Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Thromb Haemost ; 16(9): 1686-1699, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29975003

RESUMO

Platelets can contribute to tumor progression and metastasis. Cancer patients are at increased risk of thrombosis, and advanced stages of cancer are associated with thrombocytosis or increased platelet reactivity. Tyrosine kinase inhibitors (TKIs) are widely used as a targeted strategy for cancer treatment, with the aim of prolonging progression-free survival of the patients. Because of their broad kinase target spectrum, most TKIs inevitably have off-target effects. Platelets rely on tyrosine kinase activity for their activation. Frequently observed side effects are lowering of platelet count and inhibition of platelet functions, whether or not accompanied by an increased bleeding risk. In this review, we aim to give insights into: (i) 38 TKIs that are currently used for the treatment of different types of cancer, either on the market or in clinical trials; (ii) how distinct TKIs can inhibit activation mechanisms in platelets; and (iii) the clinical consequences of the antiplatelet effects of TKI treatment. For several TKIs, the knowledge on affinity for their targets does not align with the published effects on platelets and reported bleeding events. This review should raise awareness of the potential antiplatelet effects of several TKIs, which will be enhanced in the presence of antithrombotic drugs.


Assuntos
Antineoplásicos/farmacologia , Plaquetas/efeitos dos fármacos , Terapia de Alvo Molecular/efeitos adversos , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Trombofilia/etiologia , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Humanos , Interleucina-6/biossíntese , Metástase Neoplásica , Proteínas de Neoplasias/fisiologia , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neovascularização Patológica/etiologia , Neovascularização Patológica/fisiopatologia , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/fisiologia , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Inibidores de Proteínas Quinases/efeitos adversos , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Tirosina Quinases/fisiologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/fisiologia , Trombocitose/etiologia , Trombofilia/induzido quimicamente , Trombofilia/prevenção & controle , Trombopoetina/biossíntese
2.
Thromb Res ; 140 Suppl 1: S171, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27161680

RESUMO

INTRODUCTION: Severe thrombocytopenia (≤50×10(9) platelets/L) is often the consequence of hematological malignancies and intensive chemotherapy. The risk of clinically significant bleeding is increased in these patients, despite the use of prophylactic platelet transfusions. The fact that there is no clear correlation between the platelet count and the risk of hemorrhage, suggests that there are other contributing factors. The contribution of impairments in platelet and coagulant function remains poorly understood. AIM: In patients with chemotherapy-induced thrombocytopenia due to hematological malignancies, we evaluate platelet and coagulant functions and determine the effects of platelet transfusion. Ultimately, we can identify specific hemostatic factors that aid in the prediction of bleeding. MATERIALS AND METHODS: In total 58 patients were included and blood was collected before and, if indicated (≤10×10(9) platelets/L), 1 hour after transfusion with platelet concentrate. Platelet function was assessed using flow cytometry by determining: 1) integrin αIIbß3 activation (PAC-1 antibody), 2) P-selectin expression (anti-P-selectin antibody), 3) phosphatidylserine exposure (Annexin-V) and 4) intracellular calcium (Fluo-4 AM). Factor levels were determined in plasma. Thrombus and fibrin formation was assessed by perfusion of whole blood over a collagen-tissue factor surface at a shear rate of 1,000 s-1. RESULTS: Platelets from the thrombocytopenic patients before transfusion showed markedly reduced integrin αIIbß3 activation and P-selectin expression in response to thrombin, collagen-related peptide and ADP, compared to healthy donor platelets. Also, agonist-induced intracellular calcium fluxes were greatly reduced. However, calcium fluxes with thapsigargin, a SERCA pump inhibitor, were similar in patient and control platelets, suggesting a normal calcium store content in the patient platelets. Furthermore, phosphatidylserine exposure was increased in unstimulated patient platelets compared to control platelets (8.2 vs. 1.8%, p<0.0001). Coagulation factor levels were within the normal range, with the exception of von Willebrand factor and fibrinogen levels, which were elevated. Platelet transfusion partly recovered the platelet integrin αIIbß3 activation and P-selectin expression induced by agonists. Platelet deposition (6.7 vs. 1.7%, p<0.0001) and fibrin formation (7.6 vs. 0.9%, p=0.0005) under flow conditions were substantially improved after platelet transfusion. CONCLUSIONS: Platelets from cancer patients undergoing chemotherapy appear to display impaired functional responses to activating stimuli. Platelet transfusion partly restores these functional defects, resulting in improved thrombus and fibrin formation.

3.
Thromb Res ; 140 Suppl 1: S183, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27161708

RESUMO

INTRODUCTION: The myeloproliferative neoplasms ET and PV are characterized by a high incidence of both arterial and venous thrombosis, and/or microcirculatory disturbances. Three somatic mutations, i.e. JAK2-V617F, Calreticulin (CalR) and MPL, commonly found in these diseases, correlate with different thrombotic risk levels. AIM: To analyze the influence of JAK2-V617F, CalR and MPL mutations on PLT adhesion, evaluated by a dynamic method under flow conditions in a group of patients with ET and PV. MATERIALS AND METHODS: 86 patients, i.e. 51 ET (19 M/32 F; age range 32-86 years) and 35PV (22 M/13 F; 41-83 yrs.), and 24 healthy controls (13 M/11 F; 28-61 yrs.) were enrolled upon informed consent. For the adhesion assay, peripheral venous whole blood was perfused over collagen for 4' at a 1,000 s-1 shear rate. PLTs were then stained with an anti-P-selectin-FITC antibody to evaluate PLT activation, and annexin V-AlexaFluor647 to detect procoagulant phosphatidylserine expression. Then, images of adherent PLTs in random fields were taken using phase contrast and fluorescence imaging by EVOS® fluorescence microscope. Results are mean±SEM of the % area covered by PLTs, or as the % of adherent PLTs positive for P-selectin or phosphatidylserine. Main hematological parameters and mutational status were recorded. RESULTS: PLT adhesion was significantly (p<0.01) greater in ET (44.6±1.6%) and PV patients (49.0±1.9%) compared to controls (37.9±1.7%). In ET, PLT adhesion was highest in JAK2-V617F mutation carriers (n=23), followed by CalR-positive (n=16) and triple negative subjects (n=9), and lowest in the MPL-positive patients (n=3). In PV, no difference in PLT adhesion was observed between JAK2-V617F heterozygous and homozygous subjects. P-selectin expression by adherent PLTs was not statistically different between patients and controls. Differently, phosphatidylserine expression on adherent PLTs was significantly reduced (p<0.01) in both ET and PV compared to healthy subjects. In ET patients, a significant (p<0.05) correlation was found between PLT adhesion and PLT count in JAK2-V617F and CalR-positive mutation carriers. Multivariate regression analysis adjusted for age and sex, confirmed PLT count as a significant determinant of PLT adhesion in JAK2-V617F positive patients only. CONCLUSIONS: ET and PV platelets show an increased adhesion to collagen in vitro, particularly in those carrying the JAK2-V617F mutation. A prospective study is ongoing to evaluate the predictive value of our PLT thrombus formation dynamic model for the thrombotic risk in ET and PV patients. ACKNOWLEDGEMENT: Project funded by "AIRC-IG2013" grant Nr. 14505 from the "Italian Association for Cancer Research" (A.I.R.C.).

4.
J Thromb Haemost ; 13 Suppl 1: S272-80, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26149036

RESUMO

The potential relevance of murine atherothrombosis models for understanding human disease has been debated in the past. Despite this, in the last decade, many thrombosis studies with atherogenic Apoe(-/-) mice have been performed, which provide novel insight into the molecular mechanisms by which platelet and coagulation processes accomplish acute thrombus formation after plaque disruption in vivo. Support for these mechanisms has come from whole blood flow perfusion studies over plaque material in vitro, which are also reviewed in this study. The main plaque-derived triggers for thrombus formation appear to be collagen and tissue factor, next to bioactive mediators such as prostaglandin E2. The atherothrombotic process relies on collagen- and ADP-receptor-induced platelet activation as well as on thrombin/fibrin generation via the extrinsic and intrinsic coagulation pathways. Less is known of the persistent effects of a thrombus on atherosclerosis progression, but evidence suggests roles herein of activated platelets and ongoing thrombin generation.


Assuntos
Aterosclerose/sangue , Coagulação Sanguínea , Plaquetas/metabolismo , Trombose/sangue , Animais , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Artérias/metabolismo , Artérias/patologia , Aterosclerose/genética , Aterosclerose/patologia , Colágeno/metabolismo , Modelos Animais de Doenças , Fibrinólise , Humanos , Camundongos Knockout , Placa Aterosclerótica , Inibidor 1 de Ativador de Plasminogênio/sangue , Ativação Plaquetária , Ruptura Espontânea , Transdução de Sinais , Trombose/genética
5.
J Thromb Haemost ; 13(8): 1503-13, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25988848

RESUMO

BACKGROUND: Patients undergoing major cardiothoracic surgery are subjected to dilution, owing to massive fluid infusion and blood component transfusion. These patients may experience bleeding perioperatively, and are frequently treated with the endothelium-activating agent desmopressin. OBJECTIVES: To investigate the effect of desmopressin administration on von Willebrand factor (VWF)-dependent coagulant and platelet functions under flow conditions. PATIENTS/METHODS: Blood from 16 patients with postoperative bleeding was obtained before and after desmopressin treatment (0.3 µg kg(-1) body weight), and assessed for coagulant properties and platelet function. Furthermore, VWF antigen levels and multimer composition were determined in both samples. RESULTS: Desmopressin treatment did not change thrombin generation in plasma or whole blood thromboelasticity. Also coagulation factor levels (other than factor VIII) and coagulation times were unchanged, suggesting that desmopressin treatment did not have a major effect on the coagulant activity. On the other hand, desmopressin treatment raised the already high plasma levels of VWF from a median of 116 IU mL(-1) (interquartile range [IQR] 102-154 IU mL(-1) ) to a median of 160 IU mL(-1) (IQR 126-187 IU mL(-1) ) (P = 0.007), owing to accumulation of the high molecular weight VWF multimers. Furthermore, desmopressin treatment caused an increase in collagen-dependent thrombus formation and platelet phosphatidylserine exposure. Markers of thrombus formation correlated with the plasma levels of VWF. In vitro control experiments confirmed a major contribution of VWF to thrombus formation and procoagulant activity under conditions of blood dilution. CONCLUSIONS: Desmopressin treatment of patients with bleeding complications after cardiothoracic surgery induces the release of high molecular weight VWF multimers, which enhance platelet activation and thrombus formation under flow conditions.


Assuntos
Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Desamino Arginina Vasopressina/uso terapêutico , Hemostáticos/uso terapêutico , Hemorragia Pós-Operatória/tratamento farmacológico , Idoso , Testes de Coagulação Sanguínea , Plaquetas/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Fosfatidilserinas/sangue , Ativação Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Hemorragia Pós-Operatória/sangue , Hemorragia Pós-Operatória/etiologia , Resultado do Tratamento , Fator de von Willebrand/metabolismo
6.
Thromb Haemost ; 112(2): 271-5, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24740693

RESUMO

The Calibrated Automated Thrombogram (CAT) assay that measures thrombin generation (TG) in platelet-poor and -rich plasma, is increasingly being recognised as a more sensitive tool to determine the overall function of the haemostatic system. We developed a method enabling the measurement of TG in a small aliquot of blood. The objective was to validate this assay in mouse blood and to examine the rate and extent of TG in a mouse model of premature aging. TG was assayed in blood from 20- to 28-week-old brain and muscle ARNT-like protein-1 (Bmal1)-deficient (knockout, KO) mice and wild-type (WT) littermates. Bmal1-KO mice are known to display symptoms of premature aging. TG was initiated by adding calcium, tissue factor and a thrombin specific substrate. After TG, the samples were prepared for scanning electron microscopy (SEM). The intra-assay variations (%) in mouse blood of the endogenous thrombin potential (ETP), peak height, lag time, time-to-peak and velocity index were 10% or less (n=24). We found that Bmal1-KO mice have a significantly (p<0.001) higher ETP (437 ± 7 nM.min; mean ± SD, n=7) when compared with WT mice (ETP=220 ± 45 nM.min; mean ± SD, n=5). The peak heights also differed significantly (p=0.027). By applying SEM we found that Bmal1 deficient mice display a denser fibrin network with smaller pores compared to WT mice. In conclusion, the whole blood TG assay in mice revealed to be reproducible. As a proof-of-principle we have shown that the whole blood TG assay is capable of detecting a prothrombotic phenotype in Bmal1-KO mice.


Assuntos
Fatores de Transcrição ARNTL/deficiência , Testes de Coagulação Sanguínea , Coagulação Sanguínea , Trombina/metabolismo , Trombose/diagnóstico , Fatores de Transcrição ARNTL/genética , Senilidade Prematura/sangue , Senilidade Prematura/genética , Animais , Coagulação Sanguínea/genética , Modelos Animais de Doenças , Fibrina/metabolismo , Fibrina/ultraestrutura , Predisposição Genética para Doença , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Fenótipo , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Trombose/sangue , Trombose/genética
7.
J Thromb Haemost ; 12(4): 528-39, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24520961

RESUMO

BACKGROUND: Orai1, the major store-operated Ca(2+) entry (SOCE) channel in platelets, is not only critical for enhancing diverse signaling pathways, but may also regulate receptor-operated Ca(2+) entry (ROCE). Dynamic coupling of the Orai1 signalosome to canonical transient receptor potential channels (TRPCs) has been suggested as an essential step in the activation of SOCE and ROCE. However, the functional significance of the biochemical interaction between Orai and TRPC isoforms remains controversial. OBJECTIVE: We aimed to elucidate the role of Orai1 in diacylglycerol (DAG)-mediated ROCE. METHODS: Trpc6(-/-) , Orai1(-/-) and Orai1(-/-) /Trpc6(-/-) mice were generated, and their platelets were analyzed. RESULTS: Thapsigargin (TG)-induced SOCE was further reduced in Orai1(-/-) /Trpc6(-/-) platelets as compared with Orai1(-/-) platelets, thus revealing that TG-induced signaling pathways can activate TRPC6. Thapsigargin-induced SOCE leads to enhanced phospholipase C and D activity in wild-type platelets. The activity of both enzymes was significantly reduced in Orai1(-/-) platelets upon TG stimulation, whereas receptor-induced phospholipase activity was not affected. Furthermore, TG-induced and glycoprotein VI-mediated thromboxane A2 release was strongly dependent on Orai1-mediated SOCE. CONCLUSION: The regulation of TRPC6 activity can occur independently of the physical interaction with Orai1. TRPC6 operates in crosstalk with Orai1 through Orai1-induced DAG production via phospholipase activation. Orai1-induced DAG production and thromboxane release amplify the second phase of Ca(2+) signaling in platelets.


Assuntos
Canais de Cálcio/metabolismo , Fosfolipases/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Plaquetas/citologia , Cálcio/metabolismo , Diglicerídeos/química , Glicoproteínas/química , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteína ORAI1 , Agregação Plaquetária , Transdução de Sinais , Canal de Cátion TRPC6 , Tapsigargina/metabolismo , Tromboxano A2/metabolismo , Fatores de Tempo
8.
Thromb Haemost ; 111(3): 447-57, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-24258426

RESUMO

Blood dilution after transfusion fluids leads to diminished coagulant activity monitored by rotational thromboelastometry, assessing elastic fibrin clot formation, or by thrombin generation testing. We aimed to determine the contributions of blood cells (platelets, red blood cells) and plasma factors (fibrinogen, prothrombin complex concentrate) to fibrin clot formation under conditions of haemodilution in vitro or in vivo.Whole blood or plasma diluted in vitro was supplemented with platelets, red cells, fibrinogen or prothrombin complex concentrate (PCC). Thromboelastometry was measured in whole blood as well as plasma; thrombin generation was determined in parallel. Similar tests were performed with blood from 48 patients, obtained before and after massive fluid infusion during cardiothoracic surgery.Addition of platelets or fibrinogen, in additive and independent ways, reversed the impaired fibrin clot formation (thromboelastometry) in diluted whole blood. In contrast, supplementation of red blood cells or prothrombin complex concentrate was ineffective. Platelets and fibrinogen independently restored clot formation in diluted plasma, resulting in thromboelastometry curves approaching those in whole blood. In whole blood from patients undergoing dilution during surgery, elastic clot formation was determined by both the platelet count and the fibrinogen level. Thrombin generation in diluted (patient) plasma was not changed by fibrinogen, but improved markedly by prothrombin complex concentrate. In conclusion, in dilutional coagulopathy, platelets and fibrinogen, but not red blood cells or vitamin K-dependent coagulation factors, independently determine thromboelastometry parameters measured in whole blood and plasma. Clinical decisions for transfusion based on thromboelastometry should take into account the platelet concentration.


Assuntos
Plaquetas/patologia , Ponte Cardiopulmonar , Fibrina/metabolismo , Fibrinogênio/metabolismo , Hemorragia/prevenção & controle , Idoso , Coagulação Sanguínea , Plaquetas/metabolismo , Eritrócitos/patologia , Feminino , Hemodiluição/efeitos adversos , Hemorragia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Contagem de Plaquetas , Protrombina/metabolismo , Tromboelastografia , Trombina/metabolismo , Reação Transfusional
9.
Cell Death Dis ; 4: e611, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23618909

RESUMO

Immune cells and platelets maintain plasma membrane phospholipid asymmetry. Upon activation, this asymmetry is disrupted by phospholipid scrambling (PS), which is a major step during activation of immune cells, hemostasis and apoptosis. Anoctamin 6 (Ano6; TMEM16F) causes chloride (Cl(-)) and cation currents and is required for Ca(2+)-dependent PS. It is defective in blood cells from patients with Scott syndrome, a rare bleeding disorder. We examined if Cl(-) currents and PS are related, whether both processes are Ca(2+) dependent, and whether Ca(2+)-independent scrambling during intrinsic and extrinsic apoptosis is controlled by Ano6. Ca(2+) increase by ionomycin activated Ano6 Cl(-) currents and PS in normal lymphocytes, but not in B-lymphocytes from two different patients with Scott syndrome. Fas ligand (FasL) did not increase intracellular Ca(2+), but activated Cl(-) currents in normal but not in Scott lymphocytes. Whole-cell currents were inhibited by Cl(-) channel blockers and by siRNA knockdown of Ano6. In contrast, intrinsic mitochondrial apoptosis by ABT-737 did not induce Cl(-) currents in lymphocytes. PS was not inhibited by blockers of Ano6 or removal of Cl(-) ions. Remarkably, Ca(2+)-independent scrambling due to extrinsic (FasL) or intrinsic (ABT-737) apoptosis was unchanged in Scott cells. We conclude that: (i) Ano6 Cl(-) currents are activated by increase in cytosolic Ca(2+), or Ca(2+) independent by stimulation of Fas receptors; (ii) Ca(2+)-dependent PS induced by Ano6 does not require Cl(-) currents; (iii) Ca(2+)-independent PS does not require Ano6; (iv) Ano6 is necessary for Ca(2+)-dependent PS, but not by increasing intracellular Ca(2+).


Assuntos
Cálcio/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Fosfolipídeos/metabolismo , Anoctaminas , Apoptose/efeitos dos fármacos , Linfócitos B/imunologia , Linfócitos B/fisiologia , Compostos de Bifenilo/farmacologia , Transtornos da Coagulação Sanguínea/fisiopatologia , Ionóforos de Cálcio/farmacologia , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/metabolismo , Proteína Ligante Fas/farmacologia , Células HEK293 , Humanos , Transporte de Íons/efeitos dos fármacos , Ionomicina/farmacologia , Células Jurkat , Nitrofenóis/farmacologia , Técnicas de Patch-Clamp , Proteínas de Transferência de Fosfolipídeos/antagonistas & inibidores , Proteínas de Transferência de Fosfolipídeos/genética , Piperazinas/farmacologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Sulfonamidas/farmacologia
10.
J Thromb Haemost ; 11(1): 2-16, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23106920

RESUMO

Platelets in a thrombus interact with (anti)coagulation factors and support blood coagulation. In the concept of cell-based control of coagulation, three different roles of platelets can be distinguished: control of thrombin generation, support of fibrin formation, and regulation of fibrin clot retraction. Here, we postulate that different populations of platelets with distinct surface properties are involved in these coagulant functions. Platelets with elevated Ca(2+) and exposed phosphatidylserine control thrombin and fibrin generation, while platelets with activated α(IIb) ß(3) regulate clot retraction. We review how coagulation factor binding depends on the platelet activation state. Furthermore, we discuss the ligands, platelet receptors and downstream intracellular signaling pathways implicated in these coagulant functions. These insights lead to an adapted model of platelet-based coagulation.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Ativação Plaquetária , Trombose/sangue , Animais , Plaquetas/classificação , Cálcio/sangue , Retração do Coágulo , Fibrina/metabolismo , Humanos , Fosfatidilserinas/sangue , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais , Trombina/metabolismo
11.
Vox Sang ; 103(1): 25-34, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22211833

RESUMO

BACKGROUND AND OBJECTIVES: Treatment of dilutional coagulopathy by transfusing fresh frozen plasma (FFP) remains sub-optimal. We hypothesized that partial replacement of transfused FFP by fibrinogen concentrate results in improved coagulant activity and haemostasis. This was tested in a controlled clinical intervention trial with patients experiencing massive bleeding during major surgery. METHODS: Patients undergoing major elective surgery were treated according to current protocols. When transfusion with FFP was required, patients were randomized as follows: group A received 4 units FFP and group B received 2 units FFP plus 2 g fibrinogen concentrate. Blood samples were taken before and after the intervention. Analysts were blinded to the treatment type. RESULTS: Group A (B) consisted of 21 (22) patients, in 16 (17) of whom bleeding stopped after intervention. Plasma fibrinogen increased significantly more in group B (0·57 g/l) than in group A (0·05 g/l). However, levels of prothrombin and factors VIII, IX and X increased more in group A than in group B. Rotational thromboelastometry (ROTEM) of whole blood and plasma revealed improved fibrin clot formation in group B but not in group A. Thrombin generation [calibrated automated thrombogram (CAT)] in plasma increased more in group A. Principal parameters determining whole-blood thromboelastometry were the fibrinogen level and platelet count. In vitro addition of fibrinogen and prothrombin complex concentrate to pre-intervention samples restored both ROTEM and CAT parameters. CONCLUSIONS: Partial replacement of transfused FFP by fibrinogen increases fibrin clot formation at the expense of less improved thrombin generation. Coagulation factors other than fibrinogen alone are required for full restoration of haemostasis.


Assuntos
Transtornos da Coagulação Sanguínea/terapia , Transfusão de Componentes Sanguíneos , Fibrinogênio/uso terapêutico , Procedimentos Cirúrgicos Operatórios/efeitos adversos , Procedimentos Cirúrgicos Operatórios/métodos , Idoso , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/etiologia , Fatores de Coagulação Sanguínea/metabolismo , Perda Sanguínea Cirúrgica/prevenção & controle , Feminino , Fibrina/efeitos dos fármacos , Fibrina/metabolismo , Hemostasia/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Plasma/metabolismo , Contagem de Plaquetas , Hemorragia Pós-Operatória/prevenção & controle , Hemorragia Pós-Operatória/terapia , Estudos Prospectivos , Tromboelastografia
12.
Cell Death Dis ; 2: e211, 2011 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-21956548

RESUMO

Platelets shed microparticles not only upon activation, but also upon ageing by an apoptosis-like process (apoptosis-induced platelet microparticles, PM(ap)). While the activation-induced microparticles have widely been studied, not much is known about the (patho)physiological consequences of PM(ap) formation. Flow cytometry and scanning electron microscopy demonstrated that PM(ap) display activated integrins and interact to form microparticle aggregates. PM(ap) were chemotactic for monocytic cells, bound to these cells, an furthermore stimulated cell adhesion and spreading on a fibronectin surface. After prolonged incubation, PM(ap) promoted cell differentiation, but inhibited proliferation. Monocyte membrane receptor analysis revealed increased expression levels of CD11b (integrin α(M)ß(2)), CD14 and CD31 (platelet endothelial cell adhesion molecule-1), and the chemokine receptors CCR5 and CXCR4, but not of CCR2. This indicated that PM(ap) polarized the cells into resident M2 monocytes. Cells treated with PM(ap) actively consumed oxidized low-density lipoprotein (oxLDL), and released matrix metalloproteinases and hydrogen peroxide. Further confirmation for the differentiation towards resident professional phagocytes came from the finding that PM(ap) stimulated the expression of the (ox)LDL receptors, CD36 and CD68, and the production of proinflammatory and immunomodulating cytokines by monocytes. In conclusion, interaction of PM(ap) with monocytic cells has an immunomodulating potential. The apoptotic microparticles polarize the cells into a resident M2 subset, and induce differentiation to resident professional phagocytes.


Assuntos
Apoptose/fisiologia , Plaquetas/citologia , Micropartículas Derivadas de Células/metabolismo , Macrófagos/citologia , Monócitos/citologia , Plaquetas/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Macrófagos/metabolismo , Masculino , Microscopia Eletrônica , Monócitos/metabolismo
13.
J Thromb Haemost ; 9(9): 1835-46, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21696539

RESUMO

BACKGROUND AND OBJECTIVE: Platelets abundantly express glycoprotein CD36 with thrombospondin-1 (TSP1) and oxidized low-density lipoprotein (oxLDL) as proposed ligands. How these agents promote platelet activation is still poorly understood. METHODS AND RESULTS: Both TSP1 and oxLDL caused limited activation of platelets in suspension. However, immobilized TSP1 and oxLDL, but not LDL, strongly supported platelet adhesion and spreading with a major role of CD36. Platelet spreading was accompanied by potent Ca(2+) rises, and resulted in exposure of P-selectin and integrin activation, all in a CD36-dependent manner with additional contributions of α(IIb) ß(3) and ADP receptor stimulation. Signaling responses via CD36 involved activation of the protein tyrosine kinase Syk. In whole blood perfusion, co-coating of TSP1 or oxLDL with collagen enhanced thrombus formation at high-shear flow conditions, with increased expression on platelets of activated α(IIb) ß(3), P-selectin and phosphatidylserine, again in a CD36-dependent way. CONCLUSIONS: Immobilized TSP1 and oxLDL activate platelets partly via CD36 through a Syk kinase-dependent Ca(2+) signaling mechanism, which enhances collagen-dependent thrombus formation under flow. These findings provide novel insight into the role of CD36 in hemostasis.


Assuntos
Antígenos CD36/sangue , Lipoproteínas LDL/sangue , Ativação Plaquetária/fisiologia , Trombose/sangue , Trombose/imunologia , Trombospondina 1/sangue , Sinalização do Cálcio , Movimento Celular , Colágeno/farmacologia , Humanos , Proteínas Imobilizadas , Técnicas In Vitro , Microscopia de Vídeo , Modelos Biológicos , Ativação Plaquetária/efeitos dos fármacos , Adesividade Plaquetária , Transdução de Sinais , Trombose/etiologia
16.
Cardiovasc Hematol Agents Med Chem ; 9(1): 42-55, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20939828

RESUMO

The glycoprotein CD36, also known as glycoprotein IIIb/IV or FAT, is expressed on the surface of platelets, monocytes, microvascular endothelial cell, smooth muscle cells, cardiomyocytes and other cells of the cardiovascular system. In spite of its abundant presence, CD36 has remained for long a mysterious protein with a poorly understood role. In this paper, we review how CD36 can affect cellular responses by interaction with a variety of ligands, in particular thrombospondin-1, oxidized lipoproteins and fatty acids. Furthermore, given the structure of CD36 with two transmembrane domains and short cytoplasmic tails, we consider how this receptor can induce intracellular signaling, likely in junction with other cellular receptors or associated proteins in the membrane. Current literature points to activation of Src-family and mitogen-activated protein kinases, as well as to activation of the NFκB and Rho pathways. The new insights make CD36 attractive as a therapeutic target to suppress platelet and monocyte/macrophage function and thereby atherothrombosis.


Assuntos
Antígenos CD36/metabolismo , Trombose/metabolismo , Animais , Aterosclerose/metabolismo , Antígenos CD36/química , Antígenos CD36/genética , Ácidos Graxos/metabolismo , Humanos , Lipoproteínas/metabolismo , Transdução de Sinais , Trombospondina 1/metabolismo
18.
J Thromb Haemost ; 8(8): 1797-808, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20546121

RESUMO

BACKGROUND: Interaction of murine Gas6 with the platelet Gas6 receptors Tyro3, Axl and Mer (TAM) plays an important role in arterial thrombus formation. However, a role for Gas6 in human platelet activation has been questioned. OBJECTIVE: To determine the role of Gas6 in human and murine platelet activation and thrombus formation. METHODS AND RESULTS: Gas6 levels appeared to be 20-fold higher in human plasma than in platelets, suggesting a predominant role of plasma-derived Gas6. Human Gas6 synergizes with ADP-P2Y(12) by enhancing and prolonging the phosphorylation of Akt. Removal of Gas6 from plasma impaired ADP-induced platelet aggregation. Under flow conditions, absence of human Gas6 provoked gradual platelet disaggregation and integrin α(IIb) ß(3) inactivation. Recombinant human Gas6 reversed the effects of Gas6 removal. In mouse blood, deficiency in Gas6 or in one of the TAM receptors led to reduced thrombus formation and increased disaggregation, which was completely antagonized by external ADP. In contrast, collagen-induced platelet responses were unchanged by the absence of Gas6 in both human and mouse systems. CONCLUSIONS: The ADP-P2Y(12) and Gas6-TAM activation pathways synergize to achieve persistent α(IIb) ß(3) activation and platelet aggregation. We postulate a model of thrombus stabilization in which plasma Gas6, by signaling via the TAM receptors, extends and enhances the platelet-stabilizing effect of autocrine ADP, particularly when secretion becomes limited.


Assuntos
Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Ativação Plaquetária , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Trombose/metabolismo , Animais , Humanos , Camundongos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteínas Recombinantes/química , Transdução de Sinais , Trombose/patologia , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
19.
Thromb Haemost ; 103(2): 318-28, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20024495

RESUMO

Patients subjected to haemodilution during surgery are at increased risk of bleeding. We hypothesised that, in the acquired dilutional coagulopathy, insufficient haemostasis is due to either insufficient thrombin generation or insufficient fibrin clot formation. In tissue factor-activated plasmas from patients with coagulation deficiency, we measured time curves of thrombin generation and fibrin clot formation (thromboelastography). Investigated were in study A: 10 patients treated with vitamin K antagonist and five healthy subjects; in study B: 30 patients undergoing cardiopulmonary bypass (CPB) surgery and infused with on average 2,000 ml crystalloids and colloids (no major bleeding); in study C: 58 patients undergoing major general surgery, and transfused with >5,000 ml crystalloids, colloids and red cell concentrates, who experienced major bleeding and were post-transfused with fresh frozen plasma. The treatment with vitamin K antagonist led to a progressive reduction in thrombin generation but not fibrin clot formation. In CPB patients, plasma factor levels post-surgery were 53-60% of normal. This was accompanied by moderate reduction in both haemostatic processes. In plasmas from patients undergoing major surgery, factor levels were 38-41% of normal, and these levels increased after plasma transfusion. Taking preset thresholds for normal thrombin generation and fibrin clot formation, at least one of these processes was low in 88-93% of the patients with (persistent) bleeding, but only in 40-53% of the patients without bleeding. In conclusion, the ability of thrombin generation and fibrin clot formation is independently reduced in acquired dilutional coagulopathy, while minimal levels of both are required for adequate haemostasis.


Assuntos
Fibrina/metabolismo , Hemodiluição , Hemorragia/etiologia , Trombina/biossíntese , Idoso , Coagulação Sanguínea , Transtornos da Coagulação Sanguínea/etiologia , Perda Sanguínea Cirúrgica/prevenção & controle , Transfusão de Sangue , Soluções Cristaloides , Feminino , Hemorragia/prevenção & controle , Hemostasia , Humanos , Soluções Isotônicas/uso terapêutico , Cinética , Masculino , Pessoa de Meia-Idade , Assistência Perioperatória , Hemorragia Pós-Operatória/etiologia , Hemorragia Pós-Operatória/terapia , Vitamina K/antagonistas & inibidores
20.
J Thromb Haemost ; 7(1): 152-61, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18983512

RESUMO

BACKGROUND: Atherothrombosis is a major cause of cardiovascular events. However, animal models to study this process are scarce. OBJECTIVES: We describe the first murine model of acute thrombus formation upon plaque rupture to study atherothrombosis by intravital fluorescence microscopy. METHODS: Localized rupture of an atherosclerotic plaque in a carotid artery from Apoe(-/-) mice was induced in vivo using ultrasound. Rupture of the plaque and formation of localized thrombi were verified by two-photon laser scanning microscopy (TPLSM) in isolated arteries, and by immunohistochemistry. The thrombotic reaction was quantified by intravital fluorescence microscopy. RESULTS: Inspection of the ultrasound-treated plaques by histochemistry and TPLSM demonstrated local damage, collagen exposure, luminal thrombus formation as well as intra-plaque intrusion of erythrocytes and fibrin. Ultrasound treatment of healthy carotid arteries resulted in endothelial damage and limited platelet adhesion. Real-time intravital fluorescence microscopy demonstrated rapid platelet deposition on plaques and formation of a single thrombus that remained subocclusive. The thrombotic process was antagonized by thrombin inhibition, or by blocking of collagen or adenosine diphosphate receptor pathways. Multiple thrombi were formed in 70% of mice lacking CD40L. CONCLUSIONS: Targeted rupture of murine plaques results in collagen exposure and non-occlusive thrombus formation. The thrombotic process relies on platelet activation as well as on thrombin generation and coagulation, and is sensitive to established and novel antithrombotic medication. This model provides new possibilities to study atherothrombosis in vivo.


Assuntos
Coagulação Sanguínea/fisiologia , Plaquetas/fisiologia , Trombose/etiologia , Animais , Aterosclerose/complicações , Aterosclerose/patologia , Trombose das Artérias Carótidas , Colágeno , Modelos Animais de Doenças , Eritrócitos/patologia , Fibrina , Camundongos , Microscopia de Fluorescência , Trombose/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...