Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Sci Rep ; 10(1): 11389, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647264

RESUMO

Patients diagnosed with pseudohypoparathyroidism type Ia (PHP Ia) suffer from hormonal resistance and abnormal postural features, in a condition classified as Albright hereditary osteodystrophy (AHO) syndrome. This syndrome is linked to a maternally inherited mutation in the GNAS complex locus, encoding for the GTPase subunit Gsα. Here, we investigated how platelet phenotype and omics analysis can assist in the often difficult diagnosis. By coupling to the IP receptor, Gsα induces platelet inhibition via adenylyl cyclase and cAMP-dependent protein kinase A (PKA). In platelets from seven patients with suspected AHO, one of the largest cohorts examined, we studied the PKA-induced phenotypic changes. Five patients with a confirmed GNAS mutation, displayed impairments in Gsα-dependent VASP phosphorylation, aggregation, and microfluidic thrombus formation. Analysis of the platelet phosphoproteome revealed 2,516 phosphorylation sites, of which 453 were regulated by Gsα-PKA. Common changes in the patients were: (1) a joint panel of upregulated and downregulated phosphopeptides; (2) overall PKA dependency of the upregulated phosphopeptides; (3) links to key platelet function pathways. In one patient with GNAS mutation, diagnosed as non-AHO, the changes in platelet phosphoproteome were reversed. This combined approach thus revealed multiple phenotypic and molecular biomarkers to assist in the diagnosis of suspected PHP Ia.


Assuntos
Plaquetas/metabolismo , Cromograninas/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Iloprosta/farmacologia , Pseudo-Hipoparatireoidismo/diagnóstico , Biomarcadores/metabolismo , Plaquetas/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Criança , Cromograninas/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Resistência a Medicamentos/genética , Epigênese Genética , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Iloprosta/uso terapêutico , Masculino , Proteínas dos Microfilamentos/metabolismo , Mutação , Fosfoproteínas/metabolismo , Fosforilação , Agregação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/genética , Proteoma/metabolismo , Proteômica , Pseudo-Hipoparatireoidismo/sangue , Pseudo-Hipoparatireoidismo/genética
2.
Blood ; 132(24): e35-e46, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30275110

RESUMO

Antithrombotic therapies reduce cardiovascular diseases by preventing arterial thrombosis and thromboembolism, but at expense of increased bleeding risks. Arterial thrombosis studies using genetically modified mice have been invaluable for identification of new molecular targets. Because of low sample sizes and heterogeneity in approaches or methodologies, a formal meta-analysis to compare studies of mice with single-gene defects encountered major limitations. To overcome these, we developed a novel synthesis approach to quantitatively scale 1514 published studies of arterial thrombus formation (in vivo and in vitro), thromboembolism, and tail-bleeding of genetically modified mice. Using a newly defined consistency parameter (CP), indicating the strength of published data, comparisons were made of 431 mouse genes, of which 17 consistently contributed to thrombus formation without affecting hemostasis. Ranking analysis indicated high correlations between collagen-dependent thrombosis models in vivo (FeCl3 injury or ligation/compression) and in vitro. Integration of scores and CP values resulted in a network of protein interactions in thrombosis and hemostasis (PITH), which was combined with databases of genetically linked human bleeding and thrombotic disorders. The network contained 2946 nodes linked to modifying genes of thrombus formation, mostly with expression in megakaryocytes. Reactome pathway analysis and network characteristics revealed multiple novel genes with potential contribution to thrombosis/hemostasis. Studies with additional knockout mice revealed that 4 of 8 (Apoe, Fpr2, Ifnar1, Vps13a) new genes were modifying in thrombus formation. The PITH network further: (i) revealed a high similarity of murine and human hemostatic and thrombotic processes and (ii) identified multiple new candidate proteins regulating these processes.


Assuntos
Hemorragia , Trombose , Animais , Modelos Animais de Doenças , Hemorragia/genética , Hemorragia/metabolismo , Hemorragia/patologia , Humanos , Camundongos , Camundongos Knockout , Trombose/genética , Trombose/metabolismo , Trombose/patologia
3.
Blood Adv ; 2(18): 2320-2331, 2018 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-30232085

RESUMO

The platelet receptors glycoprotein Ibα (GPIbα) and GPVI are known to be cleaved by members of a disintegrin and metalloprotease (ADAM) family (ADAM10 and ADAM17), but the mechanisms and consequences of this shedding are not well understood. Our results revealed that (1) glycoprotein shedding is confined to distinct platelet populations showing near-complete shedding, (2) the heterogeneity between (non)shed platelets is independent of agonist type but coincides with exposure of phosphatidylserine (PS), and (3) distinct pathways of shedding are induced by elevated Ca2+, low Ca2+ protein kinase C (PKC), or apoptotic activation. Furthermore, we found that receptor shedding reduces binding of von Willebrand factor, enhances binding of coagulation factors, and augments fibrin formation. In response to Ca2+-increasing agents, shedding of GPIbα was abolished by ADAM10/17 inhibition but not by blockage of calpain. Stimulation of PKC induced shedding of only GPIbα, which was annulled by kinase inhibition. The proapoptotic agent ABT-737 induced shedding, which was caspase dependent. In Scott syndrome platelets that are deficient in Ca2+-dependent PS exposure, shedding occurred normally, indicating that PS exposure is not a prerequisite for ADAM activity. In whole-blood thrombus formation, ADAM-dependent glycoprotein shedding enhanced thrombin generation and fibrin formation. Together, these findings indicate that 2 major activation pathways can evoke ADAM-mediated glycoprotein shedding in distinct platelet populations and that shedding modulates platelet function from less adhesive to more procoagulant.


Assuntos
Plaquetas/metabolismo , Glicoproteínas/metabolismo , Ativação Plaquetária , Proteínas ADAM/metabolismo , Biomarcadores , Plaquetas/efeitos dos fármacos , Cálcio/metabolismo , Caspases/metabolismo , Citometria de Fluxo , Humanos , Ionomicina/farmacologia , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Transdução de Sinais , Trombina/metabolismo , Trombose/metabolismo
4.
Thromb Haemost ; 118(3): 502-513, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29452445

RESUMO

Tissue factor pathway inhibitor-alpha (TFPI-α) is a Kunitz-type serine protease inhibitor, which suppresses coagulation by inhibiting the tissue factor (TF)/factor VIIa complex as well as factor Xa. In static plasma-phospholipid systems, TFPI-α thus suppresses both factor Xa and thrombin generation. In this article, we used a microfluidics approach to investigate how TFPI-α regulates fibrin clot formation in platelet thrombi at low wall shear rate. We therefore hypothesized that the anticoagulant effect of TFPI-α in plasma is a function of the local procoagulant strength-defined as the magnitude of thrombin generation under flow, due to local activities of TF/factor VIIa and factor Xa. To test this hypothesis, we modulated local coagulation by microspot coating of flow channels with 0 to 100 pM TF/collagen, or by using blood from patients with haemophilia A or B. For blood or plasma from healthy subjects, blocking of TFPI-α enhanced fibrin formation, extending from a platelet thrombus, under flow only at <2 pM coated TF. This enhancement was paralleled by an increased thrombin generation. For mouse plasma, genetic deficiency in TFPI enhanced fibrin formation under flow also at 0 pM TF microspots. On the other hand, using blood from haemophilia A or B patients, TFPI-α antagonism markedly enhanced fibrin formation at microspots with up to 100 pM coated TF. We conclude that, under flow, TFPI-α is capable to antagonize fibrin formation in a manner dependent on and restricted by local TF/factor VIIa and factor Xa activities.


Assuntos
Plaquetas/efeitos dos fármacos , Coagulantes/química , Fator VIIa/química , Fator Xa/química , Fibrina/química , Lipoproteínas/química , Animais , Anticoagulantes/química , Coagulação Sanguínea , Plaquetas/citologia , Colágeno/química , Cruzamentos Genéticos , Feminino , Voluntários Saudáveis , Hemofilia A/sangue , Hemofilia B/sangue , Heterozigoto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Perfusão , Tromboplastina/química , Trombose
5.
Arterioscler Thromb Vasc Biol ; 35(12): 2554-61, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26471268

RESUMO

OBJECTIVE: Platelets are increasingly implicated in processes beyond hemostasis and thrombosis, such as vascular remodeling. Members of the matrix metalloproteinase (MMP) family not only remodel the extracellular matrix but also modulate platelet function. Here, we made a systematic comparison of the roles of MMP family members in acute thrombus formation under flow conditions and assessed platelet-dependent collagenolytic activity over time. APPROACH AND RESULTS: Pharmacological inhibition of MMP-1 or MMP-2 (human) or deficiency in MMP-2 (mouse) suppressed collagen-dependent platelet activation and thrombus formation under flow, whereas MMP-9 inhibition/deficiency stimulated these processes. The absence of MMP-3 was without effect. Interestingly, MMP-14 inhibition led to the formation of larger thrombi, which occurred independently of its capacity to activate MMP-2. Platelet thrombi exerted local collagenolytic activity capable of cleaving immobilized dye-quenched collagen and fibrillar collagen fibers within hours, with loss of the majority of the platelet adhesive properties of collagen as a consequence. This collagenolytic activity was redundantly mediated by platelet-associated MMP-1, MMP-2, MMP-9, and MMP-14 but occurred independently of platelet α-granule release (Nbeal2(-/-) mice). The latter was in line with subcellular localization experiments, which indicated a granular distribution of MMP-1 and MMP-2 in platelets, distinct from α-granules. Whereas MMP-9 protein could not be detected inside platelets, activated platelets did bind plasma-derived MMP-9 to their plasma membrane. Overall, platelet MMP activity was predominantly membrane-associated and influenced by platelet activation status. CONCLUSIONS: Platelet-associated MMP-1, MMP-2, MMP-9, and MMP-14 differentially modulate acute thrombus formation and at later time points limit thrombus formation by exerting collagenolytic activity.


Assuntos
Plaquetas/enzimologia , Colágeno/metabolismo , Colagenases/sangue , Trombose/enzimologia , Animais , Plaquetas/efeitos dos fármacos , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Colagenases/deficiência , Colagenases/genética , Modelos Animais de Doenças , Humanos , Inibidores de Metaloproteinases de Matriz/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativação Plaquetária , Glicoproteínas da Membrana de Plaquetas/metabolismo , Proteólise , Trombose/sangue , Trombose/genética , Fatores de Tempo
6.
Arterioscler Thromb Vasc Biol ; 34(8): 1674-80, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24855058

RESUMO

OBJECTIVE: Atherothrombosis is the main cause of myocardial infarction and ischemic stroke. Although the extrinsic (tissue factor-factor VIIa [FVIIa]) pathway is considered as a major trigger of coagulation in atherothrombosis, the role of the intrinsic coagulation pathway via coagulation FXII herein is unknown. Here, we studied the roles of the extrinsic and intrinsic coagulation pathways in thrombus formation on atherosclerotic plaques both in vivo and ex vivo. APPROACH AND RESULTS: Plaque rupture after ultrasound treatment evoked immediate formation of subocclusive thrombi in the carotid arteries of Apoe(-/-) mice, which became unstable in the presence of structurally different FXIIa inhibitors. In contrast, inhibition of FVIIa reduced thrombus size at a more initial stage without affecting embolization. Genetic deficiency in FXII (human and mouse) or FXI (mouse) reduced ex vivo whole-blood thrombus and fibrin formation on immobilized plaque homogenates. Localization studies by confocal microscopy indicated that FXIIa bound to thrombi and fibrin particularly in luminal-exposed thrombus areas. CONCLUSIONS: The FVIIa- and FXIIa-triggered coagulation pathways have distinct but complementary roles in atherothrombus formation. The tissue factor-FVIIa pathway contributes to initial thrombus buildup, whereas FXIIa bound to thrombi ensures thrombus stability.


Assuntos
Doenças da Aorta/complicações , Aterosclerose/complicações , Coagulação Sanguínea , Plaquetas/metabolismo , Doenças das Artérias Carótidas/complicações , Fator XII/metabolismo , Placa Aterosclerótica , Trombose/etiologia , Animais , Aorta Torácica/metabolismo , Aorta Torácica/patologia , Doenças da Aorta/sangue , Doenças da Aorta/genética , Doenças da Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/sangue , Aterosclerose/genética , Aterosclerose/patologia , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Doenças das Artérias Carótidas/sangue , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/patologia , Colesterol na Dieta , Modelos Animais de Doenças , Fator VIIa/metabolismo , Fator XI/metabolismo , Fator XII/genética , Deficiência do Fator XII/sangue , Deficiência do Fator XII/genética , Fator XIIa/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ruptura Espontânea , Tromboplastina/metabolismo , Trombose/sangue , Trombose/genética , Trombose/patologia , Fatores de Tempo
7.
Arterioscler Thromb Vasc Biol ; 34(1): 61-71, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24233488

RESUMO

OBJECTIVE: Platelet activation is essential for primary hemostasis and acute thrombotic vascular occlusions. On activation, platelets release their prothrombotic granules and expose phosphatidylserine, thus fostering thrombin generation and thrombus formation. In other cell types, both degranulation and phosphatidylserine exposure are modified by sphingomyelinase-dependent formation of ceramide. The present study thus explored whether acid sphingomyelinase participates in the regulation of platelet secretion, phosphatidylserine exposure, and thrombus formation. APPROACH AND RESULTS: Collagen-related peptide-induced or thrombin-induced ATP release and P-selectin exposure were significantly blunted in platelets from Asm-deficient mice (Smpd1(-/-)) when compared with platelets from wild-type mice (Smpd1(+/+)). Moreover, phosphatidylserine exposure and thrombin generation were significantly less pronounced in Smpd1(-/-) platelets than in Smpd1(+/+) platelets. In contrast, platelet integrin αIIbß3 activation and aggregation, as well as activation-dependent Ca(2+) flux, were not significantly different between Smpd1(-/-) and Smpd1(+/+) platelets. In vitro thrombus formation at shear rates of 1700 s(-1) and in vivo thrombus formation after FeCl3 injury were significantly blunted in Smpd1(-/-) mice while bleeding time was unaffected. Asm-deficient platelets showed significantly reduced activation-dependent ceramide formation, whereas exogenous ceramide rescued diminished platelet secretion and thrombus formation caused by Asm deficiency. Treatment of Smpd1(+/+) platelets with bacterial sphingomyelinase (0.01 U/mL) increased, whereas treatment with functional acid sphingomyelinase-inhibitors, amitriptyline or fluoxetine (5 µmol/L), blunted activation-dependent platelet degranulation, phosphatidylserine exposure, and thrombus formation. Impaired degranulation and thrombus formation of Smpd1(-/-) platelets were again overcome by exogenous bacterial sphingomyelinase. CONCLUSIONS: Acid sphingomyelinase is a completely novel element in the regulation of platelet plasma membrane properties, secretion, and thrombus formation.


Assuntos
Plaquetas/enzimologia , Degranulação Celular , Membrana Celular/enzimologia , Ativação Plaquetária , Esfingomielina Fosfodiesterase/sangue , Trombose/enzimologia , Trifosfato de Adenosina/sangue , Animais , Plaquetas/efeitos dos fármacos , Cálcio/sangue , Degranulação Celular/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Ceramidas/sangue , Cloretos , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Compostos Férricos , Fibrinolíticos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Selectina-P/sangue , Fosfatidilserinas/sangue , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Esfingomielina Fosfodiesterase/antagonistas & inibidores , Esfingomielina Fosfodiesterase/deficiência , Esfingomielina Fosfodiesterase/genética , Trombina/metabolismo , Trombose/sangue , Trombose/induzido quimicamente , Trombose/genética , Trombose/prevenção & controle , Fatores de Tempo
8.
PLoS One ; 8(5): e64112, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23724024

RESUMO

BACKGROUND: Recurrent bleeding can complicate the treatment of thrombosis patients with vitamin K antagonists (VKA), even at a well-regulated level of anticoagulation. In this proof-of-principle study, we investigated whether alterations in platelet function or von Willebrand factor (vWf) contribute to a bleeding phenotype in these patients. METHODS: In this case-control study 33 well-regulated patients without bleeding events (controls) and 33 patients with recurrent bleeding (cases) were retrospectively included. Thrombin generation and vWf were determined in plasma. Platelet function was assessed by light transmission aggregometry and flow cytometry using a validated panel of agonists. RESULTS: Thrombin generation was similarly reduced in controls and cases, in comparison to normal plasma. Plasma vWf level was above the normal range in 85% of controls and 67% of the cases. vWf activity was similarly increased in all patients in comparison to healthy volunteers. Platelet aggregation was in the normal range for almost all patients irrespective of the type of agonist. However, in response to a low collagen dose, platelets from 21% of controls and 27% of cases showed diminished responses. Agonist-induced secretion of alpha- and dense-granules or integrin αIIbß3 activation were affected in platelets from neither controls nor cases. CONCLUSION: Recurrent bleeding in well-controlled patients on VKA therapy is not explained by anti-hemostatic changes in platelet or vWf function.


Assuntos
Transtornos Plaquetários/induzido quimicamente , Hemorragia/induzido quimicamente , Trombose/tratamento farmacológico , Vitamina K/antagonistas & inibidores , Idoso , Plaquetas , Estudos de Casos e Controles , Feminino , Hemorragia/sangue , Humanos , Integrinas/metabolismo , Masculino , Agregação Plaquetária , Recidiva , Trombina/metabolismo , Trombose/sangue , Fator de von Willebrand/metabolismo
9.
Thromb Res ; 131(5): 411-7, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23347681

RESUMO

INTRODUCTION: Consumption of n-3 polyunsaturated fatty acids (PUFA) and antioxidant polyphenols is considered to decline the risk of cardiovascular disease. MATERIALS AND METHODS: To provide an explanation for this cardioprotective effect, we performed an intervention study with proatherogenic Apoe(-/-) mice which were fed during eight weeks with a high fat diet supplemented with either walnuts (rich in n-3 PUFA and antioxidant compounds), walnut oil (with n-3 PUFA only) or sunflower oil as a control (12 mice per group). RESULTS: Feeding walnuts, but not walnut oil, caused a 55% reduction in atherosclerotic plaque development in the aortic arch in comparison to the control diet. This was associated with reduced staining of plaques for CD36, a scavenger receptor expressed by macrophages. Feeding mice with walnuts also lowered plasma levels of triglycerides, cholesterol and prothrombin with 36%, 23% and 21 %, respectively, compared to control diet. In addition, accumulation of lipids in the liver was decreased, while plasma antioxidant capacity was increased. On the other hand, feeding mice with walnut oil did not provoke significant changes in these parameters in comparison to the control diet. Platelet activation and thrombus formation under flow remained unchanged with either diet. CONCLUSIONS: In Apoe(-/-) mice on high fat diet, intake of dietary walnut (but not walnut oil) beneficially influences lipid metabolism and atherosclerotic plaque development, with no more than limited effects on platelet and coagulation function.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Fatores de Coagulação Sanguínea/metabolismo , Juglans , Animais , Apolipoproteínas E/sangue , Doenças Cardiovasculares/prevenção & controle , Imuno-Histoquímica , Metabolismo dos Lipídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória
10.
Blood ; 121(10): 1850-7, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23303820

RESUMO

Scott syndrome, a bleeding disorder caused by defective phospholipid scrambling, has been associated with mutations in the TMEM16F gene. The role of TMEM16F in apoptosis- or agonist-induced phosphatidylserine (PS) exposure was studied in platelets from a Scott syndrome patient and control subjects. Whereas stimulation of control platelets with the BH3-mimetic ABT737 resulted in 2 distinct fractions with moderate and high PS exposure, the high PS-exposing fraction was markedly delayed in Scott platelets. High, but not moderate, PS exposure in platelets was suppressed by chelation of intracellular Ca(2+), whereas caspase inhibition completely abolished ABT737-induced PS exposure in both Scott and control platelets. On the other hand, high PS exposure induced by the Ca(2+)-mobilizing agonists convulxin/thrombin fully relied on mitochondrial depolarization and was virtually absent in Scott platelets. Finally, PS exposure induced by collagen/thrombin was partly affected in Scott platelets, and the residual PS positive fraction was insensitive to inhibition of caspases or mitochondrial depolarization. In conclusion, TMEM16F is not required for, but enhances, caspase-dependent PS exposure; convulxin-/thrombin-induced PS exposure is entirely dependent on TMEM16F, whereas collagen/thrombin-induced PS exposure results from 2 distinct pathways, one of which involves mitochondrial depolarization and is mediated by TMEM16F.


Assuntos
Apoptose , Transtornos da Coagulação Sanguínea/patologia , Plaquetas/patologia , Cálcio/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Ativação Plaquetária , Anoctaminas , Transtornos da Coagulação Sanguínea/metabolismo , Plaquetas/metabolismo , Estudos de Casos e Controles , Caspases/metabolismo , Venenos de Crotalídeos/farmacologia , Ciclofilinas/metabolismo , Citometria de Fluxo , Hemostáticos/farmacologia , Humanos , Lectinas Tipo C , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Trombina/farmacologia
11.
Cell Mol Life Sci ; 69(20): 3481-92, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22669259

RESUMO

The fibrin(ogen) receptor, integrin α(IIb)ß(3), has a well-established role in platelet spreading, aggregation and clot retraction. How α(IIb)ß(3) contributes to platelet-dependent coagulation is less well resolved. Here, we demonstrate that the potent suppressing effect of clinically used α(IIb)ß(3) blockers on tissue factor-induced thrombin generation is linked to diminished platelet Ca(2+) responses and phosphatidylserine (PS) exposure. The same blockers suppress these responses in platelets stimulated with collagen and thrombin receptor agonists, whereas added fibrinogen potentiates these responses. In platelets spreading on fibrinogen, outside-in α(IIb)ß(3) signaling similarly enhances thrombin-induced Ca(2+) rises and PS exposure. These responses are reduced in α(IIb)ß(3)-deficient platelets from patients with Glanzmann's thrombasthenia. Furthermore, the contribution of α(IIb)ß(3) to tissue factor-induced platelet Ca(2+) rises, PS exposure and thrombin generation in plasma are fully dependent on Syk kinase activity. Tyrosine phosphorylation analysis confirms a key role of Syk activation, which is largely but not exclusively dependent on α(IIb)ß(3) activation. It is concluded that the majority of tissue factor-induced procoagulant activity of platelets relies on Syk activation and ensuing Ca(2+) signal generation, and furthermore that a considerable part of Syk activation relies on α(IIb)ß(3) signaling. These results hence point to a novel role of Syk in integrin-dependent thrombin generation.


Assuntos
Plaquetas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Trombastenia/sangue , Trombina/metabolismo , Tromboplastina/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Western Blotting , Fibrinogênio/metabolismo , Citometria de Fluxo , Humanos , Fosfosserina/metabolismo , Agregação Plaquetária/efeitos dos fármacos , Transdução de Sinais , Quinase Syk
12.
Arterioscler Thromb Vasc Biol ; 32(7): 1717-23, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22580895

RESUMO

OBJECTIVE: Platelet Orai1 channels mediate store-operated Ca(2+) entry (SOCE), which is required for procoagulant activity and arterial thrombus formation. Pharmacological blockage of these channels may provide a novel way of antithrombotic therapy. Therefore, the thromboprotective effect of SOCE blockers directed against platelet Orai1 is determined. METHODS AND RESULTS: Candidate inhibitors were screened for their effects on SOCE in washed human platelets. Tested antagonists included the known compounds, SKF96365, 2-aminoethyl diphenylborate, and MRS1845 and the novel compounds, Synta66 and GSK-7975A. The potency of SOCE inhibition was in the order of Synta66>2-aminoethyl diphenylborate>GSK-7975A>SKF96365>MRS1845. The specificity of the first 3 compounds was verified with platelets from Orai1-deficient mice. Inhibitory activity on procoagulant activity and high-shear thrombus formation was assessed in plasma and whole blood. In the presence of plasma, all 3 compounds suppressed platelet responses and restrained thrombus formation under flow. Using a murine stroke model, arterial thrombus formation was provoked in vivo by transient middle cerebral artery occlusion. Postoperative administration of 2-aminoethyl diphenylborate markedly diminished brain infarct size. CONCLUSIONS: Plasma-soluble SOCE blockers such as 2-aminoethyl diphenylborate suppress platelet-dependent coagulation and thrombus formation. The platelet Orai1 channel is a novel target for preventing thrombotic events causing brain infarction.


Assuntos
Plaquetas/efeitos dos fármacos , Bloqueadores dos Canais de Cálcio/farmacologia , Fibrinolíticos/farmacologia , Animais , Compostos de Boro/farmacologia , Cálcio/metabolismo , Canais de Cálcio/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteína ORAI1 , Ativação Plaquetária/efeitos dos fármacos
13.
Thromb Haemost ; 106(6): 1179-88, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22071958

RESUMO

Platelet P2Y12 receptors play an important role in arterial thrombosis by stimulating thrombus growth. Both irreversibly (clopidogrel) and reversibly binding (ticagrelor, AZD6140) P2Y12 antagonists are clinically used for restricted periods, but possible differences in platelet function recovery after drug cessation have not been investigated. We treated WKY rats with a single, high dose of 200 mg/kg clopidogrel or 40 mg/kg ticagrelor. Blood was collected at different time points after treatment. Flow cytometry confirmed full platelet protection against ADP-induced αIIbß3 activation shortly after clopidogrel or ticagrelor treatment. At later time points after clopidogrel treatment, a subpopulation of juvenile platelets appeared that was fully responsive to ADP. Addition of ticagrelor to clopidogrel-treated blood reduced αIIbß3 activation of the unprotected platelets. In contrast, at later time points after ticagrelor treatment, all platelets gradually lost their protection against ADP activation. Perfusion experiments showed abolishment of thrombus formation shortly after clopidogrel or ticagrelor treatment. Thrombus formation on collagen was determined under high shear flow conditions. At later time points, large thrombi formed in the clopidogrel but not in the ticagrelor group, and unprotected, juvenile platelets preferentially incorporated into the formed thrombi. However, platelets from both groups were still similarly reduced in assays of whole blood aggregation. Conclusively, recovery of rat platelet function after ticagrelor differs mechanistically from that after clopidogrel. This difference is masked by conventional platelet aggregation methods, but is revealed by thrombus formation measurement under flow. Juvenile platelets formed at later time points after clopidogrel treatment promoted thrombus formation.


Assuntos
Adenosina/análogos & derivados , Plaquetas/metabolismo , Antagonistas do Receptor Purinérgico P2Y/administração & dosagem , Trombose/tratamento farmacológico , Ticlopidina/análogos & derivados , Adenosina/administração & dosagem , Adenosina/efeitos adversos , Difosfato de Adenosina/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/patologia , Clopidogrel , Citoproteção/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Ligação Proteica , Antagonistas do Receptor Purinérgico P2Y/efeitos adversos , Ratos , Ratos Endogâmicos WKY , Trombose/metabolismo , Trombose/patologia , Ticagrelor , Ticlopidina/administração & dosagem , Ticlopidina/efeitos adversos
14.
Blood ; 117(2): 651-60, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-21037087

RESUMO

A microscopic method was developed to study the role of platelets in fibrin formation. Perfusion of adhered platelets with plasma under coagulating conditions at a low shear rate (250(-1)) resulted in the assembly of a star-like fibrin network at the platelet surface. The focal fibrin formation on platelets was preceded by rises in cytosolic Ca(2+), morphologic changes, and phosphatidylserine exposure. Fibrin formation was slightly affected by α(IIb)ß(3) blockage, but it was greatly delayed and reduced by the following: inhibition of thrombin or platelet activation; interference in the binding of von Willebrand factor (VWF) to glycoprotein Ib/V/IX (GpIb-V-IX); plasma or blood from patients with type 1 von Willebrand disease; and plasma from mice deficient in VWF or the extracellular domain of GpIbα. In this process, the GpIb-binding A1 domain of VWF was similarly effective as full-length VWF. Prestimulation of platelets enhanced the formation of fibrin, which was abrogated by blockage of phosphatidylserine. Together, these results show that, in the presence of thrombin and low shear flow, VWF-induced activation of GpIb-V-IX triggers platelet procoagulant activity and anchorage of a star-like fibrin network. This process can be relevant in hemostasis and the manifestation of von Willebrand disease.


Assuntos
Coagulação Sanguínea/fisiologia , Fibrina/biossíntese , Ativação Plaquetária/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Fator de von Willebrand/metabolismo , Animais , Plaquetas/metabolismo , Humanos , Processamento de Imagem Assistida por Computador , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Resistência ao Cisalhamento , Doenças de von Willebrand/metabolismo , Doenças de von Willebrand/fisiopatologia
15.
J Biol Chem ; 285(31): 23629-38, 2010 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-20519511

RESUMO

In platelets, STIM1 has been recognized as the key regulatory protein in store-operated Ca(2+) entry (SOCE) with Orai1 as principal Ca(2+) entry channel. Both proteins contribute to collagen-dependent arterial thrombosis in mice in vivo. It is unclear whether STIM2 is involved. A key platelet response relying on Ca(2+) entry is the surface exposure of phosphatidylserine (PS), which accomplishes platelet procoagulant activity. We studied this response in mouse platelets deficient in STIM1, STIM2, or Orai1. Upon high shear flow of blood over collagen, Stim1(-/-) and Orai1(-/-) platelets had greatly impaired glycoprotein (GP) VI-dependent Ca(2+) signals, and they were deficient in PS exposure and thrombus formation. In contrast, Stim2(-/-) platelets reacted normally. Upon blood flow in the presence of thrombin generation and coagulation, Ca(2+) signals of Stim1(-/-) and Orai1(-/-) platelets were partly reduced, whereas the PS exposure and formation of fibrin-rich thrombi were normalized. Washed Stim1(-/-) and Orai1(-/-) platelets were deficient in GPVI-induced PS exposure and prothrombinase activity, but not when thrombin was present as co-agonist. Markedly, SKF96365, a blocker of (receptor-operated) Ca(2+) entry, inhibited Ca(2+) and procoagulant responses even in Stim1(-/-) and Orai1(-/-) platelets. These data show for the first time that: (i) STIM1 and Orai1 jointly contribute to GPVI-induced SOCE, procoagulant activity, and thrombus formation; (ii) a compensating Ca(2+) entry pathway is effective in the additional presence of thrombin; (iii) platelets contain two mechanisms of Ca(2+) entry and PS exposure, only one relying on STIM1-Orai1 interaction.


Assuntos
Canais de Cálcio/metabolismo , Coagulantes/metabolismo , Glicoproteínas de Membrana/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Trombina/metabolismo , Trombose/metabolismo , Animais , Plaquetas/metabolismo , Cálcio/química , Cálcio/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Proteína ORAI1 , Molécula 1 de Interação Estromal
16.
PLoS One ; 5(4): e10130, 2010 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-20405028

RESUMO

BACKGROUND: In most models of experimental thrombosis, healthy blood vessels are damaged. This results in the formation of a platelet thrombus that is stabilized by ADP signaling via P2Y(12) receptors. However, such models do not predict involvement of P2Y(12) in the clinically relevant situation of thrombosis upon rupture of atherosclerotic plaques. We investigated the role of P2Y(12) in thrombus formation on (collagen-containing) atherosclerotic plaques in vitro and in vivo, by using a novel mouse model of atherothrombosis. METHODOLOGY: Plaques in the carotid arteries from Apoe(-/-) mice were acutely ruptured by ultrasound treatment, and the thrombotic process was monitored via intravital fluorescence microscopy. Thrombus formation in vitro was assessed in mouse and human blood perfused over collagen or plaque material under variable conditions of shear rate and coagulation. Effects of two reversible P2Y(12) blockers, ticagrelor (AZD6140) and cangrelor (AR-C69931MX), were investigated. PRINCIPAL FINDINGS: Acute plaque rupture by ultrasound treatment provoked rapid formation of non-occlusive thrombi, which were smaller in size and unstable in the presence of P2Y(12) blockers. In vitro, when mouse or human blood was perfused over collagen or atherosclerotic plaque material, blockage or deficiency of P2Y(12) reduced the thrombi and increased embolization events. These P2Y(12) effects were present at shear rates >500 s(-1), and they persisted in the presence of coagulation. P2Y(12)-dependent thrombus stabilization was accompanied by increased fibrin(ogen) binding. CONCLUSIONS/SIGNIFICANCE: Platelet P2Y(12) receptors play a crucial role in the stabilization of thrombi formed on atherosclerotic plaques. This P2Y(12) function is restricted to high shear flow conditions, and is preserved in the presence of coagulation.


Assuntos
Aterosclerose/complicações , Embolia/etiologia , Receptores Purinérgicos P2Y12/fisiologia , Trombose/patologia , Animais , Aterosclerose/patologia , Coagulação Sanguínea , Trombose das Artérias Carótidas/patologia , Modelos Animais de Doenças , Hemorreologia , Humanos , Camundongos , Camundongos Knockout , Perfusão
17.
J Biol Chem ; 284(49): 33750-62, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19815551

RESUMO

Platelets are activated by adhesion to vascular collagen via the immunoglobulin receptor, glycoprotein VI (GPVI). This causes potent signaling toward activation of phospholipase Cgamma2, which bears similarity to the signaling pathway evoked by T- and B-cell receptors. Phosphoinositide 3-kinase (PI3K) plays an important role in collagen-induced platelet activation, because this activity modulates the autocrine effects of secreted ADP. Here, we identified the PI3K isoforms directly downstream of GPVI in human and mouse platelets and determined their role in GPVI-dependent thrombus formation. The targeting of platelet PI3Kalpha or -beta strongly and selectively suppressed GPVI-induced Ca(2+) mobilization and inositol 1,4,5-triphosphate production, thus demonstrating enhancement of phospholipase Cgamma2 by PI3Kalpha/beta. That PI3Kalpha and -beta have a non-redundant function in GPVI-induced platelet activation and thrombus formation was concluded from measurements of: (i) serine phosphorylation of Akt, (ii) dense granule secretion, (iii) intracellular Ca(2+) increases and surface expression of phosphatidylserine under flow, and (iv) thrombus formation, under conditions where PI3Kalpha/beta was blocked or p85alpha was deficient. In contrast, GPVI-induced platelet activation was insensitive to inhibition or deficiency of PI3Kdelta or -gamma. Furthermore, PI3Kalpha/beta, but not PI3Kgamma, contributed to GPVI-induced Rap1b activation and, surprisingly, also to Rap1b-independent platelet activation via GPVI. Together, these findings demonstrate that both PI3Kalpha and -beta isoforms are required for full GPVI-dependent platelet Ca(2+) signaling and thrombus formation, partly independently of Rap1b. This provides a new mechanistic explanation for the anti-thrombotic effect of PI3K inhibition and makes PI3Kalpha an interesting new target for anti-platelet therapy.


Assuntos
Plaquetas/metabolismo , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Trombose/patologia , Animais , Cálcio/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/genética , Ativação Plaquetária , Agregação Plaquetária , Isoformas de Proteínas , Transdução de Sinais , Trombose/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 28(11): 2023-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18757293

RESUMO

OBJECTIVE: In hyperlipidemia, dietary fish oil containing n-3 polyunsaturated fatty acids (PUFA) provokes plasma triacylglycerol lowering and hypocoagulant activity. Using APOE2 knock-in mice, the relation of these fish-oil effects with altered gene expression was investigated. METHODS AND RESULTS: Male APOE2 knock-in mice, fed regular low-fat diet, had elevated plasma levels of triacylglycerol and coagulation factors. Plasma lipids and (anti)coagulant factors reduced on feeding the mice with fish oil (n-3 PUFA) or, to a lesser degree, with sunflower seed oil (n-6 PUFA). The fish-oil diet provoked a 40% reduction in thrombin generation. Microarray (Affymetrix) and single-gene expression analysis of mouse livers showed that fish oil induced: (1) upregulation of genes contributing to lipid degradation and oxidation; (2) downregulation of genes of gamma-glutamyl carboxylase and of transcription factors implicated in lipid synthesis; (3) unchanged expression of coagulation factor genes. After fish-oil diet, vitamin K-dependent coagulation factors accumulated in periportal areas of the liver; prothrombin was partly retained in uncarboxylated form. Only part of the changes in gene expression were different from the effects of sunflower seed oil diet. CONCLUSIONS: The hypocoagulant effect of n-3 PUFA is not caused by reduced hepatic synthesis of coagulation factors, but rather results from retention of uncarboxylated coagulation factors. In contrast, the lipid-lowering effect of n-3 PUFA links to altered expression of genes that regulate transcription and fatty acid metabolism.


Assuntos
Anticoagulantes/farmacologia , Apolipoproteína E2/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Ácidos Graxos Ômega-3/farmacologia , Hiperlipidemias/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Animais , Anticoagulantes/administração & dosagem , Apolipoproteína E2/genética , Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/metabolismo , Dieta , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/administração & dosagem , Ácidos Graxos Ômega-6/farmacologia , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Hiperlipidemias/sangue , Hiperlipidemias/genética , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Análise de Sequência com Séries de Oligonucleotídeos , Óleos de Plantas/farmacologia , RNA Mensageiro/metabolismo , Óleo de Girassol , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos , Triglicerídeos/sangue
19.
Transfusion ; 48(11): 2384-94, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18673348

RESUMO

BACKGROUND: Bleeding upon major surgery or severe trauma is treated by transfusion with crystalloids, colloids, or plasma. This treatment, however, can lead to dilutional coagulopathy and impaired hemostasis. We investigated the suitability of two integrative coagulation tests to measure the hemostatic activity of diluted plasma. STUDY DESIGN AND METHODS: Plasma from healthy donors was diluted in vitro with saline or colloid (venofundin or gelofusin). Coagulant activity in response to tissue factor was monitored by calibrated automated thrombin (CAT) generation and rotational thromboelastography (TEG), detecting formation of elastic fibrin clots. Plasma from patients receiving fluid infusion during coronary artery bypass grafting (CABG) was analyzed with the same assays. RESULTS: Optimal activity of CAT and TEG assays required the presence of 10 pmol per L tissue factor and 4 micromol per L phospholipid vesicles or 100 x 10(9) platelets (PLTs) per L. Strikingly, thrombin generation and clot formation became impaired at a higher extent of dilution with PLTs present (< or =40% plasma) than with phospholipid vesicles present (< or =60% plasma). Colloids aggravated the dilution effect on clot formation, but FFP antagonized the dilution effect on thrombin and clot formation. In contrast, fibrinogen and Factor (F)XIII only restored the impaired clot formation. In plasma samples from patients undergoing CABG, CAT and TEG assay variables were altered to an extent corresponding with the volume of fluid infusion. CONCLUSION: Thrombin generation and clot formation are reduced at a plasma dilution of more than 40 percent. In either process, PLTs can partly compensate for the dilution effect. In vitro dilution with colloids impaired fibrin clot elasticity compared to saline.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/fisiologia , Plasma , Tromboelastografia , Trombina/biossíntese , Tromboplastina/farmacologia , Anticoagulantes/efeitos adversos , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico , Coloides/efeitos adversos , Coloides/farmacologia , Coloides/uso terapêutico , Ponte de Artéria Coronária , Soluções Cristaloides , Gelatina/farmacologia , Gelatina/uso terapêutico , Transtornos Hemorrágicos/sangue , Transtornos Hemorrágicos/etiologia , Transtornos Hemorrágicos/prevenção & controle , Heparina/efeitos adversos , Heparina/farmacologia , Heparina/uso terapêutico , Humanos , Derivados de Hidroxietil Amido/efeitos adversos , Derivados de Hidroxietil Amido/farmacologia , Derivados de Hidroxietil Amido/uso terapêutico , Soluções Isotônicas/efeitos adversos , Soluções Isotônicas/farmacologia , Soluções Isotônicas/uso terapêutico , Concentração Osmolar , Substitutos do Plasma/efeitos adversos , Substitutos do Plasma/farmacologia , Protaminas/farmacologia , Protaminas/uso terapêutico , Proteínas Recombinantes/farmacologia , Succinatos/farmacologia , Succinatos/uso terapêutico
20.
FEBS J ; 275(2): 371-85, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18081863

RESUMO

During thrombus formation, thrombin, which is abundantly present at sites of vascular injury, activates platelets in part via autocrine-produced ADP. We investigated the signaling pathways by which thrombin and ADP in synergy induced platelet Ca(2+) elevation and procoagulant activity, and we monitored the consequences for the coagulation process. Even at high thrombin concentration, autocrine and added ADP enhanced and prolonged Ca(2+) depletion from internal stores via stimulation of the P2Y(12) receptors. This P2Y(12)-dependent effect was mediated via two distinct signaling pathways. The first is enhanced Ca(2+) mobilization by the inositol 1,4,5-trisphosphate receptors due to inhibition of protein kinase A. The second pathway concerns prolonged activation of phosphoinositide 3-kinase (PI3-K) and phospholipase C. Experiments with phosphoinositide 3-kinase isoform-selective inhibitors and p110gamma deficient platelets demonstrated that the phosphoinositide 3-kinase beta and not the phosphoinositide 3-kinase gamma isoform is responsible for the prolonged Ca(2+) response and for the subsequent increases in procoagulant activity and coagulation. Taken together, these results demonstrate a dual P2Y(12)-dependent signaling mechanism, which increases the platelet-activating effect of thrombin by prolongation of Ca(2+) elevation, thereby facilitating the coagulation process.


Assuntos
Plaquetas/efeitos dos fármacos , Cálcio/metabolismo , Coagulantes/farmacologia , Isoenzimas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Purinérgicos P2/metabolismo , Transdução de Sinais , Trombina/farmacologia , Plaquetas/enzimologia , Plaquetas/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Citometria de Fluxo , Humanos , Técnicas In Vitro , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Receptores Purinérgicos P2Y12
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA