Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Clin Endocrinol (Oxf) ; 79(2): 252-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23278130

RESUMO

INTRODUCTION: Previous studies investigating cardiovascular (CV) risk in obese women with polycystic ovary syndrome (PCOS) have been potentially confounded by not adequately accounting for body weight. OBJECTIVE: To assess if PCOS increases CV risk independently in young obese women by examining carotid intima-media wall thickness (cIMT) and platelet function. DESIGN: A case-control study comparing women with PCOS (n = 21) to age (32·8 ± 7·2 vs 33·5 ± 6·7 years), and weight (100·9 ± 16·7 vs 99·3 ± 14·7 kg)-matched controls (n = 19). Platelet function was examined by flow cytometry, clot structure and fibrinolysis by turbidimetric assays and endothelial function by ELISA and post ischaemic reactive hyperaemia. RESULTS: The PCOS group had higher testosterone 1·2 ± 0·3 vs 0·9 ± 0·3 nmol/l (P = 0·01), HOMA-IR 2·5 ± 1·7 vs 1·7 ± 1·0 (P = 0·08), impaired glucose regulation 33·3% vs 5·3% (P = 0·02), and urinary isoprostane 16·0 ± 4·4 vs 11·8 ± 7·1 ng/ml (P = 0·04) compared to controls. Mean cIMT 0·5 ± 0·05 vs 0·48 ± 0·06 mm (P = 0·36), and basal platelet surface expression (percentage of positive cells) of P-selectin 0·52 ± 0·3 vs 0·43 ± 0·23 (P = 0·40) and fibrinogen binding 0·97 ± 0·4 vs 0·83 ± 0·3 (P = 0·48) did not significantly differ between the PCOS and control groups respectively. Furthermore, platelets sensitivity to stimulation with adenosine-5'-diphosphate or inhibition with prostacyclin, clot structure and fibrinolytic efficiency ex vivo, endothelial reactive hyperaemic index (RHI), inflammation (hsCRP) and adhesion markers (sE-selectin, sP-selectin, sVCAM-1 and sICAM-1) were not significantly different between the two groups. CONCLUSIONS: PCOS appeared not to independently increase atherothrombotic risk when matched for obesity. It is likely that any excess CV risk in young obese women with PCOS can either be attributed to obesity or is not yet apparent at this early stage of the condition.


Assuntos
Plaquetas/fisiologia , Espessura Intima-Media Carotídea , Obesidade/fisiopatologia , Síndrome do Ovário Policístico/fisiopatologia , Adulto , Doenças Cardiovasculares/etiologia , Endotélio Vascular/fisiopatologia , Feminino , Humanos , Resistência à Insulina , Isoprostanos/urina , Obesidade/sangue , Ativação Plaquetária , Síndrome do Ovário Policístico/sangue , Fatores de Risco
2.
Sci Rep ; 8(1): 3032, 2018 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-29445102

RESUMO

Evidence has emerged to suggest that thrombi are dynamic structures with distinct areas of differing platelet activation and inhibition. We hypothesised that Nitric oxide (NO), a platelet inhibitor, can modulate the actin cytoskeleton reversing platelet spreading, and therefore reduce the capability of thrombi to withstand a high shear environment. Our data demonstrates that GSNO, DEANONOate, and a PKG-activating cGMP analogue reversed stress fibre formation and increased actin nodule formation in adherent platelets. This effect is sGC dependent and independent of ADP and thromboxanes. Stress fibre formation is a RhoA dependent process and NO induced RhoA inhibition, however, it did not phosphorylate RhoA at ser188 in spread platelets. Interestingly NO and PGI2 synergise to reverse stress fibre formation at physiologically relevant concentrations. Analysis of high shear conditions indicated that platelets activated on fibrinogen, induced stress fibre formation, which was reversed by GSNO treatment. Furthermore, preformed thrombi on collagen post perfused with GSNO had a 30% reduction in thrombus height in comparison to the control. This study demonstrates that NO can reverse key platelet functions after their initial activation and identifies a novel mechanism for controlling excessive thrombosis.


Assuntos
Óxido Nítrico/metabolismo , Fibras de Estresse/metabolismo , Trombose/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Plaquetas/efeitos dos fármacos , Citoesqueleto/metabolismo , Voluntários Saudáveis , Humanos , Óxido Nítrico/fisiologia , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Resistência ao Cisalhamento/fisiologia , Fibras de Estresse/efeitos dos fármacos , Trombose/patologia , Tromboxanos/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
3.
Diabetes Metab ; 44(5): 431-436, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29784564

RESUMO

AIM: Strict glycaemic control has been associated with an increased mortality rate in subjects with type 2 diabetes (T2DM). Here we examined platelet function immediately and 24hours following induced hypoglycaemia in people with type 2 diabetes compared to healthy age-matched controls. METHODS: Hyperinsulinaemic clamps reduced blood glucose to 2.8mmol/L (50mg/dl) for 1hour. Sampling at baseline; euglycaemia 5mmol/L (90mg/dl); hypoglycaemia; and at 24 post clamp were undertaken. Platelet function was measured by whole blood flow cytometry. RESULTS: 10 subjects with T2DM and 8 controls were recruited. Platelets from people with T2DM showed reduced sensitivity to prostacyclin (PGI2, 1nM) following hypoglycaemia. The ability of PGI2 to inhibit platelet activation was significantly impaired at 24hours compared to baseline in the T2DM group. Here, inhibition of fibrinogen binding was 29.5% (10.3-43.8) compared to 50.8% (36.8-61.1), (P<0.05), while inhibition of P-selectin expression was 32% (16.1-47.6) vs. 54.4% (42.5-67.5) (P<0.05). No significant changes in platelet function were noted in controls. CONCLUSION: Induced hypoglycaemia in T2DM enhances platelet hyperactivity through impaired sensitivity to prostacyclin at 24hours.


Assuntos
Plaquetas , Diabetes Mellitus Tipo 2/sangue , Hipoglicemia/sangue , Ativação Plaquetária/fisiologia , Adulto , Glicemia , Estudos de Casos e Controles , Diabetes Mellitus Tipo 2/fisiopatologia , Feminino , Humanos , Hipoglicemia/fisiopatologia , Masculino , Pessoa de Meia-Idade , Testes de Função Plaquetária
4.
J Thromb Haemost ; 15(8): 1668-1678, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28509344

RESUMO

Essentials Platelet shape change requires cytoskeletal rearrangement via myosin-mediated actin contraction. We investigated whether nitric oxide (NO) affected thrombin-induced platelet shape change. NO inhibits shape change, RhoA/ROCK signalling and myosin light chain (MLC) phosphorylation. NO promotes MLC phosphatase activity, thus prevents MLC phosphorylation and shape change. SUMMARY: Background Platelet shape change, spreading and thrombus stability require activation of the actin cytoskeleton contractile machinery. The mechanisms controlling actin assembly to prevent unwanted platelet activation are unclear. Objectives We examined the effects of nitric oxide on the signaling pathways regulating platelet actin-myosin activation. Results S-nitrosoglutathione (GSNO) inhibited thrombin-induced platelet shape change and myosin phosphorylation of the myosin light chain (MLC). Because thrombin stimulates phospho-MLC through the RhoA/ ROCK dependent inhibition of MLC phosphatase (MLCP) we examined the effects of NO on this pathway. Thrombin caused the GTP loading and activation of RhoA, leading to the ROCK-mediated phosphorylation of MLCP on threonine 853 (thr853 ), which is known to inhibit phosphatase activity. Treatment of platelets with GSNO blocked ROCK-mediated increases in phosphoMLCP-thr853 induced by thrombin. This effect was mimicked by the direct activator of protein kinase G, 8-pCPT-PET-cGMP, and blocked by the inhibition of guanylyl cyclase, but not inhibitors of protein kinase A. Further exploration of the mechanism demonstrated that GSNO stimulated the association of RhoA with protein kinase G (PKG) and the inhibitory phosphorylation (serine188) of RhoA in a cGMP-dependent manner. Consistent with these observations, in vitro experiments revealed that recombinant PKG caused direct phosphorylation of RhoA. The inhibition of RhoA by GSNO prevented ROCK-mediated phosphorylation and inhibition of MLCP activity. Conclusions These data suggest novel crosstalk between the NO-cGMP-PKG and RhoA/ROCK signaling pathways to control platelet actin remodeling.


Assuntos
Plaquetas/enzimologia , Forma Celular , GMP Cíclico/metabolismo , Citoesqueleto/enzimologia , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Ativação Plaquetária , Sistemas do Segundo Mensageiro , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Plaquetas/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Citoesqueleto/efeitos dos fármacos , Humanos , Cadeias Leves de Miosina/metabolismo , Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/metabolismo , Doadores de Óxido Nítrico/farmacologia , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , S-Nitrosoglutationa/metabolismo , S-Nitrosoglutationa/farmacologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Trombina/farmacologia , Fatores de Tempo
5.
Sci Rep ; 7(1): 5582, 2017 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-28717253

RESUMO

Prostacyclin (PGI2) modulates platelet activation to regulate haemostasis. Evidence has emerged to suggest that thrombi are dynamic structures with distinct areas of differing platelet activation. It was hypothesised that PGI2 could reverse platelet spreading by actin cytoskeletal modulation, leading to reduced capability of platelet aggregates to withstand a high shear environment. Our data demonstrates that post-flow of PGI2 over activated and spread platelets on fibrinogen, identified a significant reduction in platelet surface area under high shear. Exploration of the molecular mechanisms underpinning this effect revealed that PGI2 reversed stress fibre formation in adherent platelets, reduced platelet spreading, whilst simultaneously promoting actin nodule formation. The effects of PGI2 on stress fibres were mimicked by the adenylyl cyclase activator forskolin and prevented by inhibitors of protein kinase A (PKA). Stress fibre formation is a RhoA dependent process and we found that treatment of adherent platelets with PGI2 caused inhibitory phosphorylation of RhoA, reduced RhoA GTP-loading and reversal of myosin light chain phosphorylation. Phospho-RhoA was localised in actin nodules with PKA type II and a number of other phosphorylated PKA substrates. This study demonstrates that PGI2 can reverse key platelet functions after their initial activation and identifies a novel mechanism for controlling thrombosis.


Assuntos
Plaquetas/química , Epoprostenol/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Fibras de Estresse/efeitos dos fármacos , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Agregação Plaquetária/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/metabolismo
6.
J Thromb Haemost ; 13(9): 1721-34, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26176741

RESUMO

BACKGROUND: Platelet adhesion to von Willebrand factor (VWF) is modulated by 3',5'-cyclic adenosine monophosphate (cAMP) signaling through protein kinase A (PKA)-mediated phosphorylation of glycoprotein (GP)Ibß. A-kinase anchoring proteins (AKAPs) are proposed to control the localization and substrate specificity of individual PKA isoforms. However, the role of PKA isoforms in regulating the phosphorylation of GPIbß and platelet response to VWF is unknown. OBJECTIVES: We wished to determine the role of PKA isoforms in the phosphorylation of GPIbß and platelet activation by VWF as a model for exploring the selective partitioning of cAMP signaling in platelets. RESULTS: The two isoforms of PKA in platelets, type I (PKA-I) and type II (PKA-II), were differentially localized, with a small pool of PKA-I found in lipid rafts. Using a combination of Far Western blotting, immunoprecipitation, proximity ligation assay and cAMP pull-down we identified moesin as an AKAP that potentially localizes PKA-I to rafts. Introduction of cell-permeable anchoring disruptor peptide, RI anchoring disruptor (RIAD-Arg11 ), to block PKA-I/AKAP interactions, uncoupled PKA-RI from moesin, displaced PKA-RI from rafts and reduced kinase activity in rafts. Examination of GPIbß demonstrated that it was phosphorylated in response to low concentrations of PGI2 in a PKA-dependent manner and occurred primarily in lipid raft fractions. RIAD-Arg11 caused a significant reduction in raft-localized phosphoGPIbß and diminished the ability of PGI2 to regulate VWF-mediated aggregation and thrombus formation in vitro. CONCLUSION: We propose that PKA-I-specific AKAPs in platelets, including moesin, organize a selective localization of PKA-I required for phosphorylation of GPIbß and contribute to inhibition of platelet VWF interactions.


Assuntos
Proteínas de Ancoragem à Quinase A/sangue , Proteína Quinase Tipo I Dependente de AMP Cíclico/sangue , AMP Cíclico/fisiologia , Microdomínios da Membrana , Adesividade Plaquetária/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Processamento de Proteína Pós-Traducional , Sistemas do Segundo Mensageiro/fisiologia , Proteínas de Ancoragem à Quinase A/fisiologia , Sequência de Aminoácidos , Proteína Quinase Tipo I Dependente de AMP Cíclico/antagonistas & inibidores , Epoprostenol/farmacologia , Humanos , Microdomínios da Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Dados de Sequência Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/farmacologia , Fosforilação , Glicoproteínas da Membrana de Plaquetas/metabolismo , Ligação Proteica , Isoformas de Proteínas/sangue , Inibidores de Proteínas Quinases/farmacologia , Fator de von Willebrand/metabolismo
7.
J Thromb Haemost ; 12(10): 1733-43, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25056834

RESUMO

BACKGROUND: Dissecting the signaling events that contribute to platelet activation will increase our understanding of platelet function and aid in the development of new antiplatelet agents. However, high-throughput methodology for the quantitative analysis of platelet signaling events is still lacking. OBJECTIVE: To develop a high-throughput assay for the analysis of platelet signaling events in whole blood. METHODS AND RESULTS: We developed a fluorescent barcoding protocol to facilitate multiplexing and enable large-scale signaling profiling in platelets in whole blood. The methodology allowed simultaneous staining and acquisition of 24-96 samples in a single analysis tube with a standard flow cytometer. This approach significantly reduced experimental numbers, data acquisition time, and antibody consumption, while providing automated statistically rich quantitative data on signaling events. Using vasodilator-stimulated phosphoprotein (VASP), an established marker of platelet inhibition and antiplatelet drug therapy, we demonstrated that the assay could detect subtle changes in phosphoVASP-Ser157/239 in response to cAMP-elevating agents of varying potency and known modulators of the cAMP signaling cascade. The assay could be used with washed platelets or whole blood, analyzed immediately or frozen, without any significant change in assay performance. To demonstrate the usefulness of the assay as a drug discovery platform, we examined a prostaglandin screening library. Our screen of 70 prostaglandin derivatives revealed three previously uncharacterized lipids that stimulated phosphorylation of VASP-Ser157. Follow-up analyses demonstrated that these agents elevated intraplatelet cAMP and inhibited collagen-induced platelet aggregation. CONCLUSIONS: This novel method enables rapid, large-scale quantitative signaling profiling and compound screening in human platelets present in whole blood.


Assuntos
Plaquetas/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Citometria de Fluxo , Animais , Anticorpos/química , Moléculas de Adesão Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Desenho de Fármacos , Eletroforese , Corantes Fluorescentes/química , Humanos , Camundongos , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Agregação Plaquetária , Inibidores da Agregação Plaquetária/química , Prostaglandinas/química , Transdução de Sinais
8.
J Thromb Haemost ; 7(12): 2106-15, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19765213

RESUMO

BACKGROUND: von Willebrand factor (VWF)-mediated platelet adhesion and spreading at sites of vascular injury is a critical step in hemostasis. This process requires two individual receptors: glycoprotein Ib (GPIb)-V-IX and integrin alpha(IIb)beta(3). However, little is known about the negative regulation of these events. OBJECTIVES: To examine if the endogenous platelet inhibitor nitric oxide (NO) has differential effects on adhesion, spreading and aggregation induced by immobilized VWF. RESULTS: S-nitrosoglutathione (GSNO) inhibited platelet aggregation on immobilized VWF under static and flow conditions, but had no effect on platelet adhesion. Primary signaling events underpinning the actions of NO required cyclic GMP but not protein kinase A. Dissecting the roles of GPIb and integrin alpha(IIb)beta(3) demonstrated that NO targeted alpha(IIb)beta(3)-mediated aggregation and spreading, but did not significantly influence GPIb-mediated adhesion. To understand the relationship between the effects of NO on adhesion and subsequent aggregation, we evaluated the activation of alpha(IIb)beta(3) on adherent platelets. NO reduced the phosphorylation of extracellular stimuli-responsive kinase (ERK) and p38, required for integrin activation resulting in reduced binding of the activated alpha(IIb)beta(3)-specific antibody PAC-1 on adherent platelets. Detailed analysis of platelet spreading initiated by VWF demonstrated key roles for integrin alpha(IIb)beta(3) and myosin light chain (MLC) phosphorylation. NO targeted both of these pathways by directly modulating integrin affinity and activating MLC phosphatase. CONCLUSION: These data demonstrate that initial activation-independent platelet adhesion to VWF via GPIb is resistant to NO, however, NO inhibits GPIb-mediated activation of alpha(IIb)beta(3) and MLC leading to reduced platelet spreading and aggregation.


Assuntos
Cadeias Leves de Miosina/metabolismo , Óxido Nítrico/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Fator de von Willebrand/metabolismo , Plaquetas/citologia , Células Cultivadas , Humanos , Glicoproteínas de Membrana/metabolismo , Adesividade Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIb-IX de Plaquetas
9.
J Thromb Haemost ; 6(12): 2121-31, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18983522

RESUMO

BACKGROUND: Platelet-derived nitric oxide (NO) has been shown to play conflicting roles in platelet function, although it is accepted that NO mediates its actions through soluble guanylyl cyclase (sGC). This confusion concerning the roles of platelet NO may have arisen because of an uncharacterized mechanism for activation of sGC. OBJECTIVES: To examine the ability of the novel platelet agonist globular adiponectin (gAd) to stimulate the NO-independent cGMP-protein kinase G (PKG) signaling cascade. METHODS: We used three independent markers of NO signaling, [(3)H]l-citrulline production, cGMP accrual, and immunoblotting of vasodilator-stimulated phosphoprotein (VASP), to examine the NO signaling cascade in response to gAd. RESULTS: gAd increased platelet cGMP formation, resulting in a dose- and time-dependent increase in phospho-VASP(157/239). Phosphorylation of VASP in response to gAd was mediated by both protein kinase A and PKG. Importantly, cGMP formation occurred in the absence of NO synthase (NOS) activation and in the presence of NOS inhibitors. Indeed, inhibition of the NOS signaling cascade had no influence on gAd-mediated platelet aggregation. Exploration of the mechanism demonstrated that NO-independent cGMP formation, phosphorylation of VASP and association of sGCalpha(1) with heat shock protein-90 induced by gAd were blocked under conditions that inhibited Src kinases, implying a tyrosine kinase-dependent mechanism. Indeed, sGCalpha1 was reversibly tyrosine phosphorylated in response to gAd, collagen, and collagen-related peptide, an effect that required Src kinases and downstream Ca(2+) mobilization. CONCLUSIONS: These data demonstrate activation of the platelet cGMP signaling cascade by a novel tyrosine kinase-dependent mechanism in the absence of NO.


Assuntos
Adiponectina/farmacologia , Plaquetas/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , GMP Cíclico/biossíntese , Óxido Nítrico/análise , Animais , Bovinos , Moléculas de Adesão Celular/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Humanos , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Fosforilação , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA