Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neuroimmunol ; 367: 577863, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35436744

RESUMO

Cerebral small vessel disease (cSVD) accounts for 25% of ischemic strokes and is a major cause of cognitive decline. Inflammatory processes, involving immune cells and platelets might drive development and progression of cSVD. The aim of the study was to identify potential novel biomarkers for cSVD, gaining new insights into its pathophysiology. We measured inflammation and platelet and neutrophil activation markers in patients with cSVD and age-matched controls. It was hypothesized that cSVD is accompanied by altered levels of these markers. The levels of interleukin 1ß, CX3CL1, CXCL4, CXCL7, myeloperoxidase (MPO), MPO-DNA complex and S100A8/A9 were measured by ELISA in plasma samples of patients with cSVD presenting with mild vascular cognitive impairment (mVCI, n = 36) or lacunar stroke (Laci, n = 44), and controls (n = 38). To determine the relevance of these ELISA markers compared with patient- and MRI-based characteristics, all characteristics were entered into three machine learning models. Among the ELISA markers measured, MPO levels were significantly elevated in patients with cSVD (48.3 (27.8-80.1, interquartile range) ng/mL) compared with controls (32.2 (19.6-47.4) ng/mL, P = 0.023), particularly in the Laci group (56.8 (33.3-84.7) ng/mL, P = 0.004). Regularized logistic regression and random forest algorithms returned MPO levels as an important feature in the detection and prediction of cSVD. Of note, logistic regression and random forest analysis also highlighted levels of CXCL4, CXCL7, MPO-DNA and S100A8/A9 as features associated with cSVD. Taken together, the neutrophil activation marker MPO is elevated in patients with Laci and machine learning indicates platelet and neutrophil markers as interesting molecules for future investigation. SHORTENED ABSTRACT: Cerebral small vessel disease (cSVD) is a major cause of cognitive decline and stroke. We aimed to identify potential novel biomarkers for cSVD and to obtain new insights into its pathophysiology. Levels of markers reflecting neutrophil activation, neutrophil extracellular trap (NET) formation, platelet activation and vascular inflammation were measured in plasma samples of patients with cSVD, and controls. Only myeloperoxidase (MPO) levels were significantly altered. Regularized logistic regression and random forest algorithms returned MPO levels as an important feature in the detection and prediction of cSVD and highlighted platelet- and NET markers as cSVD associated.


Assuntos
Doenças de Pequenos Vasos Cerebrais , Peroxidase , Inteligência Artificial , Biomarcadores , Doenças de Pequenos Vasos Cerebrais/complicações , Doenças de Pequenos Vasos Cerebrais/diagnóstico por imagem , DNA , Humanos , Inflamação/complicações , Neutrófilos , Ativação Plaquetária
2.
J Proteomics ; 261: 104577, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35351662

RESUMO

The cAMP-protein kinase A (PKA) pathway in platelets is important for both platelet activation and inactivation. We hypothesize that proteins/processes downstream of the cAMP-PKA pathway that are regulated after platelet activation ánd subsequent inactivation can serve as a "switch" in platelet activation and inhibition. We used a STRING-based protein-protein interaction network from proteins of interest distilled from publicly available quantitative platelet proteome datasets. The protein network was integrated with biological pathway information by functional enrichment analysis, phosphorylation by PKA, and drug-target information. Functional enrichment analysis revealed biological processes related to vesicle secretion and cytoskeletal reorganization to be overrepresented among these 30 proteins coinciding with topological clusters in the network. Our method identified proteins/processes with functions related to vesicle transport, cyclin-dependent protein kinases, tight junctions, and small GTPases as potential switches in platelet activation and inhibition. Next to established enzymes in cAMP-PKA signaling, such as PDE3A, proteins with an unknown/less well-known role in platelet biology, such as Stonin-2 and ABLIM-3, emerged from our analysis as interesting candidates for reversal of platelet activation. Our method can be used to repurpose existing datasets and provide a coherent overview of mechanisms involved to predict novel connections, by visually integrating multiple datasets. SIGNIFICANCE: This article presents a novel approach of visually incorporating multiple existing tools and proteomics datasets and in doing so provides novel insight into the complex molecular mechanisms involved in platelet activation. Using our approach, we also highlight several interesting candidates for future research into pathologies with high platelet reactivity.


Assuntos
AMP Cíclico , Ativação Plaquetária , Plaquetas/metabolismo , AMP Cíclico/metabolismo , Fosforilação , Ativação Plaquetária/fisiologia , Proteômica
4.
Thromb Res ; 212: 72-80, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35247800

RESUMO

BACKGROUND: Bruton's kinase (Btk) is critical for collagen-triggered platelet signal transduction. The Btk inhibitor ibrutinib has been shown to selectively block platelet adhesion to atherosclerotic plaque material under laminar arterial flow. However, this has not been studied under a shear gradient, which is characteristic for atherothrombosis. OBJECTIVE: To determine the effect of ibrutinib treatment on in vitro thrombus formation on collagen and atherosclerotic plaque material in the absence or presence of a shear gradient. METHODS: Blood was obtained from patients with chronic lymphocytic leukemia, mantle-cell lymphoma and Waldenström macroglobulinemia with and without ibrutinib treatment and perfused through a microfluidic channel with(out) 60% stenosis over Horm type I collagen or human atherosclerotic plaque homogenate. RESULTS: At a constant shear rate of 1500 s-1, platelet deposition was significantly decreased in blood from haematological malignancy patients treated with ibrutinib as compared to untreated patients, on atherosclerotic plaque material but not on collagen. However, thrombus size, stability, and height, were reduced on both plaque material and collagen. An increase in shear rate up to 3900 s-1, as induced by 60% stenosis, resulted in decreased platelet deposition and thrombus parameters on plaque material but not on collagen when compared to a laminar shear of 1500 s-1. Ibrutinib treatment decreased platelet deposition and thrombus parameters even further around the stenosis. CONCLUSION: Treatment of patients with haematological disorders with the Btk inhibitor ibrutinib reduces in vitro platelet deposition, thrombus size and contraction on human atherosclerotic plaque around a stenosis when compared to patients not receiving ibrutinib.


Assuntos
Leucemia Linfocítica Crônica de Células B , Piperidinas , Adenina/análogos & derivados , Adenina/farmacologia , Adulto , Constrição Patológica , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Piperidinas/farmacologia , Piperidinas/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
5.
Cell Mol Bioeng ; 15(1): 55-65, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35087606

RESUMO

INTRODUCTION: Studying arterial thrombus formation by in vitro flow assays is a widely used approach. Incorporating human atherosclerotic plaque material as a thrombogenic surface in these assays represents a method to model the pathophysiological environment of thrombus formation upon plaque disruption. Up until now, achieving a homogeneous coating of plaque material and subsequent reproducible platelet adhesion has been challenging. Here, we characterized a novel method for coating of plaque material on glass coverslips for use in thrombosis microfluidic assays. METHODS: A homogenate of human atherosclerotic plaques was coated on glass coverslips by conventional manual droplet coating or by spin coating. Prior to coating, a subset of coverslips was plasma treated. Water contact angle measurements were performed as an indicator for the hydrophilicity of the coverslips. Homogeneity of plaque coatings was determined using profilometric analysis and scanning electron microscopy. Thrombogenicity of the plaque material was assessed in real time by microscopic imaging while perfusing whole blood at a shear rate of 1500 s-1 over the plaque material. RESULTS: Plasma treatment of glass coverslips, prior to spin coating with plaque material, increased the hydrophilicity of the coverslip compared to no plasma treatment. The most homogeneous plaque coating and highest platelet adhesion was obtained upon plasma treatment followed by spin coating of the plaque material. Manual plaque coating on non-plasma treated coverslips yielded lowest coating homogeneity and platelet adhesion and activation. CONCLUSION: Spin coating of atherosclerotic plaque material on plasma treated coverslips leads to a more homogenous coating and improved platelet adhesion to the plaque when compared to conventional droplet coating on non-plasma treated coverslips. These properties are beneficial in ensuring the quality and reproducibility of flow experiments. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s12195-021-00713-9.

6.
Sci Rep ; 10(1): 19360, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33168914

RESUMO

While in recent trials the dual pathway inhibition with aspirin plus rivaroxaban has shown to be efficacious in patients with atherosclerotic cardiovascular disease, little is known about the effects of this combination treatment on thrombus formation and vascular remodelling upon vascular damage. The aim of this study was to examine the effects of aspirin and/or rivaroxaban on injury-induced murine arterial thrombus formation in vivo and in vitro, vessel-wall remodelling, and platelet-leukocyte aggregates. Temporary ligation of the carotid artery of C57BL/6 mice, fed a western type diet, led to endothelial denudation and sub-occlusive thrombus formation. At the site of ligation, the vessel wall stiffened and the intima-media thickened. Aspirin treatment antagonized vascular stiffening and rivaroxaban treatment led to a positive trend towards reduced stiffening. Local intima-media thickening was antagonized by both aspirin or rivaroxaban treatment. Platelet-leukocyte aggregates and the number of platelets per leukocyte were reduced in aspirin and/or rivaroxaban treatment groups. Furthermore, rivaroxaban restricted thrombus growth and height in vitro. In sum, this study shows vascular protective effects of aspirin and rivaroxaban, upon vascular injury of the mouse artery.


Assuntos
Aspirina/farmacologia , Artérias Carótidas/efeitos dos fármacos , Inibidores do Fator Xa/farmacologia , Rivaroxabana/farmacologia , Trombose/tratamento farmacológico , Animais , Artérias/efeitos dos fármacos , Plaquetas/metabolismo , Artérias Carótidas/cirurgia , Doenças das Artérias Carótidas/tratamento farmacológico , Espessura Intima-Media Carotídea , Leucócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Varredura , Inibidores da Agregação Plaquetária/uso terapêutico , Trombose/metabolismo , Trombose/fisiopatologia
9.
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
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.
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
12.
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
13.
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
14.
J Thromb Haemost ; 6(8): 1253-61, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18513212

RESUMO

In the classical concept of platelet integrin activation, it is considered that unidirectional conformational changes of alpha(IIb)beta(3) and alpha(2)beta(1) regulate the adhesiveness of platelets for fibrin(ogen) and collagen, respectively. Here, we summarize recent evidence that these conformational changes: (i) can also occur in the reverse direction; and (ii) are not independent events. Platelet stimulation through the P2Y(12) receptors provokes only transient alpha(IIb)beta(3) activation via signaling routes involving phosphoinositide 3-kinases and Rap1b. Furthermore, alpha(IIb)beta(3) can be secondarily inactivated in platelets with prolonged high Ca(2+) rises, which expose phosphatidylserine and bind coagulation factors. Thus, platelet stimulation with strong agonists (collagen and thrombin) also results in transient integrin activation. Integrin alpha(2)beta(1) is found to be activated by a mechanism that is directly linked to alpha(IIb)beta(3) activation. Integrin alpha(2)beta(1) can adopt different activation states, depending on the trigger. Conclusively, reversibility and synchrony of platelet integrin activation are newly identified mechanisms to restrict thrombus growth and to allow optimal coagulation factor binding. Back-shifting of activated integrins towards their resting state may be a novel goal of antithrombotic medication.


Assuntos
Plaquetas/fisiologia , Integrinas/sangue , Animais , Humanos , Integrina alfa2beta1/sangue , Integrina alfa2beta1/química , Integrina alfa2beta1/fisiologia , Integrinas/química , Integrinas/fisiologia , Ativação Plaquetária/fisiologia , Adesividade Plaquetária/fisiologia , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Conformação Proteica , Receptores Purinérgicos P2/sangue , Receptores Purinérgicos P2/fisiologia , Receptores Purinérgicos P2Y12 , Transdução de Sinais , Trombose/sangue , Trombose/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...