Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Blood ; 139(1): 104-117, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34329392

RESUMO

Tyrosine phosphorylation of extracellular proteins is observed in cell cultures and in vivo, but little is known about the functional roles of tyrosine phosphorylation of extracellular proteins. Vertebrate lonesome kinase (VLK) is a broadly expressed secretory pathway tyrosine kinase present in platelet α-granules. It is released from platelets upon activation and phosphorylates substrates extracellularly. Its role in platelet function, however, has not been previously studied. In human platelets, we identified phosphorylated tyrosines mapped to luminal or extracellular domains of transmembrane and secreted proteins implicated in the regulation of platelet activation. To determine the role of VLK in extracellular tyrosine phosphorylation and platelet function, we generated mice with a megakaryocyte/platelet-specific deficiency of VLK. Platelets from these mice are normal in abundance and morphology but have significant changes in function both in vitro and in vivo. Resting and thrombin-stimulated VLK-deficient platelets exhibit a significant decrease in several tyrosine phosphobands. Results of functional testing of VLK-deficient platelets show decreased protease-activated receptor 4-mediated and collagen-mediated platelet aggregation but normal responses to adenosine 5'-diphosphate. Dense granule and α-granule release are reduced in these platelets. Furthermore, VLK-deficient platelets exhibit decreased protease-activated receptor 4-mediated Akt (S473) and Erk1/2 (T202/Y204) phosphorylation, indicating altered proximal signaling. In vivo, mice lacking VLK in megakaryocytes/platelets display strongly reduced platelet accumulation and fibrin formation after laser-induced injury of cremaster arterioles compared with control mice but with normal bleeding times. These studies show that the secretory pathway tyrosine kinase VLK is critical for stimulus-dependent platelet activation and thrombus formation, providing the first evidence that a secreted protein kinase is required for normal platelet function.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária , Proteínas Tirosina Quinases/metabolismo , Trombose/metabolismo , Animais , Plaquetas/patologia , Deleção de Genes , Células HEK293 , Humanos , Camundongos Transgênicos , Proteínas Tirosina Quinases/genética , Trombose/patologia
2.
Blood ; 125(12): 1976-85, 2015 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-25587041

RESUMO

Protease-activated receptor-1 (PAR1) couples the coagulation cascade to platelet activation during myocardial infarction and to endothelial inflammation during sepsis. This receptor demonstrates marked signaling bias. Its activation by thrombin stimulates prothrombotic and proinflammatory signaling, whereas its activation by activated protein C (APC) stimulates cytoprotective and antiinflammatory signaling. A challenge in developing PAR1-targeted therapies is to inhibit detrimental signaling while sparing beneficial pathways. We now characterize a novel class of structurally unrelated small-molecule PAR1 antagonists, termed parmodulins, and compare the activity of these compounds to previously characterized compounds that act at the PAR1 ligand-binding site. We find that parmodulins target the cytoplasmic face of PAR1 without modifying the ligand-binding site, blocking signaling through Gαq but not Gα13 in vitro and thrombus formation in vivo. In endothelium, parmodulins inhibit prothrombotic and proinflammatory signaling without blocking APC-mediated pathways or inducing endothelial injury. In contrast, orthosteric PAR1 antagonists such as vorapaxar inhibit all signaling downstream of PAR1. Furthermore, exposure of endothelial cells to nanomolar concentrations of vorapaxar induces endothelial cell barrier dysfunction and apoptosis. These studies demonstrate how functionally selective antagonism can be achieved by targeting the cytoplasmic face of a G-protein-coupled receptor to selectively block pathologic signaling while preserving cytoprotective pathways.


Assuntos
Endotélio Vascular/lesões , Lactonas/efeitos adversos , Piridinas/efeitos adversos , Receptor PAR-1/antagonistas & inibidores , Trombose/tratamento farmacológico , Trombose/prevenção & controle , Animais , Apoptose , Sítios de Ligação , Células COS , Chlorocebus aethiops , Citoplasma/metabolismo , Endotélio Vascular/efeitos dos fármacos , Exocitose , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação , Ligantes , Inibidores da Agregação Plaquetária/química , Proteína C/química , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
3.
Arterioscler Thromb Vasc Biol ; 33(3): 481-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23288151

RESUMO

OBJECTIVE: Platelet granule exocytosis serves a central role in hemostasis and thrombosis. Recently, single-cell amperometry has shown that platelet membrane fusion during granule exocytosis results in the formation of a fusion pore that subsequently expands to enable the extrusion of granule contents. However, the molecular mechanisms that control platelet fusion pore expansion and collapse are not known. METHODS AND RESULTS: We identified dynamin-related protein-1 (Drp1) in platelets and found that an inhibitor of Drp1, mdivi-1, blocked exocytosis of both platelet dense and α-granules. We used single-cell amperometry to monitor serotonin release from individual dense granules and, thereby, measured the effect of Drp1 inhibition on fusion pore dynamics. Inhibition of Drp1 increased spike width and decreased prespike foot events, indicating that Drp1 influences fusion pore formation and expansion. Platelet-mediated thrombus formation in vivo after laser-induced injury of mouse cremaster arterioles was impaired after infusion of mdivi-1. CONCLUSIONS: These results demonstrate that inhibition of Drp1 disrupts platelet fusion pore dynamics and indicate that Drp1 can be targeted to control thrombus formation in vivo.


Assuntos
Plaquetas/metabolismo , Dinaminas/sangue , Exocitose , GTP Fosfo-Hidrolases/sangue , Fusão de Membrana , Proteínas Associadas aos Microtúbulos/sangue , Proteínas Mitocondriais/sangue , Vesículas Secretórias/metabolismo , Trombose/sangue , Lesões do Sistema Vascular/sangue , Animais , Arteríolas/lesões , Plaquetas/efeitos dos fármacos , Modelos Animais de Doenças , Dinaminas/antagonistas & inibidores , Exocitose/efeitos dos fármacos , Fibrinolíticos/farmacologia , GTP Fosfo-Hidrolases/antagonistas & inibidores , Humanos , Lasers , Fusão de Membrana/efeitos dos fármacos , Camundongos , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Mitocondriais/antagonistas & inibidores , Selectina-P/sangue , Quinazolinonas/farmacologia , Coelhos , Vesículas Secretórias/efeitos dos fármacos , Serotonina/sangue , Trombose/etiologia , Trombose/prevenção & controle , Fatores de Tempo , Lesões do Sistema Vascular/etiologia
4.
J Clin Invest ; 122(6): 2104-13, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22565308

RESUMO

Thrombosis, or blood clot formation, and its sequelae remain a leading cause of morbidity and mortality, and recurrent thrombosis is common despite current optimal therapy. Protein disulfide isomerase (PDI) is an oxidoreductase that has recently been shown to participate in thrombus formation. While currently available antithrombotic agents inhibit either platelet aggregation or fibrin generation, inhibition of secreted PDI blocks the earliest stages of thrombus formation, suppressing both pathways. Here, we explored extracellular PDI as an alternative target of antithrombotic therapy. A high-throughput screen identified quercetin-3-rutinoside as an inhibitor of PDI reductase activity in vitro. Inhibition of PDI was selective, as quercetin-3-rutinoside failed to inhibit the reductase activity of several other thiol isomerases found in the vasculature. Cellular assays showed that quercetin-3-rutinoside inhibited aggregation of human and mouse platelets and endothelial cell-mediated fibrin generation in human endothelial cells. Using intravital microscopy in mice, we demonstrated that quercetin-3-rutinoside blocks thrombus formation in vivo by inhibiting PDI. Infusion of recombinant PDI reversed the antithrombotic effect of quercetin-3-rutinoside. Thus, PDI is a viable target for small molecule inhibition of thrombus formation, and its inhibition may prove to be a useful adjunct in refractory thrombotic diseases that are not controlled with conventional antithrombotic agents.


Assuntos
Plaquetas/metabolismo , Fibrinolíticos/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Isomerases de Dissulfetos de Proteínas/antagonistas & inibidores , Rutina/farmacologia , Trombose/tratamento farmacológico , Animais , Inibidores Enzimáticos/farmacologia , Fibrina/genética , Fibrina/metabolismo , Humanos , Camundongos , Isomerases de Dissulfetos de Proteínas/efeitos adversos , Isomerases de Dissulfetos de Proteínas/genética , Isomerases de Dissulfetos de Proteínas/farmacologia , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Trombose/induzido quimicamente , Trombose/enzimologia
5.
ACS Med Chem Lett ; 3(3): 232-237, 2012 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-22408714

RESUMO

A high-throughput screen of the NIH-MLSMR compound collection, along with a series of secondary assays to identify potential targets of hit compounds, previously identified a 1,3-diaminobenzene scaffold that targets protease-activated receptor 1 (PAR1). We now report additional structure-activity relationship (SAR) studies that delineate the requirements for activity at PAR1 and identify plasma-stable analogues with nanomolar inhibition of PAR1-mediated platelet activation. Compound 4 was declared as a probe (ML161) with the NIH Molecular Libraries Program. This compound inhibited platelet aggregation induced by a PAR1 peptide agonist or by thrombin but not by several other platelet agonists. Initial studies suggest that ML161 is an allosteric inhibitor of PAR1. These findings may be important for the discovery of antithrombotics with an improved safety profile.

6.
Proc Natl Acad Sci U S A ; 108(7): 2951-6, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21282664

RESUMO

G protein-coupled receptors (GPCRs) can assume multiple conformations and possess multiple binding sites. Whereas endogenous agonists acting at the orthosteric binding site stabilize the active receptor conformation, small molecules that act at nonorthosteric sites can stabilize alternative conformations. The large majority of these allosteric modulators associate with extracellular loops of GPCRs. The role of intracellular domains in mediating allosteric modulation is largely unknown. In screening a small-molecule library for inhibitors of platelet activation, we identified a family of compounds that modified PAR1-mediated granule secretion. The most potent inhibitory compound, termed JF5, also demonstrated noncompetitive inhibition of the α(2A)-adrenergic receptor. Aggregation studies using a battery of platelet GPCR agonists demonstrated that sensitivity to JF5 was limited to GPCRs that possessed a constrained eighth helix, as defined by a C-terminal palmitoylation site and interactions with TM7 and the i1 loop. Inhibition by JF5 was overcome in a PAR1 mutant in which the eighth helix was deleted, confirming a role for helix 8 in JF5 activity. Evaluation of downstream signaling showed that JF5 was selective with regard to G protein coupling, blocking signaling mediated by G(αq) but not G(α12). The compound inhibited thrombus formation in vivo following vascular injury with an IC(50) of ∼1 mg/kg. These results indicate a role for helix 8 in conferring sensitivity to small molecules, and show that this sensitivity can be exploited to control platelet activation during thrombus formation.


Assuntos
Antitrombinas/metabolismo , Receptor PAR-1/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Trombose/metabolismo , Regulação Alostérica/fisiologia , Animais , Cálcio/metabolismo , Linhagem Celular , Cães , Epinefrina , Citometria de Fluxo , Luciferases , Selectina-P/metabolismo , Fragmentos de Peptídeos/metabolismo , Agregação Plaquetária , Estrutura Secundária de Proteína/fisiologia , Receptor PAR-1/agonistas
7.
Biochemistry ; 49(21): 4533-42, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20429610

RESUMO

Following platelet activation, platelets undergo a dramatic shape change mediated by the actin cytoskeleton and accompanied by secretion of granule contents. While the actin cytoskeleton is thought to influence platelet granule secretion, the mechanism for this putative regulation is not known. We found that disruption of the actin cytoskeleton by latrunculin A inhibited alpha-granule secretion induced by several different platelet agonists without significantly affecting activation-induced platelet aggregation. In a cell-free secretory system, platelet cytosol was required for alpha-granule secretion. Inhibition of actin polymerization prevented alpha-granule secretion in this system, and purified platelet actin could substitute for platelet cytosol to support alpha-granule secretion. To determine whether SNAREs physically associate with the actin cytoskeleton, we isolated the Triton X-100 insoluble actin cytoskeleton from platelets. VAMP-8 and syntaxin-2 associated only with actin cytoskeletons of activated platelets. Syntaxin-4 and SNAP-23 associated with cytoskeletons isolated from either resting or activated platelets. When syntaxin-4 and SNAP-23 were tested for actin binding in a purified protein system, only syntaxin-4 associated directly with polymerized platelet actin. These data show that the platelet cytoskeleton interacts with select SNAREs and that actin polymerization facilitates alpha-granule release.


Assuntos
Plaquetas/metabolismo , Grânulos Citoplasmáticos/metabolismo , Citoesqueleto/metabolismo , Ativação Plaquetária/fisiologia , Proteínas SNARE/metabolismo , Actinas/imunologia , Actinas/metabolismo , Plaquetas/fisiologia , Compostos Bicíclicos Heterocíclicos com Pontes , Grânulos Citoplasmáticos/imunologia , Citoesqueleto/imunologia , Citoesqueleto/fisiologia , Humanos , Octoxinol/metabolismo , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/imunologia , Proteínas Qa-SNARE/imunologia , Proteínas Qa-SNARE/metabolismo , Sintaxina 1/metabolismo , Tiazolidinas , beta-Tromboglobulina
8.
Blood ; 114(5): 1083-90, 2009 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-19395672

RESUMO

Individuals whose platelets lack dense or alpha-granules suffer various degrees of abnormal bleeding, implying that granule cargo contributes to hemostasis. Despite these clinical observations, little is known regarding the effects of impaired platelet granule secretion on thrombus formation in vivo. In platelets, SNARE proteins mediate the membrane fusion events required for granule cargo release. Endobrevin/VAMP-8 is the primary vesicle-SNARE (v-SNARE) responsible for efficient release of dense and alpha-granule contents; thus, VAMP-8(-/-) mice are a useful model to evaluate the importance of platelet granule secretion in thrombus formation. Thrombus formation, after laser-induced vascular injury, in these mice is delayed and decreased, but not absent. In contrast, thrombus formation is almost completely abolished in the mouse model of Hermansky-Pudlak syndrome, ruby-eye, which lacks dense granules. Evaluation of aggregation of VAMP-8(-/-) and ruby-eye platelets indicates that defective ADP release is the primary abnormality leading to impaired aggregation. These results demonstrate the importance of dense granule release even in the earliest phases of thrombus formation and validate the distal platelet secretory machinery as a potential target for antiplatelet therapies.


Assuntos
Transtornos da Coagulação Sanguínea/sangue , Coagulação Sanguínea/fisiologia , Plaquetas/metabolismo , Grânulos Citoplasmáticos/metabolismo , Agregação Plaquetária/fisiologia , Proteínas R-SNARE/fisiologia , Difosfato de Adenosina/metabolismo , Animais , Arteríolas/lesões , Transtornos da Coagulação Sanguínea/etiologia , Plaquetas/química , Plaquetas/ultraestrutura , Modelos Animais de Doenças , Síndrome de Hermanski-Pudlak/sangue , Humanos , Lasers/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Proteínas R-SNARE/deficiência , Proteínas R-SNARE/genética , Proteínas SNARE/sangue
9.
Blood ; 113(5): 1112-21, 2009 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-18802008

RESUMO

Platelet microparticles are a normal constituent of circulating blood. Several studies have demonstrated positive correlations between thrombotic states and platelet microparticle levels. Yet little is known about the processes by which platelet microparticles are generated in vivo. We now characterize microparticles derived directly from megakaryocytes. Video microscopy of live mouse megakaryocytes demonstrated that microparticles form as submicron beads along the lengths of slender, unbranched micropodia. These microparticles are CD41(+), CD42b(+), and express surface phosphatidylserine. Megakaryocyte microparticle generation is resistant to inhibition of microtubule assembly, which is critical to platelet formation, and augmented by inhibition of actin polymerization. To determine whether circulating microparticles are derived primarily from activated platelets or megakaryocytes, we identified markers that distinguish between these 2 populations. CD62P and LAMP-1 were found only on mouse microparticles from activated platelets. In contrast, full-length filamin A was found in megakaryocyte-derived microparticles, but not microparticles from activated platelets. Circulating microparticles isolated from mice were CD62P(-), LAMP-1(-) and expressed full-length filamin A, indicating a megakaryocytic origin. Similarly, circulating microparticles isolated from healthy volunteers were CD62P(-) and expressed full-length filamin A. Cultured human megakaryocytes elaborated microparticles that were CD41(+), CD42b(+), and express surface phosphatidylserine. These results indicate that direct production by megakaryocytes represents a physiologic means to generate circulating platelet microparticles.


Assuntos
Plaquetas/metabolismo , Micropartículas Derivadas de Células/metabolismo , Megacariócitos/metabolismo , Actinas/metabolismo , Animais , Plaquetas/ultraestrutura , Micropartículas Derivadas de Células/ultraestrutura , Células Cultivadas , Proteínas Contráteis , Filaminas , Humanos , Proteínas de Membrana Lisossomal/metabolismo , Megacariócitos/ultraestrutura , Camundongos , Proteínas dos Microfilamentos , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Selectina-P/metabolismo , Fosfatidilserinas/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo
10.
Arterioscler Thromb Vasc Biol ; 27(6): 1478-85, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17303775

RESUMO

OBJECTIVE: Several platelet proteins are palmitoylated, but whether protein palmitoylation functions in platelet activation is unknown. We sought to determine the role of platelet protein palmitoylation in platelet activation and thrombus formation. METHODS AND RESULTS: Platelet proteins were depalmitoylated by infusing acyl-protein thioesterase 1 into permeabilized platelets. In intact platelets, platelet protein palmitoylation was blocked using the protein palmitoylation inhibitor cerulein. The effects of inhibiting platelet protein palmitoylation on platelet function and on thrombus formation in vivo were evaluated. When infused into permeabilized platelets, acyl-protein thioesterase 1 reduced total platelet protein palmitoylation and inhibited protease-activated receptor-1-mediated alpha-granule secretion with an IC50 of 175 nmol/L and maximal inhibition of > or = 90%. G(alpha q) and SNAP-23, membrane-associated proteins that are constitutively palmitoylated, translocated to the cytosol when permeabilized platelets were exposed to recombinant acyl-protein thioesterase 1. The protein palmitoylation inhibitor cerulein also inhibited platelet granule secretion and aggregation. Studies using intravital microscopy showed that incubation with cerulein decreased the rate of platelet accumulation into thrombi formed after laser-induced injury of mouse arterioles and inhibited maximal platelet accumulation by >60%. CONCLUSION: These studies show that platelets possess a protein palmitoylation machinery that is required for both platelet activation and platelet accumulation into thrombi. These studies show that inhibition of platelet protein palmitoylation blocks platelet aggregation and granule secretion. In a murine model of thrombus formation, inhibition of protein palmitoylation markedly inhibits platelet accumulation into thrombi at sites of vascular injury.


Assuntos
Plaquetas/efeitos dos fármacos , Ácido Palmítico/metabolismo , Fragmentos de Peptídeos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Tioléster Hidrolases/metabolismo , Trombose/metabolismo , Aciltransferases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Arteríolas/cirurgia , Plaquetas/metabolismo , Ceruletídeo/farmacologia , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Humanos , Técnicas In Vitro , Lasers , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Selectina-P/metabolismo , Transporte Proteico/efeitos dos fármacos , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Receptor PAR-1/agonistas , Receptor PAR-1/metabolismo , Proteínas Recombinantes/farmacologia , Tioléster Hidrolases/genética , Tioléster Hidrolases/farmacologia , Trombose/sangue , Trombose/patologia
11.
Blood ; 105(10): 3879-87, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15671445

RESUMO

Stimulation of platelets with strong agonists results in centralization of cytoplasmic organelles and secretion of granules. These observations have led to the supposition that cytoskeletal contraction facilitates granule release by promoting the interaction of granules with one another and with membranes of the open canalicular system. Yet, the influence of the actin cytoskeleton in controlling the membrane fusion events that mediate granule secretion remains largely unknown. To evaluate the role of the actin cytoskeleton in platelet granule secretion, we have assessed the effects of latrunculin A and cytochalasin E on granule secretion. Exposure of platelets to low concentrations of these reagents resulted in acceleration and augmentation of agonist-induced alpha-granule secretion with comparatively modest effects on dense granule secretion. In contrast, exposure of platelets to high concentrations of latrunculin A inhibited agonist-induced alpha-granule secretion but stimulated dense granule secretion. Incubation of permeabilized platelets with low concentrations of latrunculin A primed platelets for Ca(2+)- or guanosine triphosphate (GTP)-gamma-S-induced alpha-granule secretion. Latrunculin A-dependent alpha-granule secretion was inhibited by antibodies directed at vesicle-associated membrane protein (VAMP), demonstrating that latrunculin A supports soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) protein-dependent membrane fusion. These results indicate that the actin cytoskeleton interferes with platelet exocytosis and differentially regulates alpha-granule and dense granule secretion.


Assuntos
Actinas/metabolismo , Plaquetas/citologia , Plaquetas/metabolismo , Degranulação Celular , Grânulos Citoplasmáticos/metabolismo , Citoesqueleto/fisiologia , Actinas/ultraestrutura , Plaquetas/efeitos dos fármacos , Plaquetas/ultraestrutura , Compostos Bicíclicos Heterocíclicos com Pontes/agonistas , Degranulação Celular/efeitos dos fármacos , Citocalasinas/farmacologia , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/ultraestrutura , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/ultraestrutura , Humanos , Cinética , Microscopia Eletrônica de Transmissão , Fragmentos de Peptídeos/farmacologia , Proteínas SNARE , Acetato de Tetradecanoilforbol/farmacologia , Tiazóis/agonistas , Tiazolidinas , Proteínas de Transporte Vesicular/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA