Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Platelets ; 34(1): 2224454, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37313659

RESUMO

The anucleate human platelets contain a broad pattern of mRNAs and other RNA transcripts. The high quantitative similarity of mRNAs in megakaryocytes and platelets from different sources points to a common origin, and suggests a random redistribution of mRNA species upon proplatelet formation. A comparison of the classified platelet transcriptome (17.6k transcripts) with the identified platelet proteome (5.2k proteins) indicates an under-representation of: (i) nuclear but not of other organellar proteins; (ii) membrane receptors and channels with low transcript levels; (iii) transcription/translation proteins; and (iv) so far uncharacterized proteins. In this review, we discuss the technical, normalization and database-dependent possibilities and challenges to come to a complete, genome-wide platelet transcriptome and proteome. Such a reference transcriptome and proteome can serve to further elucidate intra-subject and inter-subject differences in platelets in health and disease. Applications may also lay in the aid of genetic diagnostics.


Blood platelets contain thousands types of mRNAs and proteins. The mRNA composition is similar to the mRNAs of megakaryocytes, from which platelets are derived, suggesting a random redistribution upon platelet formation. First attempts to identify all classified platelet proteins from mass spectral analysis used the genome-wide information of all mRNA types. This analysis revealed that the so far absent proteins in platelets are especially located in the megakaryocyte nucleus, or have low mRNA levels or low copy numbers. In this review, we discuss the possibilities to come to subject-dependent identification of the platelet protein and mRNA composition. Future applications may aid the genetic diagnostics.


Assuntos
Plaquetas , Proteoma , Humanos , Transcriptoma , Megacariócitos , Bases de Dados Factuais , RNA Mensageiro
2.
J Proteome Res ; 21(4): 1181-1188, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35316605

RESUMO

As novel liquid chromatography-mass spectrometry (LC-MS) technologies for proteomics offer a substantial increase in LC-MS runs per day, robust and reproducible sample preparation emerges as a new bottleneck for throughput. We introduce a novel strategy for positive-pressure 96-well filter-aided sample preparation (PF96) on a commercial positive-pressure solid-phase extraction device. PF96 allows for a five-fold increase in throughput in conjunction with extraordinary reproducibility with Pearson product-moment correlations on the protein level of r = 0.9993, as demonstrated for mouse heart tissue lysate in 40 technical replicates. The targeted quantification of 16 peptides in the presence of stable-isotope-labeled reference peptides confirms that PF96 variance is barely assessable against technical variation from nanoLC-MS instrumentation. We further demonstrate that protein loads of 36-60 µg result in optimal peptide recovery, but lower amounts ≥3 µg can also be processed reproducibly. In summary, the reproducibility, simplicity, and economy of time provide PF96 a promising future in biomedical and clinical research.


Assuntos
Peptídeos , Proteômica , Animais , Cromatografia Líquida/métodos , Humanos , Espectrometria de Massas/métodos , Camundongos , Peptídeos/análise , Proteômica/métodos , Reprodutibilidade dos Testes
3.
BMC Cancer ; 22(1): 653, 2022 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-35698081

RESUMO

BACKGROUND: Tyrosine kinase inhibitors (TKIs), such as sunitinib, are used for cancer treatment, but may also affect platelet count and function with possible hemostatic consequences. Here, we investigated whether patient treatment with the TKI sunitinib affected quantitative and qualitative platelet traits as a function of the sunitinib level and the occurrence of bleeding. METHODS: Blood was collected from 20 metastatic renal cell carcinoma (mRCC) patients before treatment, and at 2 weeks, 4 weeks and 3 months after sunitinib administration. We measured blood cell counts, platelet aggregation, and concentrations of sunitinib as well as its N-desethyl metabolite in plasma, serum and isolated platelets. Progression of disease (PD) and bleeding were monitored after 3 months. RESULTS: In sunitinib-treated mRCC patients, concentrations of (N-desethyl-)sunitinib in plasma and serum were highly correlated. In the patients' platelets the active metabolite levels were relatively increased as compared to sunitinib. On average, a sustained reduction in platelet count was observed on-treatment, which was significantly related to the inhibitor levels in plasma/serum. Principal component and correlational analysis showed that the (N-desethyl-)sunitinib levels in plasma/serum were linked to a reduction in both platelet count and collagen-induced platelet aggregation. The reduced aggregation associated in part with reported bleeding, but did not correlate to PD. CONCLUSION: The sunitinib-induced reduction in quantitative and qualitative platelet traits may reflect the effective sunitinib levels in the patient. These novel results may serve as a proof-of-principle for other TKI-related drugs, where both platelet count and functions are affected, which could be used for therapeutic drug monitoring.


Assuntos
Antineoplásicos , Carcinoma de Células Renais , Neoplasias Renais , Antineoplásicos/efeitos adversos , Plaquetas/patologia , Carcinoma de Células Renais/patologia , Humanos , Indóis/efeitos adversos , Neoplasias Renais/patologia , Pirróis/efeitos adversos , Sunitinibe/uso terapêutico
4.
Arterioscler Thromb Vasc Biol ; 41(2): e97-e111, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33267658

RESUMO

OBJECTIVE: Fibrin is considered to strengthen thrombus formation via integrin αIIbß3, but recent findings indicate that fibrin can also act as ligand for platelet glycoprotein VI. Approach and Results: To investigate the thrombus-forming potential of fibrin and the roles of platelet receptors herein, we generated a range of immobilized fibrin surfaces, some of which were cross-linked with factor XIIIa and contained VWF-BP (von Willebrand factor-binding peptide). Multicolor microfluidics assays with whole-blood flowed at high shear rate (1000 s-1) indicated that the fibrin surfaces, regardless of the presence of factor XIIIa or VWF-BP, supported platelet adhesion and activation (P-selectin expression), but only microthrombi were formed consisting of bilayers of platelets. Fibrinogen surfaces produced similar microthrombi. Markedly, tiggering of coagulation with tissue factor or blocking of thrombin no more than moderately affected the fibrin-induced microthrombus formation. Absence of αIIbß3 in Glanzmann thrombasthenia annulled platelet adhesion. Blocking of glycoprotein VI with Fab 9O12 substantially, but incompletely reduced platelet secretion, Ca2+ signaling and aggregation, while inhibition of Syk further reduced these responses. In platelet suspension, glycoprotein VI blockage or Syk inhibition prevented fibrin-induced platelet aggregation. Microthrombi on fibrin surfaces triggered only minimal thrombin generation, in spite of thrombin binding to the fibrin fibers. CONCLUSIONS: Together, these results indicate that fibrin fibers, regardless of their way of formation, act as a consolidating surface in microthrombus formation via nonredundant roles of platelet glycoprotein VI and integrin αIIbß3 through signaling via Syk and low-level Ca2+ rises.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Fibrina/metabolismo , Adesividade Plaquetária , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Trombose/sangue , Plaquetas/ultraestrutura , Sinalização do Cálcio , Estudos de Casos e Controles , Feminino , Fibrina/ultraestrutura , Humanos , Masculino , Técnicas Analíticas Microfluídicas , Quinase Syk/sangue , Trombastenia/sangue , Trombose/patologia
5.
Int J Mol Sci ; 23(20)2022 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-36293367

RESUMO

Integrin αIIbß3 activation is essential for platelet aggregation and, accordingly, for hemostasis and arterial thrombosis. The αIIbß3 integrin is highly expressed on platelets and requires an activation step for binding to fibrinogen, fibrin or von Willebrand factor (VWF). A current model assumes that the process of integrin activation relies on actomyosin force-dependent molecular changes from a bent-closed and extended-closed to an extended-open conformation. In this paper we review the pathways that point to a functional reversibility of platelet αIIbß3 activation and transient aggregation. Furthermore, we refer to mouse models indicating that genetic defects that lead to reversible platelet aggregation can also cause instable thrombus formation. We discuss the platelet agonists and signaling pathways that lead to a transient binding of ligands to integrin αIIbß3. Our analysis points to the (autocrine) ADP P2Y1 and P2Y12 receptor signaling via phosphoinositide 3-kinases and Akt as principal pathways linked to reversible integrin activation. Downstream signaling events by protein kinase C, CalDAG-GEFI and Rap1b have not been linked to transient integrin activation. Insight into the functional reversibility of integrin activation pathways will help to better understand the effects of antiplatelet agents.


Assuntos
Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Trombose , Camundongos , Animais , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Fator de von Willebrand/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Inibidores da Agregação Plaquetária/metabolismo , Actomiosina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ativação Plaquetária , Agregação Plaquetária , Plaquetas/metabolismo , Trombose/metabolismo , Fibrinogênio/metabolismo , Proteína Quinase C/metabolismo , Difosfato de Adenosina/metabolismo , Fibrina/metabolismo , Fosfatidilinositóis/metabolismo
6.
Blood ; 132(11): 1180-1192, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30018077

RESUMO

AMP-activated protein kinase (AMPK) α1 is activated in platelets on thrombin or collagen stimulation, and as a consequence, phosphorylates and inhibits acetyl-CoA carboxylase (ACC). Because ACC is crucial for the synthesis of fatty acids, which are essential for platelet activation, we hypothesized that this enzyme plays a central regulatory role in platelet function. To investigate this, we used a double knock-in (DKI) mouse model in which the AMPK phosphorylation sites Ser79 on ACC1 and Ser212 on ACC2 were mutated to prevent AMPK signaling to ACC. Suppression of ACC phosphorylation promoted injury-induced arterial thrombosis in vivo and enhanced thrombus growth ex vivo on collagen-coated surfaces under flow. After collagen stimulation, loss of AMPK-ACC signaling was associated with amplified thromboxane generation and dense granule secretion. ACC DKI platelets had increased arachidonic acid-containing phosphatidylethanolamine plasmalogen lipids. In conclusion, AMPK-ACC signaling is coupled to the control of thrombosis by specifically modulating thromboxane and granule release in response to collagen. It appears to achieve this by increasing platelet phospholipid content required for the generation of arachidonic acid, a key mediator of platelet activation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Acetil-CoA Carboxilase/metabolismo , Plaquetas/enzimologia , Transdução de Sinais , Trombose/enzimologia , Proteínas Quinases Ativadas por AMP/genética , Acetil-CoA Carboxilase/genética , Animais , Plaquetas/patologia , Técnicas de Introdução de Genes , Camundongos , Camundongos Knockout , Fosforilação/genética , Trombose/genética , Trombose/patologia
7.
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
8.
Haematologica ; 104(6): 1256-1267, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30545925

RESUMO

In combination with microspotting, whole-blood microfluidics can provide high-throughput information on multiple platelet functions in thrombus formation. Based on assessment of the inter- and intra-subject variability in parameters of microspot-based thrombus formation, we aimed to determine the platelet factors contributing to this variation. Blood samples from 94 genotyped healthy subjects were analyzed for conventional platelet phenotyping: i.e. hematologic parameters, platelet glycoprotein (GP) expression levels and activation markers (24 parameters). Furthermore, platelets were activated by ADP, CRP-XL or TRAP. Parallel samples were investigated for whole-blood thrombus formation (6 microspots, providing 48 parameters of adhesion, aggregation and activation). Microspots triggered platelet activation through GP Ib-V-IX, GPVI, CLEC-2 and integrins. For most thrombus parameters, inter-subject variation was 2-4 times higher than the intra-subject variation. Principal component analyses indicated coherence between the majority of parameters for the GPVI-dependent microspots, partly linked to hematologic parameters, and glycoprotein expression levels. Prediction models identified parameters per microspot that were linked to variation in agonist-induced αIIbß3 activation and secretion. Common sequence variation of GP6 and FCER1G, associated with GPVI-induced αIIbß3 activation and secretion, affected parameters of GPVI-and CLEC-2-dependent thrombus formation. Subsequent analysis of blood samples from patients with Glanzmann thrombasthenia or storage pool disease revealed thrombus signatures of aggregation-dependent parameters that were subject-dependent, but not linked to GPVI activity. Taken together, this high-throughput elucidation of thrombus formation revealed patterns of inter-subject differences in platelet function, which were partly related to GPVI-induced activation and common genetic variance linked to GPVI, but also included a distinct platelet aggregation component.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária , Trombose/etiologia , Trombose/metabolismo , Biomarcadores , Citometria de Fluxo , Ensaios de Triagem em Larga Escala , Humanos , Imunofenotipagem , Agregação Plaquetária , Contagem de Plaquetas , Testes de Função Plaquetária , Glicoproteínas da Membrana de Plaquetas/metabolismo , Trombose/diagnóstico
9.
Expert Rev Proteomics ; 15(6): 467-476, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29787335

RESUMO

INTRODUCTION: Platelets are the smallest cells within the circulating blood with key roles in physiological hemostasis and pathological thrombosis regulated by the onset of activating/inhibiting processes via receptor responses and signaling cascades. Areas covered: Proteomics as well as genomic approaches have been fundamental in identifying and quantifying potential targets for future diagnostic strategies in the prevention of bleeding and thrombosis, and uncovering the complexity of platelet functions in health and disease. In this article, we provide a critical overview on current functional tests used in diagnostics and the future perspectives for platelet proteomics in clinical applications. Expert commentary: Proteomics represents a valuable tool for the identification of patients with diverse platelet associated defects. In-depth validation of identified biomarkers, e.g. receptors, signaling proteins, post-translational modifications, in large cohorts is decisive for translation into routine clinical diagnostics.


Assuntos
Biomarcadores/sangue , Plaquetas/metabolismo , Proteoma/genética , Trombose/sangue , Genômica , Humanos , Espectrometria de Massas , Processamento de Proteína Pós-Traducional/genética , Proteômica/tendências , Transdução de Sinais/genética , Trombose/genética
10.
Haematologica ; 103(9): 1557-1567, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29880611

RESUMO

Severe thrombocytopenia (≤50×109 platelets/L) due to hematological malignancy and intensive chemotherapy is associated with an increased risk of clinically significant bleeding. Since the bleeding risk is not linked to the platelet count only, other hemostatic factors must be involved. We studied platelet function in 77 patients with acute leukemia, multiple myeloma or malignant lymphoma, who experienced chemotherapy-induced thrombocytopenia. Platelets from all patients - independent of disease or treatment type - were to a variable extent compromised in Ca2+ flux, integrin a ß activation and P-selectin expression when stimulated with a panelIIbof3 agonists. The patients' platelets were also impaired in spreading on fibrinogen. Whereas the Ca2+ store content was unaffected, the patients' platelets showed ongoing phosphatidylserine exposure, which was not due to apoptotic caspase activity. Interestingly, mitochondrial function was markedly reduced in platelets from a representative subset of patients, as evidenced by a low mitochondrial membrane potential (P<0.001) and low oxygen consumption (P<0.05), while the mitochondrial content was normal. Moreover, the mitochondrial impairments coincided with elevated levels of reactive oxygen species (Spearman's rho=-0.459, P=0.012). Markedly, the impairment of platelet function only appeared after two days of chemotherapy, suggesting origination in the megakaryocytes. In patients with bone marrow recovery, platelet function improved. In conclusion, our findings disclose defective receptor signaling related to impaired mitochondrial bioenergetics, independent of apoptosis, in platelets from cancer patients treated with chemotherapy, explaining the low hemostatic potential of these patients.


Assuntos
Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias/complicações , Trombocitopenia/etiologia , Trombocitopenia/metabolismo , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Biomarcadores , Coagulação Sanguínea/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Estudos de Casos e Controles , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/sangue , Neoplasias/tratamento farmacológico , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Contagem de Plaquetas , Testes de Função Plaquetária , Índice de Gravidade de Doença , Trombocitopenia/sangue , Trombocitopenia/diagnóstico
11.
Mol Cell Proteomics ; 15(10): 3154-3169, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27535140

RESUMO

The Scott syndrome is a very rare and likely underdiagnosed bleeding disorder associated with mutations in the gene encoding anoctamin-6. Platelets from Scott patients are impaired in various Ca2+-dependent responses, including phosphatidylserine exposure, integrin closure, intracellular protein cleavage, and cytoskeleton-dependent morphological changes. Given the central role of anoctamin-6 in the platelet procoagulant response, we used quantitative proteomics to understand the underlying molecular mechanisms and the complex phenotypic changes in Scott platelets compared with control platelets. Therefore, we applied an iTRAQ-based multi-pronged strategy to quantify changes in (1) the global proteome, (2) the phosphoproteome, and (3) proteolytic events between resting and stimulated Scott and control platelets. Our data indicate a limited number of proteins with decreased (70) or increased (64) expression in Scott platelets, among those we confirmed the absence of anoctamin-6 and the strong up-regulation of aquaporin-1 by parallel reaction monitoring. The quantification of 1566 phosphopeptides revealed major differences between Scott and control platelets after stimulation with thrombin/convulxin or ionomycin. In Scott platelets, phosphorylation levels of proteins regulating cytoskeletal or signaling events were increased. Finally, we quantified 1596 N-terminal peptides in activated Scott and control platelets, 180 of which we identified as calpain-regulated, whereas a distinct set of 23 neo-N termini was caspase-regulated. In Scott platelets, calpain-induced cleavage of cytoskeleton-linked and signaling proteins was downregulated, in accordance with an increased phosphorylation state. Thus, multipronged proteomic profiling of Scott platelets provides detailed insight into their protection against detrimental Ca2+-dependent changes that are normally associated with phosphatidylserine exposure.


Assuntos
Transtornos da Coagulação Sanguínea/sangue , Plaquetas/fisiologia , Fosfoproteínas/análise , Proteômica/métodos , Transtornos da Coagulação Sanguínea/metabolismo , Plaquetas/efeitos dos fármacos , Cálcio/metabolismo , Venenos de Crotalídeos/farmacologia , Regulação da Expressão Gênica , Humanos , Ionomicina/farmacologia , Lectinas Tipo C , Fosfoproteínas/efeitos dos fármacos , Proteólise , Transdução de Sinais , Trombina/farmacologia
12.
Blood ; 125(16): 2568-78, 2015 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-25712989

RESUMO

The interaction of plasminogen with platelets and their localization during thrombus formation and fibrinolysis under flow are not defined. Using a novel model of whole blood thrombi, formed under flow, we examine dose-dependent fibrinolysis using fluorescence microscopy. Fibrinolysis was dependent upon flow and the balance between fibrin formation and plasminogen activation, with tissue plasminogen activator-mediated lysis being more efficient than urokinase plasminogen activator-mediated lysis. Fluorescently labeled plasminogen radiates from platelet aggregates at the base of thrombi, primarily in association with fibrin. Hirudin attenuates, but does not abolish plasminogen binding, denoting the importance of fibrin. Flow cytometry revealed that stimulation of platelets with thrombin/convulxin significantly increased the plasminogen signal associated with phosphatidylserine (PS)-exposing platelets. Binding was attenuated by tirofiban and Gly-Pro-Arg-Pro amide, confirming a role for fibrin in amplifying plasminogen binding to PS-exposing platelets. Confocal microscopy revealed direct binding of plasminogen and fibrinogen to different platelet subpopulations. Binding of plasminogen and fibrinogen co-localized with PAC-1 in the center of spread platelets. In contrast, PS-exposing platelets were PAC-1 negative, and bound plasminogen and fibrinogen in a protruding "cap." These data show that different subpopulations of platelets harbor plasminogen by diverse mechanisms and provide an essential scaffold for the accumulation of fibrinolytic proteins that mediate fibrinolysis under flow.


Assuntos
Plaquetas/metabolismo , Fibrinolisina/metabolismo , Fosfatidilserinas/metabolismo , Trombose/metabolismo , Plaquetas/efeitos dos fármacos , Venenos de Crotalídeos/farmacologia , Fibrina/metabolismo , Fibrinólise/efeitos dos fármacos , Fibrinolíticos/farmacologia , Citometria de Fluxo , Hemostáticos/farmacologia , Hirudinas/farmacologia , Lectinas Tipo C , Microscopia Confocal , Oligopeptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Reologia , Resistência ao Cisalhamento , Trombina/farmacologia , Tirofibana , Ativador de Plasminogênio Tecidual/metabolismo , Tirosina/análogos & derivados , Tirosina/farmacologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 36(11): 2152-2157, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27634832

RESUMO

OBJECTIVE: It is known that both platelets and coagulation strongly influence infarct progression after ischemic stroke, but the mechanisms and their interplay are unknown. Our aim was to assess the contribution of the procoagulant platelet surface, and thus platelet-driven thrombin generation, to the progression of thromboinflammation in the ischemic brain. APPROACH AND RESULTS: We present the characterization of a novel platelet and megakaryocyte-specific TMEM16F (anoctamin 6) knockout mouse. Reflecting Scott syndrome, platelets from the knockout mouse had a significant reduction in procoagulant characteristics that altered thrombin and fibrin generation kinetics. In addition, knockout mice showed significant defects in hemostasis and arterial thrombus formation. However, infarct volumes in a model of ischemic stroke were comparable with wild-type mice. CONCLUSIONS: Platelet TMEM16F activity contributes significantly to hemostasis and thrombosis but not cerebral thromboinflammation. These results highlight another key difference between the roles of platelets and coagulation in these processes.


Assuntos
Plaquetas/metabolismo , Doenças das Artérias Carótidas/sangue , Encefalite/sangue , Encefalite/genética , Hemostasia , Infarto da Artéria Cerebral Média/sangue , Fosfatidilserinas/sangue , Proteínas de Transferência de Fosfolipídeos/sangue , Trombina/metabolismo , Trombose/sangue , Animais , Anoctaminas , Coagulação Sanguínea , Doenças das Artérias Carótidas/genética , Doenças das Artérias Carótidas/patologia , Modelos Animais de Doenças , Encefalite/patologia , Fibrina/metabolismo , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Cinética , Megacariócitos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas de Transferência de Fosfolipídeos/deficiência , Proteínas de Transferência de Fosfolipídeos/genética , Ativação Plaquetária , Transdução de Sinais , Trombose/genética , Trombose/patologia
14.
Arterioscler Thromb Vasc Biol ; 36(4): 692-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26848157

RESUMO

OBJECTIVE: Platelet- and fibrin-dependent thrombus formation is regulated by blood flow and exposure of collagen and tissue factor. However, interactions between these blood-borne and vascular components are not well understood. APPROACH AND RESULTS: Here, we developed a method to assess whole-blood thrombus formation on microspots with defined amounts of collagen and tissue factor, allowing determination of the mechanical properties and intrathrombus composition. Confining the collagen content resulted in diminished platelet deposition and fibrin formation at high shear flow conditions, but this effect was compensated by a larger thrombus size and increased accumulation of fibrin in the luminal regions of the thrombi at the expense of the base regions. These thrombi were more dependent on tissue factor-triggered thrombin generation. Microforce nanoindentation analysis revealed a significantly increased microelasticity of thrombi with luminal-oriented fibrin. At a low shear rate, fibrin fibers tended to luminally cover the thrombi, again resulting in a higher microelasticity. Studies with blood from patients with distinct hemostatic insufficiencies indicated an impairment in the formation of a platelet-fibrin thrombus in the cases of dilutional coagulopathy, thrombocytopenia, Scott syndrome, and hemophilia B. CONCLUSIONS: Taken together, our data indicate that (1) thrombin increases the platelet thrombus volume; (2) tissue factor drives the formation of fibrin outside of the platelet thrombus; (3) limitation of platelet adhesion redirects fibrin from bottom to top of the thrombus; (4) a lower shear rate promotes thrombus coverage with fibrin; (5) the fibrin distribution pattern determines thrombus microelasticity; and (6) the thrombus-forming process is reduced in patients with diverse hemostatic defects.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Fibrina/metabolismo , Trombose/sangue , Transtornos da Coagulação Sanguínea/sangue , Transtornos da Coagulação Sanguínea/fisiopatologia , Testes de Coagulação Sanguínea , Velocidade do Fluxo Sanguíneo , Estudos de Casos e Controles , Colágeno/sangue , Elasticidade , Hemofilia B/sangue , Hemofilia B/fisiopatologia , Humanos , Fluxo Sanguíneo Regional , Trombocitopenia/sangue , Trombocitopenia/fisiopatologia , Tromboplastina/metabolismo , Trombose/fisiopatologia , Fatores de Tempo
15.
Platelets ; 28(5): 441-448, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28358995

RESUMO

This paper provides an overview of the various types of microfluidic devices that are employed to study the complex processes of platelet activation and blood coagulation in whole blood under flow conditions. We elaborate on how these devices are used to detect impaired platelet-dependent fibrin formation in blood from mice or patients with specific bleeding disorders. We provide a practical guide on how to assess formation of a platelet-fibrin thrombus under flow, using equipment that is present in most laboratories. In addition, we describe current insights on how blood flow and shear rate alter the location of platelet populations, von Willebrand factor, coagulation factors, and fibrin in a growing thrombus. Finally, we discuss possibilities and limitations for the clinical use of microfluidic devices to evaluate a hemostatic or prothrombotic tendency in patient blood samples.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Dispositivos Lab-On-A-Chip , Técnicas Analíticas Microfluídicas/métodos , Trombose/metabolismo , Animais , Fatores de Coagulação Sanguínea/metabolismo , Plaquetas/patologia , Fibrina/metabolismo , Humanos , Trombose/patologia
16.
Haematologica ; 101(4): 427-36, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26721892

RESUMO

Coated platelets, formed by collagen and thrombin activation, have been characterized in different ways: i) by the formation of a protein coat of α-granular proteins; ii) by exposure of procoagulant phosphatidylserine; or iii) by high fibrinogen binding. Yet, their functional role has remained unclear. Here we used a novel transglutaminase probe, Rhod-A14, to identify a subpopulation of platelets with a cross-linked protein coat, and compared this with other platelet subpopulations using a panel of functional assays. Platelet stimulation with convulxin/thrombin resulted in initial integrin α(IIb)ß3 activation, the appearance of a platelet population with high fibrinogen binding, (independently of active integrins, but dependent on the presence of thrombin) followed by phosphatidylserine exposure and binding of coagulation factors Va and Xa. A subpopulation of phosphatidylserine-exposing platelets bound Rhod-A14 both in suspension and in thrombi generated on a collagen surface. In suspension, high fibrinogen and Rhod-A14 binding were antagonized by combined inhibition of transglutaminase activity and integrin α(IIb)ß3 Markedly, in thrombi from mice deficient in transglutaminase factor XIII, platelet-driven fibrin formation and Rhod-A14 binding were abolished by blockage of integrin α(IIb)ß3. Vice versa, star-like fibrin formation from platelets of a patient with deficiency in α(IIb)ß3(Glanzmann thrombasthenia) was abolished upon blockage of transglutaminase activity. We conclude that coated platelets, with initial α(IIb)ß3 activation and high fibrinogen binding, form a subpopulation of phosphatidylserine-exposing platelets, and function in platelet-dependent star-like fibrin fiber formation via transglutaminase factor XIII and integrin α(IIb)ß3.


Assuntos
Plaquetas/metabolismo , Fator XIII/metabolismo , Fibrina/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombastenia/sangue , Animais , Coagulação Sanguínea , Plaquetas/efeitos dos fármacos , Plaquetas/patologia , Venenos de Crotalídeos/farmacologia , Fator Va/química , Fator Va/metabolismo , Fator XIII/química , Fator Xa/química , Fator Xa/metabolismo , Fibrina/química , Fibrinogênio/química , Fibrinogênio/metabolismo , Humanos , Lectinas Tipo C , Camundongos , Camundongos Knockout , Sondas Moleculares/química , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Cultura Primária de Células , Ligação Proteica , Trombastenia/patologia , Trombina/farmacologia
17.
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
18.
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
19.
Haematologica ; 100(6): 748-56, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25769543

RESUMO

The importance of factor Xa generation in thrombus formation has not been studied extensively so far. Here, we used mice deficient in either factor VIII or factor IX to determine the role of platelet-stimulated tenase activity in the formation of platelet-fibrin thrombi on collagen. With tissue factor present, deficiency in factor VIII or IX markedly suppressed thrombus growth, fibrin formation and platelet procoagulant activity (phosphatidylserine exposure). In either case, residual fibrin formation was eliminated in the absence of tissue factor. Effects of factor deficiencies were antagonized by supplementation of the missing coagulation factor. In wild-type thrombi generated under flow, phosphatidylserine-exposing platelets bound (activated) factor IX and factor X, whereas factor VIII preferentially co-localized at sites of von Willebrand factor binding. Furthermore, proteolytic activity of the generated activated factor X and thrombin was confined to the sites of phosphatidylserine exposure. With blood from a hemophilia A or B patient, the formation of platelet-fibrin thrombi was greatly delayed and reduced, even in the presence of high concentrations of tissue factor. A direct activated factor X inhibitor, rivaroxaban, added to human blood, suppressed both thrombin and fibrin formation. Together, these data point to a potent enforcement loop in thrombus formation due to factor X activation, subsequent thrombin and fibrin generation, causing activated factor X-mediated stimulation of platelet phosphatidylserine exposure. This implies that the factor VIII/factor IX-dependent stimulation of platelet procoagulant activity is a limiting factor for fibrin formation under flow conditions, even at high tissue factor concentrations.


Assuntos
Coagulação Sanguínea/fisiologia , Plaquetas/metabolismo , Cisteína Endopeptidases/fisiologia , Fibrina/metabolismo , Proteínas de Neoplasias/fisiologia , Trombose/sangue , Animais , Plaquetas/patologia , Fator IXa/metabolismo , Fator VIIIa/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Trombose/patologia
20.
Haematologica ; 100(9): 1131-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26113418

RESUMO

In patients with acute coronary syndrome, dual antiplatelet therapy with aspirin and a P2Y12 inhibitor like prasugrel is prescribed for one year. Here, we investigated how the hemostatic function of platelets recovers after discontinuation of prasugrel treatment. Therefore, 16 patients who suffered from ST-elevation myocardial infarction were investigated. Patients were treated with aspirin (100 mg/day, long-term) and stopped taking prasugrel (10 mg/day) after one year. Blood was collected at the last day of prasugrel intake and at 1, 2, 5, 12 and 30 days later. Platelet function in response to ADP was normalized between five and 30 days after treatment cessation and in vitro addition of the reversible P2Y12 receptor antagonist ticagrelor fully suppressed the regained activation response. Discontinuation of prasugrel resulted in the formation of an emerging subpopulation of ADP-responsive platelets, exhibiting high expression of active integrin αIIbß3. Two different mRNA probes, thiazole orange and the novel 5'Cy5-oligo-dT probe revealed that this subpopulation consisted of juvenile platelets, which progressively contributed to platelet aggregation and thrombus formation under flow. During offset, juvenile platelets were overall more reactive than older platelets. Interestingly, the responsiveness of both juvenile and older platelets increased in time, pointing towards a residual inhibitory effect of prasugrel on the megakaryocyte level. In conclusion, the gradual increase in thrombogenicity after cessation of prasugrel treatment is due to the increased activity of juvenile platelets.


Assuntos
Síndrome Coronariana Aguda/tratamento farmacológico , Síndrome Coronariana Aguda/metabolismo , Coagulação Sanguínea/efeitos dos fármacos , Plaquetas/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Cloridrato de Prasugrel/administração & dosagem , Difosfato de Adenosina/metabolismo , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Testes de Função Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Receptores Purinérgicos P2Y12/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA