolismo , Células Endoteliais/metabolismo , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
2.
Haematologica ; 107(7): 1643-1654, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34407603

RESUMO

Platelet-type von Willebrand disease (PT-VWD) is an inherited platelet disorder. It is characterized by macrothrombocytopenia and mucocutaneous bleeding, of variable severity, due to gain-of-function variants of GP1BA conferring to glycoprotein Ibα (GPIbα) enhanced affinity for von Willebrand factor (VWF). The bleeding tendency is conventionally attributed to thrombocytopenia and large VWF-multimer depletion. However, while some indications suggest that platelet dysfunction may contribute to the bleeding phenotype, no information on its characteristics and causes are available. The aim of the present study was to characterize platelet dysfunction in PT-VWD and shed light on its mechanism. Platelets from a PT-VWD patient carrying the p.M239V variant, and from PT-VWD mice carrying the p.G233V variant, showed a remarkable platelet function defect, with impaired aggregation, defective granule secretion and reduced adhesion under static and flow conditions. VWFbinding to GPIbα is known to trigger intracellular signaling involving Src-family kinases (SFK). We found that constitutive phosphorylation of the platelet SFK Lyn induces a negative-feedback loop downregulating platelet activation through phosphorylation of PECAM1 on Tyr686 and that this is triggered by the constitutive binding of VWF to GPIbα. These data show, for the first time, that the abnormal triggering of inhibitory signals mediated by Lyn and PECAM1 may lead to platelet dysfunction. In conclusion, our study unravels the mechanism of platelet dysfunction in PT-VWD caused by deranged inhibitory signaling. This is triggered by the constitutive binding of VWF to GPIbα which may significantly contribute to the bleeding phenotype of these patients.


Assuntos
Trombocitopenia , Doenças de von Willebrand , Animais , Plaquetas/metabolismo , Hemorragia/genética , Camundongos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Trombocitopenia/genética , Trombocitopenia/metabolismo , Doenças de von Willebrand/genética , Doenças de von Willebrand/metabolismo , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
3.
Haematologica ; 107(6): 1374-1383, 2022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35142150

RESUMO

Deep vein thrombosis results from the cooperative action of leukocytes, platelets, and endothelial cells. The proline-rich tyrosine kinase Pyk2 regulates platelet activation and supports arterial thrombosis. In this study, we combined pharmacological and genetic approaches to unravel the role of Pyk2 in venous thrombosis. We found that mice lacking Pyk2 almost completely failed to develop deep venous thrombi upon partial ligation of the inferior vena cava. Pyk2-deficient platelets displayed impaired exposure of phosphatidylserine and tissue factor expression by endothelial cells and monocytes was completely prevented by inhibition of Pyk2. In human umbilical vein endothelial cells (HUVEC), inhibition of Pyk2 hampered IL-1b-induced expression of VCAM and P-selectin, and von Willebrand factor release. Pyk2-deficient platelets showed defective adhesion on von Willebrand factor and reduced ability to bind activated HUVEC under flow. Moreover, inhibition of Pyk2 in HUVEC strongly reduced platelet adhesion. Similarly, Pyk2-deficient neutrophils were unable to efficiently roll and adhere to immobilized endothelial cells under venous flow conditions. Moreover, platelets and neutrophils from Pyk2- knockout mice showed defective ability to form heterogeneous aggregates upon stimulation, while platelet monocyte interaction occurred normally. Consequently, platelet neutrophil aggregates, abundant in blood of wild-type mice upon inferior vena cava ligation, were virtually undetectable in Pyk2-knockout mice. Finally, we found that expression of Pyk2 was required for NETosis induced by activated platelets. Altogether our results demonstrate a critical role of Pyk2 in the regulation of the coordinated thromboinflammatory responses of endothelial cells, leukocytes and platelets leading to venous thrombosis. Pyk2 may represent a novel promising target in the treatment of deep vein thrombosis.


Assuntos
Quinase 2 de Adesão Focal/metabolismo , Trombose Venosa , Animais , Plaquetas/metabolismo , Células Endoteliais/metabolismo , Quinase 2 de Adesão Focal/genética , Humanos , Camundongos , Fosforilação , Prolina/metabolismo , Trombose Venosa/genética , Trombose Venosa/metabolismo , Fator de von Willebrand/metabolismo
4.
Haematologica ; 106(12): 3034-3045, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34407607

RESUMO

The COVID-19 pandemic has had a heavy impact on global health and economy and vaccination remains the primary way of controlling the infection. During the ongoing vaccination campaign some unexpected thrombotic events have emerged in subjects who had recently received the AstraZeneca (Vaxzevria) vaccine or the Johnson and Johnson (Janssen) vaccine, two adenovirus vector-based vaccines. Epidemiological studies confirm that the observed/expected ratio of these unusual thromboses is abnormally increased, especially in women in fertile age. The characteristics of this complication, with venous thromboses at unusual sites, most frequently in the cerebral vein sinuses but also in splanchnic vessels, often with multiple associated thromboses, thrombocytopenia, and sometimes disseminated intravascular coagulation, are unique and the time course and tumultuous evolution are suggestive of an acute immunological reaction. Indeed, plateletactivating anti-PF4 antibodies have been detected in a large proportion of the affected patients. Several data suggest that adenoviruses may interact with platelets, the endothelium and the blood coagulation system. Here we review interactions between adenoviral vectors and the hemostatic system that are of possible relevance in vaccine-associated thrombotic thrombocytopenia syndrome. We systematically analyze the clinical data on the reported thrombotic complications of adenovirus-based therapeutics and discuss all the current hypotheses on the mechanisms triggering this novel syndrome. Although, considering current evidence, the benefit of vaccination clearly outweighs the potential risks, it is of paramount importance to fully unravel the mechanisms leading to vaccineassociated thrombotic thrombocytopenia syndrome and to identify prognostic factors through further research.


Assuntos
COVID-19 , Trombocitopenia , Trombose , Vacinas , Adenoviridae , Coagulação Sanguínea , Plaquetas , Vacinas contra COVID-19 , Feminino , Humanos , Pandemias , SARS-CoV-2 , Trombocitopenia/etiologia , Trombose/etiologia
5.
Blood ; 129(7): 883-895, 2017 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-28034890

RESUMO

Platelets contain and release several matrix metalloproteinases (MMPs). Among these, active MMP-2 enhances platelet aggregation by favoring the activation of phosphatidylinositol 3- kinase (PI3K) and contributes to arterial thrombosis. The platelet surface target of MMP-2 and the mechanism through which it primes platelets to respond to subsequent stimuli are still unknown. We show that active MMP-2 enhances platelet activation induced by weak stimuli by cleaving PAR1 at a noncanonical extracellular site different from the thrombin-cleavage site and thus initiates biased receptor signaling, triggering only some of the signaling pathways normally activated by full PAR1 agonism. The novel PAR1-tethered ligand exposed by MMP-2 stimulates PAR1-dependent Gq and G12/13 pathway activation, triggering p38-MAPK phosphorylation, Ca+2 fluxes, and PI3K activation, but not Gi signaling; this is insufficient to cause platelet aggregation, but it is enough to predispose platelets to fully respond to Gi-activating stimuli. Integrin αIIbß3 is a necessary cofactor for PAR1 cleavage by MMP-2 by binding the MMP-2 hemopexin domain, thus favoring the interaction of the enzyme with PAR1. Our studies unravel a novel mechanism regulating platelet activation that involves the binding of MMP-2 to integrin αIIbß3 and the subsequent cleavage of PAR1 by active MMP-2 at a noncanonical site, exposing a previously undescribed tethered ligand that triggers biased G-protein agonism and thus predisposes platelets to full activation by other stimuli. These results identify the MMP-2-αIIbß3-PAR1 interaction as a potential target for the prevention of arterial thrombosis.


Assuntos
Plaquetas/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Ativação Plaquetária , Receptor PAR-1/metabolismo , Transdução de Sinais , Animais , Plaquetas/citologia , Células CHO , Cricetulus , Humanos , Camundongos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Mapas de Interação de Proteínas
6.
Blood ; 130(4): 527-536, 2017 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-28611024

RESUMO

The amyloid precursor protein (APP), primarily known as the precursor of amyloid peptides that accumulate in the brain of patients with Alzheimer disease, is abundant in platelets, but its physiological function remains unknown. In this study, we investigated the role of APP in hemostasis and thrombosis, using APP knockout (KO) mice. Ex vivo aggregation, secretion, and integrin αIIbß3 inside-out activation induced by several agonists were normal in APP-deficient platelets, but the number of circulating platelets was reduced by about 20%, and their size was slightly increased. Tail bleeding time was normal, and in vivo, the absence of APP did not alter thrombus formation in the femoral artery. In contrast, in a model of vein thrombosis induced by flow restriction in the inferior vena cava, APP-KO mice, as well as chimeric mice with selective deficiency of APP in blood cells, developed much larger thrombi than control animals, and were more sensitive to embolization. Consistent with this, in a pulmonary thromboembolism model, larger vessels were occluded. APP-KO mice displayed a shorter APTT, but not PT, when measured in the presence of platelets. Moreover, the activity of factor XIa (FXIa), but not FXIIa, was higher in APP-KO mice compared with controls. APP-KO mice presented a higher number of circulating platelet-leukocyte aggregates, and neutrophils displayed a greater tendency to protrude extracellular traps, which were more strongly incorporated into venous thrombi. These results indicate that platelet APP limits venous thromboembolism through a negative regulation of both fibrin formation and neutrophil function.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Plaquetas/metabolismo , Veia Cava Inferior/metabolismo , Tromboembolia Venosa/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Plaquetas/patologia , Fator XIa/genética , Fator XIa/metabolismo , Camundongos , Camundongos Knockout , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Veia Cava Inferior/patologia , Tromboembolia Venosa/genética , Tromboembolia Venosa/patologia
7.
Haematologica ; 104(7): 1473-1481, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30655369

RESUMO

Platelet-type von Willebrand disease is an inherited platelet disorder characterized by thrombocytopenia with large platelets caused by gain-of-function variants in GP1BA leading to enhanced GPIbα-von Willebrand factor (vWF) interaction. GPIbα and vWF play a role in megakaryocytopoiesis, thus we aimed to investigate megakaryocyte differentiation and proplatelet-formation in platelet-type von Willebrand disease using megakaryocytes from a patient carrying the Met239Val variant and from mice carrying the Gly233Val variant. Platelet-type von Willebrand disease megakaryocytes bound vWF at an early differentiation stage and generated proplatelets with a decreased number of enlarged tips compared to control megakaryocytes. Moreover, they formed proplatelets upon contact with collagen, differently from normal megakaryocytes. Similarly, collagen triggered megakaryocytes showed defective activation of the RhoA-MLC2 axis, which prevents proplatelet formation, and increased phosphorylation of Lyn, which acts as a negative regulator of GPVI signaling, thus preventing ectopic proplatelet-formation on collagen. Consistently, human and murine bone marrow contained an increased number of extravascular platelets compared to controls. In addition, platelet survival of mutant mice was shortened compared to control mice, and the administration of desmopressin, raising circulating vWF, caused a marked drop in platelet count. Taken together, these results show for the first time that thrombocytopenia in platelet-type von Willebrand disease is due to the combination of different pathogenic mechanisms, i.e. the formation of a reduced number of platelets by megakaryocytes, the ectopic release of platelets in the bone marrow, and the increased clearance of platelet/vWF complexes.


Assuntos
Plaquetas/patologia , Megacariócitos/patologia , Mutação , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Trombocitopenia/fisiopatologia , Doenças de von Willebrand/patologia , Fator de von Willebrand/metabolismo , Animais , Plaquetas/metabolismo , Estudos de Casos e Controles , Movimento Celular , Humanos , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Trombocitopenia/metabolismo , Trombopoese , Doenças de von Willebrand/metabolismo , Fator de von Willebrand/genética
8.
Blood ; 125(4): 697-705, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25480660

RESUMO

Nitric oxide (NO) exerts vasodilatatory, antiplatelet, antioxidant, and antiproliferative effects. Endothelium-derived NO has been shown to be of crucial importance in cardiovascular protection, whereas evidence that NO is synthesized by platelets and regulates platelet function is still controversial. By using a sensitive and specific fluorescent probe, 4-amino-5-methylamino-2',7'-difluorofluorescein diacetate (DAF-FM), we visualized NO production in individual platelets undergoing adhesion on a collagen substrate under flow conditions. NO production, monitored in real time, was dependent on the shear rates applied, increasing with the raising of the shear rates. Furthermore, NO production increased in the presence of l-arginine (nitric-oxide synthase [NOS] substrate), and it decreased in the presence of L-NG-monomethyl arginine (L-NMMA) (NOS inhibitor) but not of D-NG-monomethyl arginine (D-NMMA) (L-NMMA-inactive enantiomer). Platelet deposition, measured with mepacrine-labeled platelets, was inversely related to NO production. A correlation was evident between Ca(++) elevation and NO production, suggesting that platelet NO formation is triggered by intracytoplasmic Ca(++) elevation. Simultaneous measurement of NO and Ca(++) indicated that NO production in individual platelets is preceded by Ca(++) elevations, with a lag phase of 33 ± 9.5 s. Our studies provide the first direct demonstration of platelet NO production triggered by the interaction with an activating surface under flow and suggest that intraplatelet Ca(++) elevation elicits the production of NO which, in turn, modulates thrombus size.


Assuntos
Plaquetas/metabolismo , Óxido Nítrico/metabolismo , Adesividade Plaquetária/fisiologia , Animais , Velocidade do Fluxo Sanguíneo , Plaquetas/citologia , Cálcio/metabolismo , Colágeno/farmacologia , Inibidores Enzimáticos/farmacologia , Fluoresceínas/farmacocinética , Masculino , Camundongos , Camundongos Knockout , Adesividade Plaquetária/efeitos dos fármacos , Quinacrina/farmacologia , ômega-N-Metilarginina/farmacologia
9.
Clin Chem Lab Med ; 54(1): 97-103, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26087065

RESUMO

BACKGROUND: Subconjunctival hemorrage (SCH) is a frequent, mild bleeding manifestation and a common cause of consultation. Hemostatic alterations are possible causes of SCH but their role and prevalence is unknown. We assessed the prevalence of hemostatic abnormalities in patients with spontaneous, recurrent SCH to clarify the role of the hemostasis laboratory in this clinical setting. METHODS: A total of 105 SCH patients (21-78 years, 65 females) with no identifiable cause (hypertension-trauma-conjunctivitis) or concomitant treatments (NSAIDs- aspirin-oral anticoagulants-antiplatelet agents) and 53 age and sex-matched healthy controls (HCs) (22-72 years, 29 females) were evaluated for skin bleeding time, PFA-100®, blood clotting screening, platelet count, light transmission aggregomery, VWF:Ag, VWF:RCo, RIPA, FVIII activity, FXIII antigen and activity and ISTH Bleeding Severity Score (BSS). RESULTS: Prevalence of hemostatic abnormalities was not higher in the SCH population than in HCs BSS was 0.83 (95% CI 0.62-1.06) in SCH and 0.66 (0.37-0.95) in HC (p=NS). Type I Von Willebrand disease was diagnosed in one SCH and none HC patients, a prevalence not significantly different (p=NS by χ2). CONCLUSIONS: The prevalence of hemostatic alterations in patients with recurrent, spontaneous SCH is not different from the general population; hemostatic screening or second level tests are of no use in patients with recurrent SCH and no other bleedings.


Assuntos
Hemorragia Ocular/tratamento farmacológico , Hemorragia Ocular/epidemiologia , Hemostáticos/efeitos adversos , Adulto , Idoso , Idoso de 80 Anos ou mais , Hemorragia Ocular/diagnóstico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Prevalência , Recidiva , Adulto Jovem
10.
J Allergy Clin Immunol ; 135(2): 528-38, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25445826

RESUMO

BACKGROUND: Clinical studies reveal platelet activation in patients with asthma, allergic rhinitis, and eczema. This is distinct from platelet aggregation, which is critical for the maintenance of hemostasis and in which a role for platelet purinergic receptors is well documented. However, purines are also essential for inflammatory cell trafficking in animal models of allergic lung inflammation, which are known to be platelet dependent, yet the role of purines in the platelet activation accompanying inflammation is unknown. OBJECTIVES: We investigated whether the involvement of purine activation of platelets during allergic inflammation is distinct from purine involvement in platelet aggregation. METHODS: BALB/c mice were sensitized to ovalbumin and subsequent airway ovalbumin challenge. Bronchoalveolar lavage fluid was analyzed for inflammatory cells, and blood samples were assessed for platelet activation. The role of platelet purinergic receptors and associated signaling mechanisms (RhoA) were assessed. RESULTS: P2Y1, but not P2Y12 or P2X1, antagonism inhibited pulmonary leukocyte recruitment. The formation of platelet-leukocyte complexes in vivo and platelet/P-selectin-dependent polymorphonuclear cell migration in vitro were exclusively platelet P2Y1 receptor dependent. Furthermore, platelet P2Y1 activation resulted in RhoA activity in vivo after allergen challenge, and RhoA signaling in platelets through P2Y1 stimulation was required for platelet-dependent leukocyte chemotaxis in vitro. Leukocyte recruitment in thrombocytopenic mice remained suppressed after reinfusion of platelets pretreated with a P2Y1 antagonist or a Rho-associated kinase 1 inhibitor, confirming the crucial role of platelet P2Y1 receptor and subsequent activation of RhoA. CONCLUSION: RhoA signaling downstream of platelet P2Y1, but not P2Y12, represents a clear dichotomy in platelet activation during allergic inflammation versus hemostasis.


Assuntos
Difosfato de Adenosina/farmacologia , Quimiotaxia de Leucócito/imunologia , Hipersensibilidade/imunologia , Hipersensibilidade/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Alérgenos/imunologia , Animais , Plaquetas/imunologia , Quimiocinas/metabolismo , Modelos Animais de Doenças , Feminino , Pulmão/imunologia , Pulmão/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Modelos Biológicos , Ovalbumina/imunologia , Selectina-P/metabolismo , Agregação Plaquetária , Agonistas do Receptor Purinérgico P2Y/farmacologia , Transdução de Sinais
11.
Blood ; 121(25): 5088-97, 2013 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-23589671

RESUMO

Thrombolytic therapy is the cornerstone of treatment of acute atherothrombotic ischemic stroke but is associated with brain hemorrhage; antiplatelet therapy has limited efficacy and is still associated with intracranial bleeding. Therefore, new antithrombotic approaches with a better efficacy/safety ratio are required. We have assessed the effect of ALX-0081, a Nanobody against the A1 domain of von Willebrand factor (VWF) that blocks VWF binding to GPIb, of the thrombolytic agent recombinant tissue plasminogen activator (rtPA), and of the GPIIb/IIIa antagonist tirofiban, in a middle cerebral artery (MCA) thrombosis model in guinea pigs. Drugs were administered before, immediately after, or 15 or 60 minutes after the total occlusion of the MCA. ALX-0081 prevented MCA thrombosis and induced reperfusion when given immediately after and 15 minutes after complete occlusion and reduced brain damage without inducing hemorrhage, whereas tirofiban prevented thrombosis but did not induce reperfusion and induced striking brain hemorrhage. rtPA also induced reperfusion when given 60 minutes after occlusion but provoked brain hemorrhage. Skin bleeding time was not modified or was moderately prolonged by ALX-0081, whereas tirofiban and rtPA prolonged it. The inhibition of the GPIb-VWF axis in guinea pigs prevents cerebral artery thrombosis and induces early reperfusion without provoking intracerebral bleeding thus reducing brain infarct area.


Assuntos
Infarto Encefálico/prevenção & controle , Fibrinolíticos/farmacologia , Trombose Intracraniana/tratamento farmacológico , Anticorpos de Domínio Único/farmacologia , Terapia Trombolítica/métodos , Animais , Artérias Cerebrais/patologia , Modelos Animais de Doenças , Cobaias , Masculino , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Reperfusão/métodos , Fator de von Willebrand/antagonistas & inibidores
12.
Blood ; 121(4): 648-57, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23175689

RESUMO

In the present study, we used a knockout murine model to analyze the contribution of the Ca(2+)-dependent focal adhesion kinase Pyk2 in platelet activation and thrombus formation in vivo. We found that Pyk2-knockout mice had a tail bleeding time that was slightly increased compared with their wild-type littermates. Moreover, in an in vivo model of femoral artery thrombosis, the time to arterial occlusion was significantly prolonged in mice lacking Pyk2. Pyk2-deficient mice were also significantly protected from collagen plus epinephrine-induced pulmonary thromboembolism. Ex vivo aggregation of Pyk2-deficient platelets was normal on stimulation of glycoprotein VI, but was significantly reduced in response to PAR4-activating peptide, low doses of thrombin, or U46619. Defective platelet aggregation was accompanied by impaired inside-out activation of integrin α(IIb)ß(3) and fibrinogen binding. Granule secretion was only slightly reduced in the absence of Pyk2, whereas a marked inhibition of thrombin-induced thromboxane A(2) production was observed, which was found to be responsible for the defective aggregation. Moreover, we have demonstrated that Pyk2 is implicated in the signaling pathway for cPLA(2) phosphorylation through p38 MAPK. The results of the present study show the importance of the focal adhesion kinase Pyk2 downstream of G-protein-coupled receptors in supporting platelet aggregation and thrombus formation.


Assuntos
Quinase 2 de Adesão Focal/genética , Ativação Plaquetária/genética , Trombina/metabolismo , Trombose/genética , Trombose/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Cálcio/metabolismo , Fosfolipases A2 do Grupo II/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/genética , Transdução de Sinais , Trombina/farmacologia , Tromboxano A2/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 34(4): 820-9, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24558107

RESUMO

OBJECTIVE: dl-Nebivolol, a selective ß1-adrenergic receptor antagonist, besides its hypotensive activity exerts vasodilatory and platelet inhibitory effects in vitro by a mechanism involving nitric oxide (NO). Our aim was to evaluate whether nebivolol exerts in vivo antithrombotic effects, to unravel the mechanism of this action and to clarify the relative roles of its 2 enantiomers: d- and l-nebivolol. METHODS AND RESULTS: In wild-type mice, dl-nebivolol, l-nebivolol, and d-nebivolol, but not bisoprolol, reduced mortality consequent to platelet pulmonary thromboembolism induced by the intravenous injection of collagen plus epinephrine (-44%, -45%, -29%, respectively; P<0.05), whereas in eNOS(-/-) mice only dl-nebivolol and d-nebivolol were effective. dl-Nebivolol, l- and d-nebivolol reduced photochemical damage-induced femoral artery thrombosis in wild-type mice, whereas in eNOS(-/-) mice only dl-nebivolol and d-nebivolol were active. Moreover, dl-nebivolol and l-nebivolol increased plasma, urinary-, and platelet-derived nitrites and nitrates (NOx), NO degradation products, in wild-type but not in eNOS(-/-) mice. In vivo platelet activation, assessed by platelet P-selectin expression, was reduced by dl-nebivolol and l- and d-nebivolol in wild-type mice but only by dl-nebivolol and d-nebivolol in eNOS(-/-) mice. In bone marrow-transplanted, chimeric mice with only blood cells, and not the endothelium, producing NO dl-nebivolol and l-nebivolol maintained their antithrombotic activity, whereas they lose it in chimeras with only endothelium, and not blood cells, producing NO. In vitro, with isolated platelets, dl-nebivolol and l-nebivolol, but not d-nebivolol and bisoprolol, increased platelet cGMP and NOx formation. Treatment with dl-nebivolol and l-nebivolol increased phophorylated eNOS in platelets. CONCLUSIONS: Our data show that dl-nebivolol exerts an antithrombotic activity by stimulating the formation of NO by platelets, and that this effect is generated by its l-enantiomer, whereas the d-enantiomer exerts a weak antiplatelet effect because of ß-adrenergic receptor-independent stimulation of adenyly cyclase. These results confirm that platelet-derived NO plays a role in thrombosis prevention and it may represent a target of pharmacological intervention.


Assuntos
Benzopiranos/farmacologia , Plaquetas/efeitos dos fármacos , Etanolaminas/farmacologia , Fibrinolíticos/farmacologia , Óxido Nítrico/sangue , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Tromboembolia/prevenção & controle , Trombose/prevenção & controle , Antagonistas de Receptores Adrenérgicos beta 1/farmacologia , Animais , Antioxidantes/farmacologia , Benzopiranos/química , Plaquetas/metabolismo , Pressão Sanguínea/efeitos dos fármacos , GMP Cíclico/sangue , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Etanolaminas/química , Fibrinolíticos/química , Isomerismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nebivolol , Óxido Nítrico Sintase Tipo III/deficiência , Óxido Nítrico Sintase Tipo III/genética , Fosforilação , Inibidores da Agregação Plaquetária/química , Selenoproteína P/sangue , Tromboembolia/sangue , Tromboembolia/fisiopatologia , Trombose/sangue , Trombose/fisiopatologia , Fatores de Tempo , Regulação para Cima
14.
Rev Cardiovasc Med ; 15 Suppl 1: S9-20, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24987863

RESUMO

Highly active antiretroviral therapy (HAART) has transformed human immunodeficiency virus (HIV) infection into a chronic condition, which has allowed the infected population to age and become prone to chronic degenerative diseases common to the general population, including atherosclerotic cardiovascular disease, and coronary artery disease (CAD). Possible causative mechanisms of HIV-associated CAD are related to classic cardiovascular risk factors, such as dyslipidemia, insulin resistance, and fat redistribution, which may be due to either HIV infection or to HAART-associated toxicity. However, other mechanisms are emerging as crucial for the cardiovascular complication of HIV and HAART. This article analyzes the effects of HIV and HAART on endothelial function, endothelium-leukocyte interactions, and platelets as possible mechanisms of enhanced cardiovascular risk.


Assuntos
Fármacos Anti-HIV/efeitos adversos , Plaquetas/efeitos dos fármacos , Doenças Cardiovasculares/induzido quimicamente , Células Endoteliais/efeitos dos fármacos , Infecções por HIV/tratamento farmacológico , Animais , Terapia Antirretroviral de Alta Atividade , Plaquetas/imunologia , Plaquetas/metabolismo , Plaquetas/virologia , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/imunologia , Doenças Cardiovasculares/virologia , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Infecções por HIV/sangue , Infecções por HIV/imunologia , Infecções por HIV/virologia , Sobreviventes de Longo Prazo ao HIV , Humanos , Inflamação/sangue , Inflamação/imunologia , Inflamação/virologia , Prognóstico , Fatores de Risco , Fatores de Tempo
15.
Arthritis Rheum ; 64(4): 1089-97, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22042442

RESUMO

OBJECTIVE: To assess the role of matrix metalloproteinase 2 (MMP-2) in the evolution of septic arthritis induced by group B streptococci (GBS) in mice. METHODS: Mice deficient in MMP-2 (MMP-2(-/-) ) and wild-type controls were injected intravenously with 1 × 10(7) colony-forming units of type IV GBS (strain 1/82). Levels of MMP-2, mortality rates, evolution of arthritis, bacterial clearance, joint histopathologic features, and production of cytokines and chemokines were examined in both experimental groups of mice on days 3, 6, and 9 after infection. RESULTS: MMP-2 was produced during GBS infection. Disruption of the gene for MMP-2 resulted in a decrease in the incidence and severity of arthritis, as demonstrated by both clinical and histologic findings, without affecting mortality rates. Amelioration of arthritis was accompanied by a dramatic reduction in the local production of interleukin-1ß (IL-1ß), IL-6, macrophage inflammatory protein 1α (MIP-1α), and MIP-2 and a reduced bacterial burden. CONCLUSION: MMP-2, produced early during GBS infection in mice, is involved in the degradation of extracellular matrix components at the level of the joint. This degradation is the first step in a cascade of events (joint invasion by GBS, extravasation and accumulation of inflammatory cells, proinflammatory cytokine production), all of which contribute to the damage of articular tissue. Thus, MMP-2 should be regarded as a potential therapeutic target in GBS-induced arthritis.


Assuntos
Artrite Experimental/metabolismo , Artrite Infecciosa/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Infecções Estreptocócicas/metabolismo , Streptococcus agalactiae , Animais , Artrite Experimental/microbiologia , Artrite Experimental/patologia , Artrite Infecciosa/microbiologia , Artrite Infecciosa/patologia , Feminino , Interleucina-1beta/metabolismo , Articulações/metabolismo , Articulações/patologia , Metaloproteinase 2 da Matriz/genética , Camundongos , Camundongos Knockout , Infecções Estreptocócicas/patologia
16.
Handb Exp Pharmacol ; (210): 287-309, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22918736

RESUMO

The formation of platelet-rich thrombi, a critical step in the pathogenesis of atherothrombotic events, is a multistep process involving several components, among which von Willebrand Factor (VWF) plays a central role. Ruptured atherosclerotic plaques expose subendothelial matrix proteins which bind VWF that represents a bridge between the injured blood vessel and activated platelets, playing a crucial role in platelet adhesion and aggregation, especially in conditions of high-shear rate. Due to these peculiarities, the binding of VWF to GPIbα is an attractive drug target. Here we summarize the present knowledge on the different classes of drugs targeting the VWF-GPIb interaction and we give an account of their level of clinical development. In particular, the following compounds are discussed: AJW200, an IgG4 humanized monoclonal antibody against VWF-A1; 82D6A3, a monoclonal antibody against VWF-A3; ALX-0081 and ALX-0681, bivalent humanized nanobodies targeting the VWF-A1 domain; ARC1779 and its advanced formulation ARC15105, second-generation aptamers that bind the VWF-A1 domain; h6B4-Fab, a murine monoclonal antibody, and GPG-290, a recombinant chimeric protein, both directed against GPIbα.


Assuntos
Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Fator de von Willebrand/antagonistas & inibidores , Animais , Aptâmeros de Nucleotídeos/uso terapêutico , Humanos , Ativação Plaquetária , Inibidores da Agregação Plaquetária/uso terapêutico , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Trombose/tratamento farmacológico , Fator de von Willebrand/química , Fator de von Willebrand/fisiologia
17.
Eur Heart J ; 32(3): 316-25, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21036774

RESUMO

AIMS: To investigate whether selected matrix metalloproteinases (MMPs) are released in the coronary circulation of patients with acute coronary syndrome (ACS), whether this release is related to platelet activation, and whether it contributes to sustained platelet activation. METHODS AND RESULTS: Blood from the aorta (Ao) and the coronary sinus (Cs) was obtained from 21 controls (non-cardiac chest pain), 24 stable angina (SA), and 30 ACS patients, before performing percutaneous transluminal coronary angioplasty. Selected MMPs, some platelet activation- and atheroma-related markers, and the platelet activation-potentiating activity of plasma were measured. Total MMP-2, active MMP-2, and MMP-9 were released in the coronary circulation of patients with ACS, but not of those with SA or controls. Similarly, transcoronary gradients of ß-thromboglobulin (ß-TG) and platelet factor 4, two platelet-specific proteins, and of soluble CD40L and secretory phospholipase A2 (sPLA2), markers of inflammation and platelet activation, were higher in ACS patients than in the other groups. In contrast, plasma monocyte chemoattractant protein-1, a platelet-unrelated marker of atherogenesis, was not increased in the Cs compared with Ao in any of the groups. Transcoronary gradients of both ß-TG and sPLA2 correlated with those of total and active MMP-2 in ACS, but not in controls or SA. Plasma from the Cs of ACS patients potentiated platelet activation, an effect suppressed by the specific MMP-2-inhibitor, tissue inhibitor of MMP-2 (TIMP-2). CONCLUSION: Matrix metalloproteinase-2 is released in the coronary circulation of ACS patients, derives in part from activated platelets, and may contribute to sustained intracoronary platelet activation.


Assuntos
Síndrome Coronariana Aguda/enzimologia , Plaquetas/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Ativação Plaquetária/fisiologia , Análise de Variância , Estudos de Casos e Controles , Quimiocina CCL2/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosfolipases A2 Secretórias/metabolismo , Inibidor Tecidual de Metaloproteinase-2/metabolismo
18.
Biochem Pharmacol ; 206: 115297, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36243098

RESUMO

Platelets are the main effectors of the thrombotic events occurring at a ruptured atherosclerotic plaque and therefore antiplatelet agents are the mainstay of antithrombotic treatment for the prevention of myocardial infarction, atherotrombotic ischemic stroke and critical limb ischemia due to the thrombotic occlusion of the peripheral arteries. Despite great progress in antiplatelet agents over the last two decades, a number of important unmet medical needs still remain, like insufficient efficacy and a high incidence of hemorrhagic complications. Advances in the knowledge of the molecular mechanisms regulating platelet participation in hemostasis and in thrombosis and progress in pharmaceutical design have allowed to identify new drugs for established antiplatelet targets and novel targets for the development of new agents. Among the latter, several innovative approaches have already proceeded to clinical testing, like GPVI antagonism, PAR4 antagonism, PI3K inhibition, and some preliminary results seem promising. Here we review the pharmacologic approaches to platelet inhibition currently available and in development for their effects on platelet activation in vitro and on thrombosis in animal models and in humans. An ideal antithrombotic agent should selectively target events crucial for pathological thrombus formation without affecting hemostasis, an objective so far not achieved: if one or more of the novel agents in development will reach this goal this will represent a great step forward in the prevention of ischemic cardiovascular events.


Assuntos
Inibidores da Agregação Plaquetária , Trombose , Animais , Humanos , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Ativação Plaquetária , Plaquetas , Trombose/tratamento farmacológico , Trombose/prevenção & controle , Fibrinolíticos/farmacologia , Fibrinolíticos/uso terapêutico
19.
Am J Physiol Heart Circ Physiol ; 300(6): H2072-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21378140

RESUMO

Activated platelets express CD40L on their plasma membrane and release the soluble fragment sCD40L. The interaction between platelet surface CD40L and endothelial cell CD40 leads to the activation of endothelium contributing to atherothrombosis. Few studies have directly demonstrated an increased expression of platelet CD40L in conditions of in vivo platelet activation in humans, and no data are available on its relevance for endothelial activation. We aimed to assess whether platelets activated in vivo at a localized site of vascular injury in humans express CD40L and release sCD40L, whether the level of platelet CD40L expression attained in vivo is sufficient to induce endothelial activation, and whether platelet CD40L expression is inhibited by aspirin intake. We used the skin-bleeding-time test as a model to study the interaction between platelets and a damaged vessel wall by measuring CD40L in the blood emerging from a skin wound in vivo in healthy volunteers. In some experiments, shed blood was analyzed before and 1 h after the intake of 500 mg of aspirin. Platelets from the bleeding-time blood express CD40L and release soluble sCD40L, in a time-dependent way. In vivo platelet CD40L expression was mild but sufficient to induce VCAM-1 expression and IL-8 secretion in coincubation experiments with cultured human endothelial cells. Moreover, platelets recovered from the bleeding-time blood activated endothelial cells; an anti-CD40L antibody blocked this effect. On the contrary, the amount of sCD40L released by activated platelets at a localized site of vascular injury did not reach the concentrations required to induce endothelial cell activation. Soluble monocyte chemoattractant protein-1, a marker of endothelium activation, was increased in shed blood and correlated with platelet CD40L expression. Aspirin intake did not inhibit CD40L expression by platelets in vivo. We concluded that CD40L expressed by platelets in vivo in humans upon contact with a damaged vessel wall activates endothelium; aspirin treatment does not inhibit this mechanism.


Assuntos
Aspirina/farmacologia , Plaquetas/citologia , Plaquetas/metabolismo , Ligante de CD40/metabolismo , Comunicação Celular/fisiologia , Endotélio Vascular/citologia , Endotélio Vascular/lesões , Adulto , Aspirina/uso terapêutico , Aterosclerose/fisiopatologia , Aterosclerose/prevenção & controle , Plaquetas/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/metabolismo , Endotélio Vascular/fisiopatologia , Feminino , Humanos , Inflamação/fisiopatologia , Interleucina-8/metabolismo , Masculino , Pessoa de Meia-Idade , Modelos Cardiovasculares , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Pele/lesões , Molécula 1 de Adesão de Célula Vascular/metabolismo
20.
Br J Clin Pharmacol ; 72(4): 634-46, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21649691

RESUMO

Inhibition of platelet aggregation can be achieved either by the blockade of membrane receptors or by interaction with intracellular signalling pathways. Cyclic adenosine 3',5'-monophosphate (cAMP) and cyclic guanosine 3',5'-monophosphate (cGMP) are two critical intracellular second messengers provided with strong inhibitory activity on fundamental platelet functions. Phosphodiesterases (PDEs), by catalysing the hydrolysis of cAMP and cGMP, limit the intracellular levels of cyclic nucleotides, thus regulating platelet function. The inhibition of PDEs may therefore exert a strong platelet inhibitory effect. Platelets possess three PDE isoforms (PDE2, PDE3 and PDE5), with different selectivity for cAMP and cGMP. Several nonselective or isoenzyme-selective PDE inhibitors have been developed, and some of them have entered clinical use as antiplatelet agents. This review focuses on the effect of PDE2, PDE3 and PDE5 inhibitors on platelet function and on the evidence for an antithrombotic action of some of them, and in particular of dipyridamole and cilostazol.


Assuntos
Plaquetas/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Trombose/prevenção & controle , Cilostazol , Ensaios Clínicos como Assunto , Dipiridamol/farmacologia , Humanos , Tetrazóis/farmacologia , Trombose/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA