Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Heart Vessels ; 37(11): 1962-1970, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35545686

RESUMO

Transforming growth factor-ß-induced protein (TGFBIp) is released from activated platelets and promotes pro-thrombotic complications like pulmonary embolism. The role of TGFBIp in acute coronary syndrome, especially with a focus on platelets, has not been investigated so far. Using ELISA and immunoblotting, we demonstrate platelet TGFBIp release in patients with myocardial infarction (MI). We investigated TGFBIp-induced platelet adhesion and rolling by flow chamber and chemotactic effects of TGFBIp in transwell experiments. Immunochemistry staining of arterial vessels detected TGFBIp and the platelet-specific protein GPVI in the vessel wall.We demonstrate for the first time that platelet TGFBIp release is significantly increased in MI and correlates with the severity of acute coronary syndromes (STEMI, NSTEMI). After activation with TRAP, platelets release TGFBIp and TGFBIp itself activates platelets. Under flow, TGFBIp-mediated platelet rolling and adherence similarly to collagen. TGFBIp significantly increased platelet transmigration and we demonstrate TGFBIp deposits in the wall of human arteries. In this study, we add novel aspects to the role of TGFBIp in acute coronary syndrome by demonstrating that TGFBIp is partially released from platelets during MI and has activating, pro-adhesive and pro-migratory effects on platelets that could contribute to the disease development of coronary vascular inflammation and MI.


Assuntos
Síndrome Coronariana Aguda , Proteínas da Matriz Extracelular/sangue , Infarto do Miocárdio , Fator de Crescimento Transformador beta/sangue , Plaquetas/metabolismo , Colágeno/metabolismo , Humanos , Infarto do Miocárdio/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fatores de Crescimento Transformadores/metabolismo
2.
Int J Mol Sci ; 21(21)2020 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-33142786

RESUMO

Platelets are exposed to extracellular matrix (ECM) proteins like collagen and laminin and to fibrinogen during acute vascular events. However, beyond hemostasis, platelets have the important capacity to migrate on ECM surfaces, but the translational response of platelets to different extracellular matrix stimuli is still not fully characterized. Using 2D-gel electrophoresis, confocal microscopy, polysome analysis and protein sequencing by mass spectrometry, we demonstrate that platelets show a differential expression profile of newly synthesized proteins on laminin, collagen or fibrinogen. In this context, we observed a characteristic, ECM-dependent translocation phenotype of translation initiation factor eIF4E to the ribosomal site. eIF4E accumulated in polysomes with increased binding of mRNA and co-localization with vinculin, leading to de novo synthesis of important cytoskeletal regulator proteins. As the first study, we included a proteome analysis of laminin-adherent platelets and interestingly identified upregulation of essentially important proteins that mediate cytoskeletal regulation and mobility in platelets, such as filamin A, talin, vinculin, gelsolin, coronin or kindlin-3. In summary, we demonstrate that platelet activation with extracellular matrix proteins results in a distinct stimulus-specific translational response of platelets that will help to improve our understanding of the regulation of platelet mobility and migration.


Assuntos
Plaquetas/fisiologia , Proteínas do Citoesqueleto/metabolismo , Matriz Extracelular/metabolismo , Ativação Plaquetária , Biossíntese de Proteínas , Processamento de Proteína Pós-Traducional , Plaquetas/citologia , Colágeno/metabolismo , Citoesqueleto/metabolismo , Fator de Iniciação 4E em Eucariotos/metabolismo , Fibrinogênio/metabolismo , Hemostasia , Humanos
3.
Int J Mol Sci ; 20(24)2019 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-31817594

RESUMO

Kindlins are important proteins for integrin signaling and regulation of the cytoskeleton, but we know little about their precise function and regulation in platelets during acute ischemic events. In this work, we investigated kindlin-3 protein levels in platelets isolated from patients with ST-elevation myocardial infarction (STEMI) compared to patients with non-ischemic chest pain. Platelets from twelve patients with STEMI and twelve patients with non-ischemic chest pain were isolated and analyzed for kindlin-3 protein levels and intracellular localization by immunoblotting and two-dimensional gel electrophoresis. Platelet proteome analysis by two-dimensional gel electrophoresis and protein sequencing identified kindlin-3 as a protein that is cleaved in platelets from patients with myocardial infarction. Kindlin-3 full-length protein was significantly decreased in patients with STEMI compared to patients with non-ischemic chest pain (1.0 ± 0.2 versus 0.28 ± 0.2, p < 0.05) by immunoblotting. Kindlin-3 showed a differential distribution and was primarily cleaved in the cytosolic and membrane compartment of platelets in myocardial infarction. Platelet activation with thrombin alone did not affect kindlin-3 protein levels. The present study demonstrates that kindlin-3 protein levels become significantly reduced in platelets of patients with myocardial infarction compared to controls. The results suggest that kindlin-3 cleavage in platelets is associated with the ischemic event of myocardial infarction.


Assuntos
Plaquetas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Infarto do Miocárdio com Supradesnível do Segmento ST/metabolismo , Idoso , Plaquetas/efeitos dos fármacos , Eletroforese em Gel Bidimensional , Feminino , Humanos , Immunoblotting , Masculino , Pessoa de Meia-Idade , Trombina/farmacologia
4.
Int J Mol Sci ; 20(23)2019 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-31783528

RESUMO

Heat-shock proteins are a family of proteins which are upregulated in response to stress stimuli including inflammation, oxidative stress, or ischemia. Protective functions of heat-shock proteins have been studied in vascular disease models, and malfunction of heat-shock proteins is associated with vascular disease development. Heat-shock proteins however have not been investigated in human platelets during acute myocardial infarction ex vivo. Using two-dimensional electrophoresis and immunoblotting, we observed that heat-shock protein 27 (HSPB1) levels and phosphorylation are significantly increased in platelets of twelve patients with myocardial infarction compared to patients with nonischemic chest pain (6.4 ± 1.0-fold versus 1.0 ± 0.9-fold and 5.9 ± 1.8-fold versus 1.0 ± 0.8-fold; p < 0.05). HSP27 (HSPB1) showed a distinct and characteristic intracellular translocation from the cytoskeletal fraction into the membrane fraction of platelets during acute myocardial infarction that did not occur in the control group. In this study, we could demonstrate for the first time that HSP27 (HSPB1) is upregulated and phosphorylated in human platelets during myocardial infarction on a cellular level ex vivo with a characteristic intracellular translocation pattern. This HSP27 (HSPB1) phenotype in platelets could thus represent a measurable stress response in myocardial infarction and potentially other acute ischemic events.


Assuntos
Plaquetas/metabolismo , Proteínas de Choque Térmico/genética , Chaperonas Moleculares/genética , Fosforilação/genética , Infarto do Miocárdio com Supradesnível do Segmento ST/genética , Regulação para Cima/genética , Citoesqueleto/genética , Feminino , Proteínas de Choque Térmico HSP27/genética , Resposta ao Choque Térmico/genética , Humanos , Masculino , Pessoa de Meia-Idade , Estresse Oxidativo/genética , Ativação Transcricional/genética
5.
PLoS Pathog ; 7(11): e1002355, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22102811

RESUMO

Human ß-defensins (hBD) are antimicrobial peptides that curb microbial activity. Although hBD's are primarily expressed by epithelial cells, we show that human platelets express hBD-1 that has both predicted and novel antibacterial activities. We observed that activated platelets surround Staphylococcus aureus (S. aureus), forcing the pathogens into clusters that have a reduced growth rate compared to S. aureus alone. Given the microbicidal activity of ß-defensins, we determined whether hBD family members were present in platelets and found mRNA and protein for hBD-1. We also established that hBD-1 protein resided in extragranular cytoplasmic compartments of platelets. Consistent with this localization pattern, agonists that elicit granular secretion by platelets did not readily induce hBD-1 release. Nevertheless, platelets released hBD-1 when they were stimulated by α-toxin, a S. aureus product that permeabilizes target cells. Platelet-derived hBD-1 significantly impaired the growth of clinical strains of S. aureus. hBD-1 also induced robust neutrophil extracellular trap (NET) formation by target polymorphonuclear leukocytes (PMNs), which is a novel antimicrobial function of ß-defensins that was not previously identified. Taken together, these data demonstrate that hBD-1 is a previously-unrecognized component of platelets that displays classic antimicrobial activity and, in addition, signals PMNs to extrude DNA lattices that capture and kill bacteria.


Assuntos
Toxinas Bacterianas/imunologia , Plaquetas/metabolismo , Proteínas Hemolisinas/imunologia , Neutrófilos/imunologia , Staphylococcus aureus/imunologia , beta-Defensinas/metabolismo , Toxinas Bacterianas/metabolismo , Plaquetas/enzimologia , Plaquetas/imunologia , Linhagem Celular Tumoral , Células HeLa , Proteínas Hemolisinas/metabolismo , Humanos , Neutrófilos/metabolismo , Ativação Plaquetária , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transdução de Sinais , Staphylococcus aureus/crescimento & desenvolvimento , beta-Defensinas/genética
6.
Cell Physiol Biochem ; 30(1): 259-68, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22759972

RESUMO

Recent observations pointed to the ability of platelets to migrate and thus to invade the inflamed vascular wall. Platelet migration could be stimulated by stromal cell-derived factor-1 (SDF-1), an effect dependent on phosphatidylinositide-3-kinase (PI3K) and paralleled by activation and phosphorylation of Wiskott-Aldrich syndrome protein (WASP). Migration is inhibited by vinculin, which is similarly regulated by phosphorylation. PI3K-sensitive kinases include the serum- and glucocorticoid-inducible kinase 1 (SGK1). The present study explored whether SGK1 modifies WASP and vinculin phosphorylation in murine platelets and participates in the regulation of platelet migration. Platelets were isolated from gene-targeted mice lacking SGK1 (sgk1(-/-)) and from their wild type littermates (sgk1(+/+)). Platelet migration stimulated with SDF-1 was significantly less pronounced in sgk1(-/-)platelets than in sgk1(+/+) platelets. Moreover, SDF-1 significantly induced WASP phosphorylation, an effect again reduced in platelets lacking SGK1. Phosphorylation of vinculin was significantly enhanced in sgk1(-/-)platelets and was significantly reduced following treatment of platelets with Ca(2+) chelator BAPTA. Immunohistochemical analysis of in vivo experiments in intestinal vessels after vascular inflammation revealed that transmigration of platelets into inflamed vessel walls was significantly less pronounced in sgk1(-/-)than in sgk1(+/+) mice. In conclusion, SGK1 is a powerful regulator of platelet migration.


Assuntos
Plaquetas/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Migração Transendotelial e Transepitelial , Animais , Células Cultivadas , Quelantes/farmacologia , Quimiocina CXCL12/fisiologia , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Feminino , Proteínas Imediatamente Precoces/genética , Intestinos/irrigação sanguínea , Isquemia/metabolismo , Isquemia/patologia , Masculino , Camundongos , Camundongos Knockout , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Vinculina/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
7.
Platelets ; 23(1): 1-6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21671729

RESUMO

Vascular lesion formation and collagen presentation are key events leading to the development of vulnerable plaques. Glycoprotein VI (GPVI) significantly contributes to plaque-associated collagen binding and thrombus formation. The aim of this study was to image endothelial injury using fluorescence-labelled GPVI-Fc (Fc, fragment crystallized), a soluble form of GPVI that was generated by cloning and fusing GPVI to an Fc-domain, in an ex-vivo rabbit model. This study serves as a proof-of-principle study to demonstrate that GPVI-Fc is a useful tool for detecting endothelial damage. The carotid and femoral arteries and the aorta abdominalis were isolated from rabbits and perfused with phosphate buffered saline (PBS) to remove all blood, and a catheter was placed into the vessels in situ. Endothelial damage was achieved by pulling an inflated balloon approximately 1 inch through the vessels, while control vessels were not balloon-treated. After balloon deflation, the catheter was removed. Fluorescence-labelled GPVI-Fc (50 µg/mL) was injected into the injured and control intact vessels, and the opened vessels were sealed by clamps. After incubation, the vessels were rinsed with PBS, and optical imaging was performed to measure GPVI-Fc binding to injured endothelium. The optical data corresponding to the mean detected optical signal of the regions of interest were corrected by subtracting the mean data of the background fluorescence (arbitrary units). After denudation, fluorescence was enhanced in injured femoral and carotid arteries when compared to intact femoral (41.1 ± 17.5 vs. 14.6 ± 6.5; P = 0.021) and carotid (30.2 ± 7.6 vs. 7.9 ± 3.9; P = 0.005) arteries. This preclinical GPVI-Fc-based vascular lesion imaging approach may be the first step towards a method that allows identification of vascular lesions in vivo.


Assuntos
Lesões das Artérias Carótidas/patologia , Endotélio Vascular/patologia , Artéria Femoral/patologia , Angiofluoresceinografia/métodos , Fragmentos Fc das Imunoglobulinas/farmacologia , Glicoproteínas da Membrana de Plaquetas/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Animais , Artérias Carótidas/patologia , Endotélio Vascular/lesões , Feminino , Artéria Femoral/lesões , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/genética , Glicoproteínas da Membrana de Plaquetas/química , Glicoproteínas da Membrana de Plaquetas/genética , Coelhos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética
8.
Biochem Biophys Res Commun ; 415(1): 54-60, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-22005466

RESUMO

Platelets have been shown to migrate and thus to invade the vascular wall. Platelet migration is stimulated by SDF-1. In other cell types, migration is dependent on Ca(2+) entry via Ca(2+) channels. Ca(2+) influx is sensitive to cell membrane potential which is maintained by K(+) channel activity and/or Cl(-) channel activity. The present study explored the role of ion channels in the regulation of SDF-1 induced migration. Platelets were isolated from human volunteers as well as from gene targeted mice lacking the Ca(2+) activated K(+) channel SK4 (sk4(-/-)) and their wild type littermates (sk4(+/+)). According to confocal microscopy human platelets expressed the Ca(2+) channel Orai1 and the Ca(2+)-activated K(+) channel K(Ca)3.1 (SK4). SDF-1 (100 ng/ml) stimulated migration in human platelets, an effect blunted by Orai1 inhibitors 2-aminoethoxydiphenyl borate 2-APB (10 µM) and SKF-96365 (10 µM), by unspecific K(+) channel inhibitor TEA (30 mM), by SK4 specific K(+) channel blocker clotrimazole (10 µM), but not by Cl(-) channel inhibitor 5-nitro-2-(3-phenylpropylamino) benzoic acid NPPB (100 µM). Significant stimulation of migration by SDF-1 was further observed in sk4(+/+) platelets but was virtually absent in sk4(-/-) platelets. In conclusion, platelet migration requires activity of the Ca(2+) channel Orai1 and of the Ca(2+) activated K(+) channel SK4, but not of NPPB-sensitive Cl(-) channels.


Assuntos
Plaquetas/fisiologia , Movimento Celular , Quimiocina CXCL12/metabolismo , Canais Iônicos/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Canais de Cálcio/metabolismo , Canais de Cloreto/metabolismo , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Camundongos , Nitrobenzoatos/farmacologia , Proteína ORAI1
9.
Arterioscler Thromb Vasc Biol ; 30(6): 1127-36, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20378847

RESUMO

OBJECTIVE: To investigate the role of junctional adhesion molecule A (JAM-A) on adhesion and differentiation of human CD34(+) cells into endothelial progenitor cells. METHODS AND RESULTS: Tissue healing and vascular regeneration is a multistep process requiring firm adhesion of circulating progenitor cells to the vascular wall and their further differentiation into endothelial cells. The role of JAM-A in platelet-mediated adhesion of progenitor cells was investigated by adhesion assays in vitro and with the help of intravital fluorescence microscopy in mice. Preincubation of human CD34(+) progenitor cells with soluble JAM-A-Fc (sJAM-A-Fc) resulted in significantly decreased adhesion over immobilized platelets or inflammatory endothelium under high shear stress in vitro and after carotid ligation in vivo or ischemia/reperfusion injury in the microcirculation of mice. Human CD34(+) cells express JAM-A, as defined by flow cytometry and Western blot analysis. JAM-A mediates differentiation of CD34(+) cells to endothelial progenitor cells and facilitates CD34(+) cell-induced reendothelialization in vitro. Pretreatment of human CD34(+) cells with sJAM-A-Fc resulted in increased neointima formation 3 weeks after endothelial denudation in the carotid arteries of nonobese diabetic/severe combined immunodeficient mice. CONCLUSIONS: These results indicate that the expression of JAM-A on CD34(+) cells mediates adhesion to the vascular wall after injury and differentiation into endothelial progenitor cells, a mechanism potentially involved in vascular regeneration. Human CD34(+) cells express JAM-A, mediating their interaction with platelets and endothelial cells. Specifically, JAM-A expressed on human CD34(+) progenitor cells regulates their adhesion over immobilized platelets or inflammatory endothelium under high shear stress in vitro and after carotid ligation in vivo or ischemia/reperfusion injury in the microcirculation of mice. Moreover, it mediates differentiation of CD34(+) cells to endothelial progenitor cells and facilitates reendothelialization.


Assuntos
Antígenos CD34/análise , Lesões das Artérias Carótidas/metabolismo , Moléculas de Adesão Celular/metabolismo , Adesão Celular , Diferenciação Celular , Células Endoteliais/metabolismo , Imunoglobulinas/metabolismo , Intestinos/irrigação sanguínea , Traumatismo por Reperfusão/metabolismo , Células-Tronco/metabolismo , Animais , Plaquetas/metabolismo , Western Blotting , Células CHO , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/imunologia , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/fisiopatologia , Moléculas de Adesão Celular/genética , Proliferação de Células , Cricetinae , Cricetulus , Células Endoteliais/imunologia , Células Endoteliais/transplante , Citometria de Fluxo , Humanos , Fragmentos Fc das Imunoglobulinas/metabolismo , Imunoglobulinas/genética , Antígeno-1 Associado à Função Linfocitária/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia de Fluorescência , Microscopia de Vídeo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Receptores de Superfície Celular , Proteínas Recombinantes de Fusão/metabolismo , Traumatismo por Reperfusão/sangue , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia , Transplante de Células-Tronco , Células-Tronco/imunologia , Fatores de Tempo , Transfecção , Cicatrização
10.
Platelets ; 22(6): 415-21, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21413844

RESUMO

Shear forces are generated in all parts of the vascular system and contribute directly and indirectly to vascular disease progression. Endothelial cells are able to adapt to flow conditions, and are known to polarize and migrate in response to shear forces. Platelets exposed to shear stress are activated and release bioactive molecules from their alpha granules. So far, platelets have been considered to be static cells that do not leave the site of tight adhesion. However, we have recently been able to demonstrate the capacity of platelets to migrate in response to stromal derived factor-1 (SDF-1). In this project, we have demonstrated that platelets accumulate in areas with a high concentration of SDF-1 under flow conditions and respond to high shear stress by cellular polarization, cytoskeletal reorganisation, and flow-directed migration. In this context, we have shown increased Wiskott-Aldrich Syndrome protein (WASP) phosphorylation and intracellular redistribution of focal adhesion kinase (FAK) under high-shear stress conditions. The effect of flow-induced platelet migration has not previously been recognized and offers a new role for platelets as mobile cells. Their migratory potential may enable platelets to cover intimal lesions and contribute to vascular repair.


Assuntos
Plaquetas , Movimento Celular , Quimiocina CXCL12/farmacologia , Plaquetas/citologia , Plaquetas/metabolismo , Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Polaridade Celular , Quimiocina CXCL12/metabolismo , Endotélio Vascular/metabolismo , Citometria de Fluxo , Proteína-Tirosina Quinases de Adesão Focal/biossíntese , Hemodinâmica/efeitos dos fármacos , Humanos , Microscopia Confocal , Fosforilação/efeitos dos fármacos , Estresse Mecânico , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
11.
J Exp Med ; 200(5): 671-80, 2004 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-15337793

RESUMO

In addition to releasing preformed granular proteins, polymorphonuclear leukocytes (PMNs) synthesize chemokines and other factors under transcriptional control. Here we demonstrate that PMNs express an inducible transcriptional modulator by signal-dependent activation of specialized mechanisms that regulate messenger RNA (mRNA) translation. HL-60 myelocytic cells differentiated to surrogate PMNs respond to activation by platelet activating factor by initiating translation and with appearance of specific mRNA transcripts in polyribosomes. cDNA array analysis of the polyribosome fraction demonstrated that retinoic acid receptor (RAR)-alpha, a transcription factor that controls the expression of multiple genes, is one of the polyribosome-associated transcripts. Quiescent surrogate HL60 PMNs and primary human PMNs contain constitutive message for RAR-alpha but little or no protein. RAR-alpha protein is rapidly synthesized in response to platelet activating factor under the control of a specialized translational regulator, mammalian target of rapamycin, and is blocked by the therapeutic macrolide rapamycin, events consistent with features of the 5' untranslated region of the transcript. Newly synthesized RAR-alpha modulates production of interleukin-8. Rapid expression of a transcription factor under translational control is a previously unrecognized mechanism in human PMNs that indicates unexpected diversity in gene regulation in this critical innate immune effector cell.


Assuntos
Regulação da Expressão Gênica , Neutrófilos/metabolismo , Biossíntese de Proteínas , Receptores do Ácido Retinoico/biossíntese , Transcrição Gênica , Regiões 5' não Traduzidas , Diferenciação Celular , DNA Complementar/metabolismo , Ensaio de Imunoadsorção Enzimática , Células HL-60 , Humanos , Interleucina-8/metabolismo , Modelos Moleculares , Análise de Sequência com Séries de Oligonucleotídeos , Polirribossomos/metabolismo , Estrutura Secundária de Proteína , RNA Mensageiro/metabolismo , Receptor alfa de Ácido Retinoico , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo
12.
Semin Thromb Hemost ; 36(2): 203-11, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20414836

RESUMO

Platelets respond immediately to vascular injury by adhesion, aggregation, and thrombus formation. Disruption of the endothelial cell layer exposes extracellular matrix to the bloodstream. Collagen binding to platelet glycoprotein VI (GPVI) mediates the initial adhesion of the rolling platelet to the vascular wound. Signaling by GPVI leads to the onset of the platelet activation cascade that is finally crowned by a firm and shear-resistant integrin-based adhesive clot. Blockade of collagen binding to GPVI would prevent initial adhesion and further activation of the platelet and would have an enormous impact in antithrombotic therapy. Besides the therapeutical implication, radiolabeled GPVI gives us a valuable diagnostic means that may be used clinically for plaque imaging in the near future.


Assuntos
Glicoproteínas da Membrana de Plaquetas/análise , Glicoproteínas da Membrana de Plaquetas/metabolismo , Aterosclerose/diagnóstico , Aterosclerose/tratamento farmacológico , Colágeno/metabolismo , Fibrinolíticos/farmacologia , Humanos , Ativação Plaquetária/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Solubilidade
13.
Semin Thromb Hemost ; 36(2): 146-56, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20414829

RESUMO

Platelets are involved in the initiation of atherosclerosis by adherence to inflamed endothelium. Monocytes bind to these platelets and transmigrate into the vessel wall, transforming into macrophages and foam cells. We have previously shown that lipid-laden platelets are phagocytosed by macrophages. In this study we investigated the functional consequences of oxidized low-density lipoprotein (oxLDL) uptake on platelet function and interaction with the endothelium. Human platelets were isolated from healthy donors and activated by adenosine diphosphate. Immunofluorescence microscopy and flow cytometry revealed that oxLDL is located intracellularly in vesicles. With mepacrine costaining and confocal microtomography, we were able to identify dense granules as the vesicles that contain oxLDL. OxLDL-laden platelets induced intercellular adhesion molecule 1 expression in endothelial cells more than exogenous native LDL, oxLDL, and oxLDL-negative platelets. Furthermore, oxLDL-laden platelets induced foam cell development from CD34(+) progenitor cells. On endothelial regeneration, oxLDL-laden platelets had the opposite effect: The number of CD34(+) progenitor cells (colony-forming units) able to transform into endothelial cells was significantly reduced in the presence of oxLDL-platelets, whereas native LDL had no effect. Our results demonstrate that activated platelets internalize oxLDL and that oxLDL-laden platelets activate endothelium, inhibit endothelial regeneration, and promote foam cell development. Platelet oxLDL contributes significantly to vascular inflammation and is able to promote atherosclerosis.


Assuntos
Plaquetas/fisiologia , Endotélio Vascular/patologia , Inflamação/etiologia , Lipoproteínas LDL/fisiologia , Plaquetas/patologia , Endocitose , Células Endoteliais/patologia , Células Espumosas/citologia , Células-Tronco Hematopoéticas/citologia , Humanos , Ativação Plaquetária , Regeneração
14.
Circ Res ; 102(3): 302-9, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18048771

RESUMO

In atherosclerosis, circulating platelets interact with endothelial cells and monocytes, leading to cell activation and enhanced recruitment of leukocytes into the vascular wall. The invasion of monocytes is accompanied by overexpression of matrix metalloproteinases (MMPs), which are thought to promote atherosclerosis and trigger plaque rupture. Following interaction with itself, the extracellular matrix metalloproteinase inducer (EMMPRIN) induces MMP synthesis via a little-known intracellular pathway. Recently, we showed upregulation of EMMPRIN on monocytes during acute myocardial infarction. EMMPRIN also stimulates secretion of MMP-9 by monocytes and of MMP-2 by smooth muscle cells, indicating that it may be an important regulator of MMP activity. Expression of EMMPRIN on platelets has not been described until now. Here, we demonstrate that resting platelets show low surface expression of EMMPRIN, which is upregulated by various platelet stimulators (flow cytometry). EMMPRIN is located in the open canalicular system and in alpha granules of platelets (according to electron microscopy and sucrose gradient ultracentrifugation). Platelet stimulation with recombinant EMMPRIN-Fc induced surface expression of CD40L and P-selectin (according to flow cytometry), suggesting that EMMPRIN-EMMPRIN interaction activates platelets. Coincubation of platelets with monocytes induced EMMPRIN-mediated nuclear factor kappaB activation (according to Western blot) in monocytes with increased MMP-9 (zymography), interleukin-6, and tumor necrosis factor-alpha secretion (according to ELISA) by monocytes. In conclusion, EMMPRIN displays a new platelet receptor that is upregulated on activated platelets. Binding of EMMPRIN to platelets fosters platelet degranulation. Platelet-monocyte interactions via EMMPRIN stimulate nuclear factor kappaB-driven inflammatory pathways in monocytes, such as MMP and cytokine induction. Thus, EMMPRIN may represent a novel target to diminish the burden of protease activity and inflammation in atherosclerosis.


Assuntos
Basigina/metabolismo , Plaquetas/metabolismo , Regulação da Expressão Gênica , Monócitos/metabolismo , NF-kappa B/metabolismo , Ativação Plaquetária , Aterosclerose/metabolismo , Aterosclerose/patologia , Basigina/genética , Basigina/farmacologia , Plaquetas/ultraestrutura , Ligante de CD40/metabolismo , Técnicas de Cocultura , Células Endoteliais/metabolismo , Células Endoteliais/ultraestrutura , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Regiões Constantes de Imunoglobulina/genética , Regiões Constantes de Imunoglobulina/metabolismo , Regiões Constantes de Imunoglobulina/farmacologia , Inflamação/metabolismo , Inflamação/patologia , Interleucina-6/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Monócitos/ultraestrutura , Selectina-P/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Vesículas Secretórias/metabolismo , Vesículas Secretórias/ultraestrutura , Fator de Necrose Tumoral alfa/metabolismo
15.
Eur Heart J ; 30(5): 584-93, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19109356

RESUMO

AIMS: Previous experimental studies have suggested that platelet stromal-cell-derived factor-1 (SDF-1) regulates mobilization and recruitment of haematopoietic progenitor cells supporting revascularization in mice. However, there are no clinical data available regarding platelet-bound SDF-1 in patients with acute coronary syndrome (ACS). The objective of this study was to evaluate the platelet-surface expression of SDF-1 in patients with ACS. METHODS AND RESULTS: Patients with ACS (n = 418) showed a significantly enhanced SDF-1 expression on admission compared with those with stable angina pectoris (SAP, n = 486) [SAP (mean fluorescence intensity (MFI) +/- SD): 13.48 +/- 5.27; ACS: 18.45 +/- 12.85; P < 0.001) independent of cardiovascular risk factors and medication. Enhanced platelet-bound SDF-1 expression was found in patients with reduced left ventricular ejection fraction (LVEF <55%) in comparison to patients with normal LVEF (P = 0.005). Platelet-bound SDF-1 expression positively correlated with the degree of platelet activation [CD62P: r = 0.325; glycoprotein VI (GPVI): r = 0.277; PAC-1: r = 0.501; P < 0.001 for all] and showed a significant, but slight association with plasma levels of SDF-1 (r = 0.084; P = 0.045). In a subgroup of patients with coronary artery disease, platelet-bound SDF-1, but not other platelet activation markers, significantly correlated with the number of circulating CD34(+) progenitor cells (r = 0.252; P = 0.002) or CD34(+)/CD133(+) endothelial progenitor cells (r = 0.352; P = 0.008). CONCLUSION: Platelet-bound SDF-1 may play an important role in peripheral homing of circulating progenitor cells thus in tissue regeneration.


Assuntos
Síndrome Coronariana Aguda/sangue , Plaquetas/metabolismo , Quimiocina CXCL12/sangue , Células-Tronco Hematopoéticas/metabolismo , Síndrome Coronariana Aguda/fisiopatologia , Idoso , Idoso de 80 Anos ou mais , Antígenos CD34/sangue , Biomarcadores/sangue , Contagem de Células , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Ativação Plaquetária , Volume Sistólico , Disfunção Ventricular Esquerda/sangue , Disfunção Ventricular Esquerda/fisiopatologia
16.
Circulation ; 117(2): 206-15, 2008 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-18086932

RESUMO

BACKGROUND: Peripheral homing of progenitor cells in areas of diseased organs is critical for tissue regeneration. The chemokine stromal cell-derived factor-1 (SDF-1) regulates homing of CD34+ stem cells. We evaluated the role of platelet-derived SDF-1 in adhesion and differentiation of human CD34+ cells into endothelial progenitor cells. METHODS AND RESULTS: Adherent platelets express substantial amounts of SDF-1 and recruit CD34+ cells in vitro and in vivo. A monoclonal antibody to SDF-1 or to its counterreceptor, CXCR4, inhibits stem cell adhesion on adherent platelets under high arterial shear in vitro and after carotid ligation in mice, as determined by intravital fluorescence microscopy. Platelets that adhere to human arterial endothelial cells enhance the adhesion of CD34+ cells on endothelium under flow conditions, a process that is inhibited by anti-SDF-1. During intestinal ischemia/reperfusion in mice, anti-SDF-1 and anti-CXCR4, but not isotype control antibodies, abolish the recruitment of CD34+ cells in microcirculation. Moreover, platelet-derived SDF-1 binding to CXCR4 receptor promotes platelet-induced differentiation of CD34+ cells into endothelial progenitor cells, as verified by colony-forming assays in vitro. CONCLUSIONS: These findings imply that platelet-derived SDF-1 regulates adhesion of stem cells in vitro and in vivo and promotes differentiation of CD34+ cells to endothelial progenitor cells. Because tissue regeneration depends on recruitment of progenitor cells to peripheral vasculature and their subsequent differentiation, platelet-derived SDF-1 may contribute to vascular and myocardial regeneration.


Assuntos
Plaquetas/fisiologia , Adesão Celular , Diferenciação Celular , Quimiocina CXCL12/fisiologia , Células Endoteliais/citologia , Células-Tronco/citologia , Animais , Antígenos CD34 , Plaquetas/química , Plaquetas/citologia , Células Cultivadas , Humanos , Camundongos , Regeneração
17.
Basic Res Cardiol ; 104(3): 352-7, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19190951

RESUMO

BACKGROUND: The platelet collagen receptor glycoprotein VI (GPVI) mediates platelet adhesion to subendothelial matrix and thrombus formation in acute coronary syndrome (ACS). This study examined patients with both ACS and stable coronary artery disease (CAD), which presented with atrial fibrillation (AF) and sinus rhythm (SR). METHODS AND RESULTS: We evaluated 992 patients with acute or stable CAD, and determined platelet surface expression of GPVI using flow cytometry. Seventy-eight patients presented with nonvalvular persistent AF. After 1:1 propensity score matching 156 matched cases with 78 pairs were obtained. Patients with AF and ACS showed a significantly decreased GPVI expression compared to patients with ACS and SR, whereas patients with stable angina pectoris (SA) presented with low level activation and no significant difference between SR and AF [mean fluorescence intensity (MFI) for ACS (SR Vs. AF): 20 +/- 6.3 Vs. 17.7 +/- 4.4; P = 0.023; SA (SR Vs. AF): 18.8 +/- 9.4 Vs. 18.1 +/- 6.1; P = 0.649]. In contrast, soluble GPVI was increased in ACS and AF accordingly [plasma GPVI (ng/ml) for ACS (SR Vs. AF): 1.4 +/- 0.8 Vs. 1.9 +/- 1.1; P = 0.038; SA (SR Vs. AF): 0.9 +/- 0.4 Vs. 1.1 +/- 0.5; P = 0.127]. CONCLUSION: Platelet GPVI surface expression is decreased in patients with AF and ACS compared to patients with SR and ACS. Nonvalvular AF is related to indices of chronic platelet activation and might be responsible for a down-regulation of GPVI receptor density on platelets, while soluble GPVI was increased in ACS and AF accordingly.


Assuntos
Síndrome Coronariana Aguda/metabolismo , Fibrilação Atrial/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Idoso , Plaquetas/metabolismo , Doença da Artéria Coronariana/metabolismo , Feminino , Citometria de Fluxo , Humanos , Masculino
18.
Thromb Haemost ; 101(5): 911-5, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19404545

RESUMO

Platelets play a key role in the development of an acute coronary syndrome (ACS) and contribute to cardiovascular events. Platelet collagen receptor glycoprotein VI (GPVI) contributes significantly to platelet adhesion and thrombus formation in ACS. We consecutively investigated both the platelet count and the platelet surface expression of GPVI in 843 patients with a symptomatic coronary artery disease verified by coronary angiography. Four hundred fourteen patients presented with stable angina pectoris and 429 patients with ACS. Platelet surface expression of GPVI and CD62P was determined by flow cytometry and platelet count with a coulter counter, plasmatic soluble GPVI was measured by ELISA. Platelet GPVI expression in patients with ACS was compared to platelet count. Patients with ACS showed significantly elevated GPVI expression levels in the first and second quartiles of platelet count compared to patients with higher platelet count [mean fluorescence intensity (MFI) +/- standard deviation): 1(st) vs. 4(th): 20.44 +/- 6.1 vs. 18.62 +/- 3.7; p=0.012; 2(nd)vs.3(rd):21.2+/-8.5vs.18.76+/-3.7;P=0.03; 2(nd)vs.4(th): 21.2+/-8.5vs.18.62+/-3.7;P=0.004], which was paralleled in trend for the CD62P expression [MFI: 1(st) vs. 4(th): 11.2 +/- 6.8 vs. 12.3 +/- 9; p=0.057; 2(nd) vs. 3(rd): 16.3 +/- 16 vs.12.7 +/- 5.3; p=0.138; 2(nd) vs. 4(th): 16.3 +/- 16 vs.11 +/- 4.4; p=0.043]. In a subgroup of 48 patients with ACS, determination of soluble GPVI showed similar results [plasma GPVI (ng/ml): 1(st)vs.4(th): 1.6 +/- 0.6 vs. 1.2 +/- 0.4; p=0.046; 1(st) vs. 3(rd): 1.6 +/- 0.6 vs. 1.1 +/- 0.5; p=0.038; 2(nd) vs. 3(rd): 1.9 +/- 0.8 vs. 1.1 +/- 0.5; p=0.04; 2(nd) vs. 4(th): 1.9 +/- 0.8 vs. 1.2 +/- 0.4; p=0.056]. Thus, a lower platelet count comes along with a higher GPVI surface expression and plasma concentration in patients with ACS, which potentially reflects increased activation and enhanced recruitment of platelets to the site of vascular injury.


Assuntos
Síndrome Coronariana Aguda/sangue , Angina Pectoris/sangue , Antígenos CD36/sangue , Doença da Artéria Coronariana/sangue , Ativação Plaquetária , Contagem de Plaquetas , Síndrome Coronariana Aguda/diagnóstico por imagem , Síndrome Coronariana Aguda/etiologia , Síndrome Coronariana Aguda/mortalidade , Idoso , Angina Pectoris/diagnóstico por imagem , Angina Pectoris/etiologia , Angina Pectoris/mortalidade , Biomarcadores/sangue , Angiografia Coronária , Doença da Artéria Coronariana/complicações , Doença da Artéria Coronariana/diagnóstico por imagem , Doença da Artéria Coronariana/mortalidade , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/sangue , Infarto do Miocárdio/etiologia , Selectina-P/sangue , Valor Preditivo dos Testes , Prognóstico , Medição de Risco , Fatores de Risco , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/etiologia , Fatores de Tempo
19.
Semin Thromb Hemost ; 35(3): 295-306, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19452405

RESUMO

This article reviews gender-specific differences in platelet function and response to aspirin. In contrast with men, women under 65 years of age do not benefit from aspirin therapy in primary prevention of coronary artery disease. Furthermore, the overall mortality after myocardial infarction is higher among women, which cannot simply be explained by age or individual treatment. Platelets play a central role in thrombo-ischemic events leading to myocardial infarction and ischemic stroke. Gender differences in platelet reactivity and "aspirin resistance" have been reported. Several studies using female platelets have shown increased platelet reactivity at baseline and a less effective inhibition of platelet aggregation by aspirin. The mechanisms underlying these differences are still to be elucidated, but influences of female sex hormones may play an important role. As a consequence, inhibition of platelet aggregation in women treated with aspirin may be insufficient, and female patients might benefit from higher maintenance dosages or the use of alternative antiplatelet medications. A potential future role of pharmacogenomics and laboratory monitoring of antiplatelet medications in predicting individual responsiveness is likely. Overall, primary prevention of myocardial infarction in women should be correlated with the individual risk score of the female patient. More studies focused on women are needed to optimize gender-specific therapy and, therefore, to improve clinical outcomes in women with atherosclerosis.


Assuntos
Aspirina/administração & dosagem , Infarto do Miocárdio/prevenção & controle , Inibidores da Agregação Plaquetária/administração & dosagem , Agregação Plaquetária/fisiologia , Trombose/prevenção & controle , Fatores Etários , Animais , Ensaios Clínicos como Assunto , Feminino , Hormônios Esteroides Gonadais/sangue , Humanos , Masculino , Infarto do Miocárdio/sangue , Agregação Plaquetária/efeitos dos fármacos , Fatores de Risco , Fatores Sexuais
20.
Cardiovasc Res ; 78(1): 8-17, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18218686

RESUMO

In the last decade, it was recognized that platelets and lipoproteins play a pivotal role in both early and late atherogenesis. Beside cellular interactions of platelets with other blood cells and vascular cells, interactions with lipoproteins seem to be quite important. Lipoproteins are fundamental 'players' in atherogenesis since they change the properties of different cells involved in atherosclerosis and thrombosis. Several studies have already shown that low density lipoproteins (LDL) are involved in the initiation of platelet signalling pathways. Platelets of hypercholesterolemic patients show hyperaggregability in vitro and enhanced activity in vivo. This review elucidates the major aspects concerning how native and modified lipoproteins influence the activation and metabolic behaviour of platelets, and shows a new way by which platelet-mediated lipoprotein transfer might contribute to foam cell formation. In hyperlipidaemia, circulating platelets are activated. This is accompanied by increased platelet aggregation, platelet-leukocyte aggregate formation, and platelet-induced superoxide anion production. Furthermore, oxidized LDL induces monocyte adhesion to the endothelium, migration and proliferation of smooth muscle cells, injures cells, interferes with nitric oxide release, and promotes procoagulant properties of vascular cells. New data about platelet-mediated lipoprotein transport and consequent foam cell formation, however, provide proof of how platelets might contribute to atheromatous lesion formation.


Assuntos
Aterosclerose/metabolismo , Plaquetas/metabolismo , Células Espumosas/metabolismo , Lipoproteínas/metabolismo , Ativação Plaquetária , Animais , Aterosclerose/sangue , Aterosclerose/patologia , Membrana Celular/metabolismo , Endocitose , Humanos , Lipoproteínas/sangue , Lipoproteínas LDL/metabolismo , Monócitos/metabolismo , Fagocitose , Receptores Depuradores/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa