Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
J Pharm Bioallied Sci ; 15(Suppl 1): S180-S182, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37654390

RESUMEN

Introduction: Achieving a correct occlusion depends heavily on the axial inclination of the teeth, particularly the maxillary anteriors, which have the longest crowns. This study, which expanded the use of angular measures to the mandible, was created to determine the prevalence of adversely angulated canines using orthopantomograms. Methods: A sample of 200 people between the ages of 18 and 25 was chosen and separated into two groups based on the degree of crowding; one group served as the control group while the other, which experienced severe, crowding, served as the study group. All of the individuals had their Orthopantomogram and study models taken. Results: While canine angulation in controls typically ranges from 85 to 95 degrees, cases of crowding were observed to have angulation as low as 67 degrees and as high as 105 degrees. Conclusion: Orthopantomogram can be used to predict canine angulation, with distally and mesially angulated canines occurring at rates of roughly 18% and 37%, respectively.

2.
J Orthod Sci ; 12: 65, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38234635

RESUMEN

BACKGROUND AND OBJECTIVES: The curve of Spee is a naturally occurring phenomenon in the human dentition. Leveling the curve of Spee can affect the Frankfort-mandibular plane (FMP) Angle and thus the lower anterior facial height. This study examined the degree of change in FMP angle after leveling the curve of Spee in different malocclusion groups. METHODS: In this study, 75 patients who were aged >14 years and had undergone fixed appliance therapy using a 0.022-slot MBT bracket system were included. The pre- and post-treatment casts and lateral cephalograms of the patients were divided into three groups, namely Class I, II, and III malocclusions, with 25 patients in each group. The curve of Spee and FMP angle were measured before and after orthodontic treatment, and their correlation was evaluated. RESULTS: After leveling the curve of Spee, the FMP angle decreased in Class I and II groups and increased in Class III group. These results were statistically significant except in Class I malocclusion group. A mild positive correlation was observed between the curve of Spee and FMP angle in Class I and III malocclusion groups and a negative correlation in Class II malocclusion group. CONCLUSION: The change in FMP angle, following the leveling of the curve of Spee, in Class II and III malocclusion group, is attributed to alterations in lower anterior facial height. The study observed a mild positive correlation between the curve of Spee and FMP angle in Class I and III malocclusion groups and a negative correlation in Class II malocclusion group.

3.
Diab Vasc Dis Res ; 17(6): 1479164120963048, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33334146

RESUMEN

BACKGROUND: Fibrin network characteristics determine predisposition to cardiovascular disease (CVD). Individuals with type 1 (T1DM) and type 2 diabetes mellitus (T2DM) have higher risk of CVD and display deranged fibrin network structure. Those with maturity onset diabetes of the young (MODY) may also be at increased risk but their fibrin clot properties have not been studied. METHODS: Plasma clots properties from 13 individuals with HNF1A-MODY, 12 matched-individuals with T2DM and 12 with T1DM were studied using a validated turbidimetric assay and confocal microscopy. Plasma levels of fibrinogen, plasminogen activator inhibitor-1, complement C3 and C-reactive protein were also measured. RESULTS: MODY clot maximum absorbance was 0.37 ± 0.03 AU, similar to T1DM (0.32 ± 0.03 AU; p = 0.26), but lower than T2DM (0.49 ± 0.03 AU; p = 0.02), with confocal microscopy confirming structural differences. Clot lysis time in MODY was similar to T1DM (456 ± 50 and 402 ± 20 s, respectively; p = 0.09) but shorter than T2DM (588 ± 58 s; p = 0.006). Comparing inflammatory/thrombotic proteins in HNF1A-MODY and T2DM, C3 levels were lower in MODY than T2DM (0.58 ± 0.09 and 0.80 ± 0.1 mg/ml, respectively; p < 0.01). CONCLUSIONS: HNF1A-MODY fibrin network alterations are at least as pronounced as in T1DM but less thrombotic than T2DM clots. Differences in fibrin clot characteristics comparing HNF1A-MODY and T2DM may, in part, relate to lower C3 levels.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Fibrina/análisis , Trombosis/etiología , Adulto , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Complemento C3/análisis , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Femenino , Predisposición Genética a la Enfermedad , Factor Nuclear 1-alfa del Hepatocito/genética , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Inhibidor 1 de Activador Plasminogénico/sangre , Factores de Riesgo , Trombosis/sangre , Trombosis/diagnóstico , Adulto Joven
4.
Diabet Med ; 37(5): 726-734, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32141628

RESUMEN

Cardiovascular complications remain the main cause of mortality and morbidity in diabetes. This is related to advanced vascular pathology in this population, together with an enhanced thrombotic environment. The increased risk in thrombosis is secondary to platelet hyper-reactivity and increased levels and/or altered activity of coagulation factors. The current review is focused on the role of antiplatelet agents in modulating the thrombotic milieu in diabetes and improving vascular outcome in this high-risk population. We review the latest evidence for the use of aspirin in primary vascular prevention together with long-term treatment with this agent for secondary prevention. We also discuss the effects of the various P2Y12 inhibitors, including clopidogrel, prasugrel and ticagrelor, on both short- and long-term secondary vascular prevention. Moreover, we briefly review antiplatelet therapies in special groups of people including those intolerant to aspirin, individuals with peripheral vascular disease and those with cerebrovascular pathology. The overall aim of this review is to provide the healthcare professional with a pragmatic guide for the management of thrombotic risk using established antiplatelet therapies to improve vascular outcome in persons with diabetes.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Diabetes Mellitus/tratamiento farmacológico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Trombosis/prevención & control , Aspirina/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Clopidogrel/uso terapéutico , Diabetes Mellitus/sangre , Humanos , Clorhidrato de Prasugrel/uso terapéutico , Prevención Primaria , Prevención Secundaria , Ticagrelor/uso terapéutico
5.
J Thromb Haemost ; 16(11): 2300-2314, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30151891

RESUMEN

Essentials The metabolic integration processes required for platelet activation are unclear. The metabolic plasticity of human platelets were investigated. Activated platelets exhibit a glycolytic phenotype while preserving mitochondrial function. Platelets can switch freely between glucose/glycogen and fatty acids to support aggregation. SUMMARY: Background Platelet activation is an energy-dependent process, but the type and integrated use of metabolic fuels required to drive activation remain unclear. Objective To dissect the metabolic fuel and pathway plasticity required for platelet activation. Methods Platelet oxygen consumption rate and extracellular acidification rate were measured as markers of oxidative phosphorylation (OXPHOS) and glycolysis, respectively. Glucose and glycogen were quantified by enzyme-coupled fluorometric assay. Results Blood platelets switched freely between glycolysis and OXPHOS, using either glucose or fatty acids at rest. The transition of platelets from a quiescent to an activated state promoted rapid uptake of exogenous glucose, associated with a shift to a predominantly glycolytic phenotype coupled with a minor rise in mitochondrial oxygen consumption. Consistent with this metabolic plasticity, under nutrient-limiting conditions, platelets utilized glucose, glycogen or fatty acids independently to support activation. Importantly, the glycolytic switch occurred even in the absence of extracellular glucose, originating from endogenous glycogen. Focusing on the relative flexibility of mitochondrial fuel oxidation of glucose and fatty acids, we found that inhibition of oxidation of a single fuel was compensated for by increased oxidation of the other, but, when oxidation was inhibited, glycolysis was upregulated. Glutamine made little contribution to mitochondrial oxygen consumption. Analysis of platelet functional dependency on ATP from different pathways demonstrated that inhibition of both fuel oxidation and glycolysis were required to prevent agonist-driven platelet activation. Conclusion Platelets have significant metabolic fuel and pathway flexibility, but preferentially use glycolysis for ATP generation when activated.


Asunto(s)
Glucemia/metabolismo , Plaquetas/metabolismo , Adenosina Trifosfato/metabolismo , Coagulación Sanguínea , Ácidos Grasos/metabolismo , Glucógeno/metabolismo , Glucólisis , Humanos , Mitocondrias/metabolismo , Fosforilación Oxidativa , Oxígeno/metabolismo , Consumo de Oxígeno , Fenotipo , Fosforilación , Activación Plaquetaria , Receptor PAR-1/metabolismo , Receptores de Trombina/metabolismo , Trombina/metabolismo
6.
Diabetes Metab ; 44(5): 431-436, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29784564

RESUMEN

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.


Asunto(s)
Plaquetas , Diabetes Mellitus Tipo 2/sangre , Hipoglucemia/sangre , Activación Plaquetaria/fisiología , Adulto , Glucemia , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/fisiopatología , Femenino , Humanos , Hipoglucemia/fisiopatología , Masculino , Persona de Mediana Edad , Pruebas de Función Plaquetaria
7.
Acta Physiol (Oxf) ; 223(3): e13071, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29633517

RESUMEN

Platelet-based applications such as platelet-rich plasma (PRP) and platelet releasate have gained unprecedented attention in regenerative medicine across a variety of tissues as of late. The rationale behind utilizing PRP originates in the delivery of key cytokines and growth factors from α-granules to the targeted area, which in turn act as cell cycle regulators and promote the healing process across a variety of tissues. The aim of the present review is to assimilate current experimental evidence on the role of platelets as biomaterials in tissue regeneration, particularly in skeletal muscle, by integrating findings from human, animal and cell studies. This review is composed of 3 parts: firstly, we review key aspects of platelet biology that precede the preparation and use of platelet-related applications for tissue regeneration. Secondly, we critically discuss relevant evidence on platelet-mediated regeneration in skeletal muscle focusing on findings from (i) clinical trials, (ii) experimental animal studies and (iii) cell culture studies; and thirdly, we discuss the application of platelets in the regeneration of several other tissues including tendon, bone, liver, vessels and nerve. Finally, we review key technical variations in platelet preparation that may account for the large discrepancy in outcomes from different studies. This review provides an up-to-date reference tool for biomedical and clinical scientists involved in platelet-mediated tissue regenerative applications.


Asunto(s)
Plaquetas/fisiología , Músculo Esquelético/fisiología , Plasma Rico en Plaquetas , Regeneración , Medicina Regenerativa/métodos , Animales , Humanos
8.
Sci Rep ; 8(1): 3032, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29445102

RESUMEN

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.


Asunto(s)
Óxido Nítrico/metabolismo , Fibras de Estrés/metabolismo , Trombosis/metabolismo , Citoesqueleto de Actina/efectos de los fármacos , Plaquetas/efectos de los fármacos , Citoesqueleto/metabolismo , Voluntarios Sanos , Humanos , Óxido Nítrico/fisiología , Fosforilación , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Resistencia al Corte/fisiología , Fibras de Estrés/efectos de los fármacos , Trombosis/patología , Tromboxanos/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
9.
Sci Rep ; 7(1): 5582, 2017 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-28717253

RESUMEN

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.


Asunto(s)
Plaquetas/química , Epoprostenol/farmacología , Inhibidores de Agregación Plaquetaria/farmacología , Fibras de Estrés/efectos de los fármacos , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Agregación Plaquetaria/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
10.
J Thromb Haemost ; 15(8): 1668-1678, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28509344

RESUMEN

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.


Asunto(s)
Plaquetas/enzimología , Forma de la Célula , GMP Cíclico/metabolismo , Citoesqueleto/enzimología , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Activación Plaquetaria , Sistemas de Mensajero Secundario , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Plaquetas/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Citoesqueleto/efectos de los fármacos , Humanos , Cadenas Ligeras de Miosina/metabolismo , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Fosforilación , Activación Plaquetaria/efectos de los fármacos , S-Nitrosoglutatión/metabolismo , S-Nitrosoglutatión/farmacología , Sistemas de Mensajero Secundario/efectos de los fármacos , Trombina/farmacología , Factores de Tiempo
11.
J Thromb Haemost ; 13(9): 1721-34, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26176741

RESUMEN

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.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/sangre , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/sangre , AMP Cíclico/fisiología , Microdominios de Membrana , Adhesividad Plaquetaria/fisiología , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Procesamiento Proteico-Postraduccional , Sistemas de Mensajero Secundario/fisiología , Proteínas de Anclaje a la Quinasa A/fisiología , Secuencia de Aminoácidos , Proteína Quinasa Tipo I Dependiente de AMP Cíclico/antagonistas & inhibidores , Epoprostenol/farmacología , Humanos , Microdominios de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Datos de Secuencia Molecular , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/farmacología , Fosforilación , Glicoproteínas de Membrana Plaquetaria/metabolismo , Unión Proteica , Isoformas de Proteínas/sangre , Inhibidores de Proteínas Quinasas/farmacología , Factor de von Willebrand/metabolismo
12.
J Thromb Haemost ; 12(10): 1733-43, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25056834

RESUMEN

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.


Asunto(s)
Plaquetas/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Citometría de Flujo , Animales , Anticuerpos/química , Moléculas de Adhesión Celular/metabolismo , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diseño de Fármacos , Electroforesis , Colorantes Fluorescentes/química , Humanos , Ratones , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/química , Prostaglandinas/química , Transducción de Señal
13.
Clin Endocrinol (Oxf) ; 79(2): 252-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23278130

RESUMEN

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.


Asunto(s)
Plaquetas/fisiología , Grosor Intima-Media Carotídeo , Obesidad/fisiopatología , Síndrome del Ovario Poliquístico/fisiopatología , Adulto , Enfermedades Cardiovasculares/etiología , Endotelio Vascular/fisiopatología , Femenino , Humanos , Resistencia a la Insulina , Isoprostanos/orina , Obesidad/sangre , Activación Plaquetaria , Síndrome del Ovario Poliquístico/sangre , Factores de Riesgo
14.
J Thromb Haemost ; 9(4): 820-33, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21320283

RESUMEN

BACKGROUND: Activated coagulation factor XIII (FXIIIa) is a transglutaminase that crosslinks fibrin at sites of vascular injury. FXIIIa also associates with blood platelets, although its role in platelet function is unclear and requires clarification. OBJECTIVES: To evaluate the ability of FXIIIa to support platelet adhesion and spreading under conditions of physiologic flow, and to identify the underpinning receptors and signaling events. METHODS AND RESULTS: Platelet adhesion to immobilized FXIIIa was measured by fluorescence microscopy, and signaling events were characterized by immunoblotting. Immobilized FXIIIa supported platelet adhesion and spreading under static conditions through mechanisms that were dually and differentially dependent on integrins α(IIb)ß(3) and α(v)ß(3). Platelet adhesion was independent of FXIIIa transglutaminase or protein disulfide isomerase activity. Moreover, adhesion was abolished by antibodies that prevented interaction with FXIIIa, but maintained when potential interactions with fibrinogen were blocked. Platelet adhesion to FXIIIa was reduced significantly by either the specific α(IIb)ß(3) antagonist tirofiban or the selective α(v)ß(3)-blocking antibody LM609, and abolished when they were used in combination. Importantly, platelet adhesion was preserved under venous and arterial flow conditions in which both integrins played essential roles. In contrast, FXIIIa stimulated the formation of filopodia and lamellipodia in adherent platelets that was mediated exclusively by α(IIb)ß(3) and eliminated by the Src-family inhibitor 4-amino-5-(4-methylphenyl-7-(t-butyl)pyrazolo(3,4-d)pyrimidine, indicating a tyrosine kinase-dependent mechanism. Crucially, under conditions of arterial shear, FXIIIa accentuated platelet recruitment by von Willebrand factor and collagen. CONCLUSIONS: Our data demonstrate a potential role for FXIIIa in supporting platelet adhesion at sites of vascular damage, particularly in association with other thrombogenic matrix proteins.


Asunto(s)
Proteínas de la Matriz Extracelular/fisiología , Factor XIII/fisiología , Activación Plaquetaria/fisiología , Trombosis/fisiopatología , Plaquetas/citología , Adhesión Celular , Humanos , Inmunoprecipitación , Integrina alfaVbeta3/fisiología , Microscopía Fluorescente , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología
15.
J Thromb Haemost ; 7(12): 2106-15, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19765213

RESUMEN

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.


Asunto(s)
Cadenas Ligeras de Miosina/metabolismo , Óxido Nítrico/farmacología , Activación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Factor de von Willebrand/metabolismo , Plaquetas/citología , Células Cultivadas , Humanos , Glicoproteínas de Membrana/metabolismo , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIb-IX de Glicoproteína Plaquetaria
16.
J Thromb Haemost ; 6(12): 2175-85, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18983487

RESUMEN

BACKGROUND: Nitric oxide (NO) inhibits platelet adhesion to collagen, although the precise molecular mechanisms underlying this process are unclear. OBJECTIVES: Collagen-mediated adhesion is a multifaceted event requiring multiple receptors and platelet-derived soluble agonists. We investigated the influence of NO on these processes. RESULTS: S-nitrosoglutathione (GSNO) induced a concentration-dependent inhibition of platelet adhesion to immobilized collagen. Maximal adhesion to collagen required platelet-derived ADP and TxA(2). GSNO-mediated inhibition was lost in the presence of apyrase and indomethacin, suggesting that NO reduced the availability of, or signaling by, ADP and TxA(2). Exogenous ADP, but not the TxA(2) analogue U46619, reversed the inhibitory actions of GSNO on adhesion. Under adhesive conditions NO inhibited dense granule secretion but did not influence TxA(2) generation. These data indicated that NO may block signaling by TxA(2) required for dense granule secretion, thereby reducing the availability of ADP. Indeed, we found TxA(2)-mediated activation of PKC was required to drive dense granule secretion, a pathway that was inhibited by NO. Because our data demonstrated that NO only inhibited the activation-dependent component of adhesion, we investigated the effects of NO on individual collagen receptors. GSNO inhibited platelet adhesion and spreading on alpha(2)beta(1) specific peptide ligand GFOGER. In contrast, GSNO did not inhibit GPVI-mediated adhesion to collagen, or adhesion to the GPVI specific ligand, collagen related peptide (CRP). CONCLUSIONS: NO targets activation-dependent adhesion mediated by alpha(2)beta(1), possibly by reducing bioavailability of platelet-derived ADP, but has no effect on activation-independent adhesion mediated by GPVI. Thus, NO regulates platelet spreading and stable adhesion to collagen.


Asunto(s)
Colágeno/metabolismo , Integrinas/metabolismo , Óxido Nítrico/farmacología , Adhesividad Plaquetaria/efectos de los fármacos , Adenosina Difosfato , Plaquetas/química , Plaquetas/citología , Células Cultivadas , Humanos , Integrina alfa2beta1/metabolismo , Péptidos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Unión Proteica/efectos de los fármacos , S-Nitrosoglutatión/farmacología , Tromboxano A2
17.
J Thromb Haemost ; 6(12): 2121-31, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18983522

RESUMEN

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.


Asunto(s)
Adiponectina/farmacología , Plaquetas/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , GMP Cíclico/biosíntesis , Óxido Nítrico/análisis , Animales , Bovinos , Moléculas de Adhesión Celular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Humanos , Proteínas de Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Fosforilación , Transducción de Señal
19.
J Thromb Haemost ; 6(6): 1012-20, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18419742

RESUMEN

BACKGROUND: The adipocyte-derived cytokine, adiponectin (Ad), exerts potent vascular effects, although the direct effects of Ad on blood platelets are unclear. OBJECTIVE: The influence of globular Ad (gAd) on blood platelet function was investigated. RESEARCH DESIGN AND METHODS: We measured platelet aggregation and tyrosine phosphorylation signaling events in human and mouse platelets. The ability of gAd to activate Glycoprotein VI (GPVI) activity was determined with a NFAT luciferase reporter assay. RESULTS: gAd, but not full length Ad, induced rapid aggregation and granule secretion of human and mouse platelets through a pathway that is ablated under conditions of Src kinase inhibition, indicating a tyrosine kinase-dependent mechanism. Consistent with this, gAd stimulates rapid tyrosine phosphorylation of several proteins in human and mouse platelets. The pattern of increase in tyrosine phosphorylation was similar to that induced by collagen, with the tyrosine kinase Syk and PLCgamma2 being identified among the list of tyrosine phosphorylated proteins. As collagen activates platelet through the GPVI-Fc receptor gamma-chain (FcRgamma) complex, we used FcRgamma null platelets (which also lack GPVI) to explore the mechanism by which gAd stimulates platelets. Stimulation of tyrosine phosphorylation and platelet aggregation by gAd was abolished in FcRgamma null platelets and markedly reduced in the absence of PLCgamma2. Further, GPVI was confirmed as a collagen receptor for gAd by increased luciferase activity in Jurkat T-cells transfected with GPVI. CONCLUSIONS: We identify gAd as a novel ligand for GPVI that stimulates tyrosine kinase-dependent platelet aggregation. Our data raise the possibility that gAd may promote unwanted platelet activation at sites of vascular injury.


Asunto(s)
Adiponectina/metabolismo , Regulación de la Expresión Génica , Activación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Fc/metabolismo , Animales , Plaquetas/metabolismo , Humanos , Células Jurkat , Ratones , Fosforilación , Agregación Plaquetaria , Isoformas de Proteínas , Linfocitos T/citología
20.
J Thromb Haemost ; 6(1): 10-9, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17944990

RESUMEN

Endothelial-derived nitric oxide (NO) is a key regulator of platelet function, inhibiting both adhesion to the extracellular matrix and aggregation at sites of vascular injury. Platelets also have the capacity to synthesize and release bioactive NO, which is thought to make a significant contribution to the vascular pool of NO. The regulation of platelet NO production is poorly understood and studies examining the physiological role of platelet-derived NO have produced contradictory and controversial findings. In the present article, we discuss the current understanding of the biochemical and molecular regulation of platelet NO synthesis and outline the potential physiological and clinical significance of this molecule.


Asunto(s)
Plaquetas/enzimología , Óxido Nítrico Sintasa de Tipo III/fisiología , Plaquetas/metabolismo , Humanos , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III/metabolismo , Activación Plaquetaria , Trombosis/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...