Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nanoscale Adv ; 6(5): 1497-1506, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38419863

RESUMEN

In-stent restenosis (ISR) and stent thrombosis (ST) are the most serious complications of coronary angioplasty and stenting. Although the evolution of drug-eluting stents (DES) has significantly restricted the incidence of ISR, they are associated with an enhanced risk of ST. In the present study, we explore the photothermal ablation of a thrombus using a nano-enhanced thermogenic stent (NETS) as a modality for revascularization following ST. The photothermal activity of NETS, fabricated by coating bare metal stents with gold nanorods generating a thin plasmonic film of gold, was found to be effective in rarefying clots formed within the stent lumen in various in vitro assays including those under conditions mimicking blood flow. NETS implanted in the rat common carotid artery generated heat following exposure to a NIR-laser that led to effective restoration of blood flow within the occluded vessel in a model of ferric chloride-induced thrombosis. Our results present a proof-of-concept for a novel photothermal ablation approach by employing coated stents in the non-invasive management of ST.

2.
Cell Commun Signal ; 22(1): 13, 2024 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-38172855

RESUMEN

Circulating platelets derived from bone marrow megakaryocytes play a central role in thrombosis and hemostasis. Despite being anucleate, platelets express several proteins known to have nuclear niche. These include transcription factors and steroid receptors whose non-genomic functions are being elucidated in platelets. Quite remarkably, components of some of the best-studied morphogen pathways, namely Notch, Sonic Hedgehog (Shh), and Wnt have also been described in recent years in platelets, which regulate platelet function in the context of thrombosis as well as influence their survival. Shh and Notch pathways in stimulated platelets establish feed-forward loops of autocrine/juxtacrine/paracrine non-canonical signaling that helps perpetuate thrombosis. On the other hand, non-canonical Wnt signaling is part of a negative feedback loop for restricting platelet activation and possibly limiting thrombus growth. The present review will provide an overview of these signaling pathways in general. We will then briefly discuss the non-genomic roles of transcription factors and steroid receptors in platelet activation. This will be followed by an elaborate description of morphogen signaling in platelets with a focus on their bearing on platelet activation leading to hemostasis and thrombosis as well as their potential for therapeutic targeting in thrombotic disorders.


Asunto(s)
Receptores de Esteroides , Trombosis , Humanos , Proteínas Hedgehog/metabolismo , Plaquetas/metabolismo , Activación Plaquetaria , Trombosis/metabolismo , Receptores de Esteroides/metabolismo
3.
bioRxiv ; 2023 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-37662286

RESUMEN

Background: Antibodies to ß2-glycoprotein I (ß2GPI) cause thrombosis in antiphospholipid syndrome, however the role of ß2GPI itself in regulation of coagulation pathways in vivo is not well understood. Methods: We developed ß2GPI-deficient mice (Apoh -/- ) by deleting exon 2 and 3 of Apoh using CRISPR/Cas9 and compared the propensity of wild-type (WT) and Apoh -/- mice to develop thrombosis using rose bengal and FeCl 3 -induced carotid thrombosis, laser-induced cremaster arteriolar injury, and inferior vena cava (IVC) stasis models. We also compared tail bleeding times and assessed platelet activation in WT and Apoh -/- mice in the absence and presence of exogenous ß2GPI. Results: Compared to WT littermates, Apoh -/- mice demonstrated a prolonged time to occlusion of the carotid artery after exposure to rose bengal or FeCl 3 , and reduced platelet and fibrin accumulation in cremasteric arterioles after laser injury. Similarly, significantly smaller thrombi were retrieved from the IVC of Apoh -/- mice 48 hours after IVC occlusion. The activated partial thromboplastin time (aPTT) and prothrombin time, as well as aPTT reagent- and tissue factor-induced thrombin generation times using plasma from Apoh -/- and WT mice revealed no differences. However, we observed significant prolongation of tail bleeding in Apoh -/- mice, and reduced P-selectin expression and binding of fibrinogen to the activated α2bß3 integrin on platelets from these mice after stimulation with low thrombin concentrations; these changes were reversed by exogenous ß2GPI. An antibody to PAR3 blocked thrombin-induced activation of WT, but not Apoh -/- platelets, as well as the ability of ß2GPI to restore the activation response of Apoh -/- platelets to thrombin. ß2GPI deficiency did not affect platelet activation by a PAR4-activator peptide, or ADP. Conclusions: In mice, ß2GPI may mediate procoagulant activity by enhancing the ability of PAR3 to present thrombin to PAR4, promoting platelet activation at low thrombin concentrations. Key Points: ß2GPI deficient mice are protected from experimental arterial, venous, and microvascular thrombosis.ß2GPI deficient mice display prolonged tail bleeding times and reduced PAR3-facilitated platelet activation by low concentrations of thrombin.

5.
Cell Death Differ ; 30(8): 1886-1899, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37301927

RESUMEN

Necroptosis is a form of programmed cell death executed by receptor-interacting serine/threonine protein kinase 1 (RIPK1), RIPK3, and mixed lineage kinase domain-like (MLKL). Platelets are circulating cells that play central roles in haemostasis and pathological thrombosis. In this study we demonstrate seminal contribution of MLKL in transformation of agonist-stimulated platelets to active haemostatic units progressing eventually to necrotic death on a temporal scale, thus attributing a yet unrecognized fundamental role to MLKL in platelet biology. Physiological agonists like thrombin instigated phosphorylation and subsequent oligomerization of MLKL in platelets in a RIPK3-independent but phosphoinositide 3-kinase (PI3K)/AKT-dependent manner. Inhibition of MLKL significantly curbed agonist-induced haemostatic responses in platelets that included platelet aggregation, integrin activation, granule secretion, procoagulant surface generation, rise in intracellular calcium, shedding of extracellular vesicles, platelet-leukocyte interactions and thrombus formation under arterial shear. MLKL inhibition, too, prompted impairment in mitochondrial oxidative phosphorylation and aerobic glycolysis in stimulated platelets, accompanied with disruption in mitochondrial transmembrane potential, augmented proton leak and drop in both mitochondrial calcium as well as ROS. These findings underscore the key role of MLKL in sustaining OXPHOS and aerobic glycolysis that underlie energy-intensive platelet activation responses. Prolonged exposure to thrombin provoked oligomerization and translocation of MLKL to plasma membranes forming focal clusters that led to progressive membrane permeabilization and decline in platelet viability, which was prevented by inhibitors of PI3K/MLKL. In summary, MLKL plays vital role in transitioning of stimulated platelets from relatively quiescent cells to functionally/metabolically active prothrombotic units and their ensuing progression to necroptotic death.


Asunto(s)
Plaquetas , Proteínas Quinasas , Proteínas Quinasas/metabolismo , Plaquetas/metabolismo , Necroptosis , Calcio/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Trombina/farmacología , Trombina/metabolismo , Muerte Celular , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
6.
Metabolism ; 145: 155596, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37244415

RESUMEN

Platelets are circulating cells central to haemostasis that follows vessel injury, as well as thrombosis that ensues as a consequence of pathological stasis or plaque rupture. Platelet responses to various stimuli that mediate these processes are all energy-intensive. Hence, platelets need to adapt their energy metabolism to fulfil the requirements of clot formation while overcoming the adversities of the thrombus niche such as restricted access to oxygen and nutrient. In the present review, we describe the changes in energy metabolism of platelets upon agonist challenge and their underlying molecular mechanisms. We briefly discuss the metabolic flexibility and dependency of stimulated platelets in terms of choice of energy substrates. Finally, we discuss how targeting the metabolic vulnerabilities of stimulated platelets such as aerobic glycolysis and/or beta oxidation of fatty acids could forestall platelet activation and thrombus formation. Thus, we present a case for modulating platelet energy metabolism using small-molecules as a novel anti-platelet strategy in the management of vaso-occlusive disorders like acute myocardial infarction, ischemic stroke, deep vein thrombosis and pulmonary embolism.


Asunto(s)
Plaquetas , Trombosis , Humanos , Metabolismo Energético , Ácidos Grasos/metabolismo , Activación Plaquetaria , Agregación Plaquetaria
7.
FASEB J ; 37(2): e22768, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36624703

RESUMEN

Platelet mitochondria possess remarkable plasticity for oxidation of energy substrates, where metabolic dependency on glucose or fatty acids is higher than glutamine. Since platelets metabolize nearly the entire pool of glucose to lactate rather than fluxing through mitochondrial tricarboxylic acid cycle, we posit that majority of mitochondrial ATP, which is essential for platelet granule secretion and thrombus formation, is sourced from oxidation of fatty acids. We performed a comprehensive analysis of bioenergetics and function of stimulated platelets in the presence of etomoxir, trimetazidine and oxfenicine, three pharmacologically distinct inhibitors of ß-oxidation. Each of them significantly impaired oxidative phosphorylation in unstimulated as well as thrombin-stimulated platelets leading to a small but consistent drop in ATP level in activated cells due to a lack of compensation from glycolytic ATP. Trimetazidine and oxfenicine attenuated platelet aggregation, P-selectin externalization and integrin αIIb ß3 activation. Both etomoxir and trimetazidine impeded agonist-induced dense granule release and platelet thrombus formation on collagen under arterial shear. The effect of inhibitors on platelet aggregation and dense granule release was dose- and incubation time- dependent with significant inhibition at higher doses and prolonged incubation times. Neither of the inhibitors could protect mice from collagen-epinephrine-induced pulmonary embolism or prolong mouse tail bleeding times. However, mice pre-administered with etomoxir, trimetazidine and oxfenicine were protected from ferric chloride-induced mesenteric thrombosis. In conclusion, ß-oxidation of fatty acids sustains ATP level in stimulated platelets and is therefore essential for energy-intensive agonist-induced platelet responses. Thus, fatty acid oxidation may constitute an attractive therapeutic target for novel antiplatelet agents.


Asunto(s)
Trombosis , Trimetazidina , Ratones , Animales , Ácidos Grasos/metabolismo , Trimetazidina/efectos adversos , Trimetazidina/metabolismo , Plaquetas/metabolismo , Activación Plaquetaria , Agregación Plaquetaria , Trombosis/inducido químicamente , Trombosis/prevención & control , Trombosis/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/efectos adversos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Fosforilación Oxidativa , Colágeno/metabolismo , Adenosina Trifosfato/metabolismo , Glucosa/metabolismo
8.
Blood Adv ; 6(17): 5024-5040, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-35704688

RESUMEN

Sonic Hedgehog (Shh) is a morphogen in vertebrate embryos that is also associated with organ homeostasis in adults. We report here that human platelets, though enucleate, synthesize Shh from preexisting mRNAs upon agonist stimulation, and mobilize it for surface expression and release on extracellular vesicles, thus alluding to its putative role in platelet activation. Shh, in turn, induced a wave of noncanonical signaling in platelets leading to activation of small GTPase Ras homolog family member A and phosphorylation of myosin light chain in activated protein kinase-dependent manner. Remarkably, agonist-induced thrombogenic responses in platelets, which include platelet aggregation, granule secretion, and spreading on immobilized fibrinogen, were significantly attenuated by inhibition of Hedgehog signaling, thus, implicating inputs from Shh in potentiation of agonist-mediated platelet activation. In consistence, inhibition of the Shh pathway significantly impaired arterial thrombosis in mice. Taken together, the above observations strongly support a feed-forward loop of platelet stimulation triggered locally by Shh, similar to ADP and thromboxane A2, that contributes significantly to the stability of occlusive arterial thrombus and that can be investigated as a potential therapeutic target in thrombotic disorders.


Asunto(s)
Plaquetas , Proteínas Hedgehog , Trombosis , Animales , Plaquetas/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Activación Plaquetaria , Agregación Plaquetaria , Transducción de Señal , Trombosis/metabolismo
9.
Gynecol Endocrinol ; 38(4): 288-295, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35007186

RESUMEN

BACKGROUND: Altered irisin levels have been reported in pregnancy-associated disorders, such as preeclampsia. OBJECTIVE: A systematic review and meta-analysis were conducted to evaluate the changes in maternal circulatory irisin levels in preeclampsia as compared to normotensive healthy pregnant controls. METHODS: Relevant studies were identified by searching PubMed and other databases. Random-effects model was used to obtain standardized mean differences (SMDs) and its 95% confidence intervals (CIs). The sub-group meta-regression analyses were conducted to explore heterogeneity. The presence of publication bias and the study robustness was tested using funnel plot and sensitivity analyses, respectively. RESULTS: This meta-analysis finally included 14 observations from eight studies. Compared with controls, preeclampsia patients showed significantly decreased serum irisin levels (SMD: -1.13; 95% CI: -1.63 to -0.62, p < .0001). The sub-group analysis showed that this decrease in irisin is regardless of body mass index (BMI) and gestational age of preeclampsia patients. The meta-regression analysis indicated that blood pressure is significantly associated with the observed results. There was no significant publication bias as indicated by the funnel plot analysis followed by Begg's (p = .35) and Egger's tests (p = .39). The sensitivity analysis indicated that no single study had a significant influence on the overall outcome, suggesting the robustness of this meta-analysis. CONCLUSIONS: This meta-analysis showed significantly decreased maternal serum irisin level in preeclampsia patients as compared to normotensive pregnant women. This study highlights the need for future studies evaluating the diagnostic utilities and associations of irisin with the fetal and neonatal outcomes in preeclampsia.


Asunto(s)
Preeclampsia , Presión Sanguínea , Femenino , Edad Gestacional , Humanos , Recién Nacido , Embarazo
11.
Sci Rep ; 11(1): 21888, 2021 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-34750495

RESUMEN

Hypercoagulability and the need for prioritizing coagulation markers for prognostic abilities have been highlighted in COVID-19. We aimed to quantify the associations of D-dimer with disease progression in patients with COVID-19. This systematic review and meta-analysis was registered with PROSPERO, CRD42020186661.We included 113 studies in our systematic review, of which 100 records (n = 38,310) with D-dimer data) were considered for meta-analysis. Across 68 unadjusted (n = 26,960) and 39 adjusted studies (n = 15,653) reporting initial D-dimer, a significant association was found in patients with higher D-dimer for the risk of overall disease progression (unadjusted odds ratio (uOR) 3.15; adjusted odds ratio (aOR) 1.64). The time-to-event outcomes were pooled across 19 unadjusted (n = 9743) and 21 adjusted studies (n = 13,287); a strong association was found in patients with higher D-dimers for the risk of overall disease progression (unadjusted hazard ratio (uHR) 1.41; adjusted hazard ratio (aHR) 1.10). The prognostic use of higher D-dimer was found to be promising for predicting overall disease progression (studies 68, area under curve 0.75) in COVID-19. Our study showed that higher D-dimer levels provide prognostic information useful for clinicians to early assess COVID-19 patients at risk for disease progression and mortality outcomes. This study, recommends rapid assessment of D-dimer for predicting adverse outcomes in COVID-19.


Asunto(s)
COVID-19/diagnóstico , COVID-19/inmunología , Productos de Degradación de Fibrina-Fibrinógeno/química , Adulto , Anciano , Área Bajo la Curva , Biomarcadores/sangre , COVID-19/epidemiología , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Pronóstico , Modelos de Riesgos Proporcionales , Respiración Artificial , Riesgo , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Trombofilia/sangre
12.
Thromb Res ; 196: 175-185, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32890901

RESUMEN

AMP-activated protein kinase (AMPK) is a metabolic master switch that has critical role in wide range of pathologies including cardiovascular disorders. As AMPK-α2 knockout mice exhibit impaired thrombus stability, we asked whether pharmacological inhibition of AMPK with a specific small-molecule inhibitor, compound C, could protect against arterial thrombosis without affecting hemostasis. Mice pre-administered with compound C exhibited decreased mesenteric arteriolar thrombosis but normal tail bleeding time compared to vehicle-treated animals. Compound C potently restricted platelet aggregation, clot retraction and integrin activation induced by thrombin and collagen. It impaired platelet spreading on both immobilized fibrinogen and collagen matrices; it, however, had no significant effect on thrombin-induced phosphatidylserine exposure that is characteristic of procoagulant platelets. In parallel, compound C brought about significant drop in thrombin-induced phosphorylation of myosin light chain (MLC) and MLC phosphatase (MYPT1) as well as abrogated rise in level of RhoA-GTP in thrombin-stimulated platelets. Thus, effects of compound C on agonist-induced platelet responses could be at least in part attributed to modulation of cytoskeletal changes mediated by RhoA-MYPT1-MLC signaling. An ideal antithrombotic drug would spare hemostatic responses that maintain vascular integrity while preferentially protecting against thrombosis. The present study suggests that AMPK could be one such potential therapeutic target.


Asunto(s)
Plaquetas , Trombosis , Proteínas Quinasas Activadas por AMP , Animales , Hemostasis , Ratones , Activación Plaquetaria , Agregación Plaquetaria , Trombosis/tratamiento farmacológico , Trombosis/prevención & control
13.
Mol Med ; 25(1): 34, 2019 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-31362709

RESUMEN

Following publication of the original article [1], the author reported an error in Figure 1. The correct version of Figure 1 is as follows.

14.
Haematologica ; 104(12): 2482-2492, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31004026

RESUMEN

Oxygen-compromised environments, such as high altitude, are associated with platelet hyperactivity. Platelets confined within the relatively impervious core of an aggregate/thrombus have restricted access to oxygen, yet they continue to perform energy-intensive procoagulant activities that sustain the thrombus. Studying platelet signaling under hypoxia is, therefore, critical to our understanding of the mechanistic basis of thrombus stability. We report here that hypoxia-inducible factor (HIF)-2α is translated from pre-existing mRNA and stabilized against proteolytic degradation in enucleate platelets exposed to hypoxia. Hypoxic stress, too, stimulates platelets to synthesize plasminogen-activator inhibitor-1 (PAI-1) and shed extracellular vesicles, both of which potentially contribute to the prothrombotic phenotype associated with hypoxia. Stabilization of HIF-α by administering hypoxia-mimetics to mice accelerates thrombus formation in mesenteric arterioles. In agreement, platelets from patients with chronic obstructive pulmonary disease and high altitude residents exhibiting thrombogenic attributes have abundant expression of HIF-2α and PAI- 1. Thus, targeting platelet hypoxia signaling could be an effective anti-thrombotic strategy.


Asunto(s)
Mal de Altura/complicaciones , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Plaquetas/patología , Vesículas Extracelulares/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Trombosis/patología , Animales , Plaquetas/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Estudios de Seguimiento , Humanos , Ratones , Pronóstico , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Transducción de Señal , Trombosis/etiología , Trombosis/metabolismo
15.
Haematologica ; 104(4): 806-818, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30381300

RESUMEN

Platelets are critical to arterial thrombosis, which underlies myocardial infarction and stroke. Activated platelets, regardless of the nature of their stimulus, initiate energy-intensive processes that sustain thrombus, while adapting to potential adversities of hypoxia and nutrient deprivation within the densely packed thrombotic milieu. We report here that stimulated platelets switch their energy metabolism to aerobic glycolysis by modulating enzymes at key checkpoints in glucose metabolism. We found that aerobic glycolysis, in turn, accelerates flux through the pentose phosphate pathway and supports platelet activation. Hence, reversing metabolic adaptations of platelets could be an effective alternative to conventional anti-platelet approaches, which are crippled by remarkable redundancy in platelet agonists and ensuing signaling pathways. In support of this hypothesis, small-molecule modulators of pyruvate dehydrogenase, pyruvate kinase M2 and glucose-6-phosphate dehydrogenase, all of which impede aerobic glycolysis and/or the pentose phosphate pathway, restrained the agonist-induced platelet responses ex vivo These drugs, which include the anti-neoplastic candidate, dichloroacetate, and the Food and Drug Administration-approved dehydroepiandrosterone, profoundly impaired thrombosis in mice, thereby exhibiting potential as anti-thrombotic agents.


Asunto(s)
Plaquetas/metabolismo , Fibrinolíticos/farmacología , Glucólisis/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Trombosis , Aerobiosis/efectos de los fármacos , Animales , Femenino , Humanos , Masculino , Ratones , Vía de Pentosa Fosfato/efectos de los fármacos , Trombosis/tratamiento farmacológico , Trombosis/metabolismo , Trombosis/patología
16.
Mol Med ; 22: 224-232, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27262026

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disorder, characterized by extensive loss of neurons, and deposition of amyloid beta (Aß) in the form of extracellular plaques. Aß is considered to have critical role in synaptic loss and neuronal death underlying cognitive decline. Platelets contribute to 95% of circulating amyloid-precursor protein that releases Aß into circulation. We have recently demonstrated that, Aß active fragment containing amino acid sequence 25-35 (Aß25-35) is highly thrombogenic in nature, and elicits strong aggregation of washed human platelets in RhoA-dependent manner. In the present study we evaluated the influence of fibrinogen on Aß-induced platelet activation. Intriguingly, Aß failed to induce aggregation of platelets suspended in plasma but not in buffer. Fibrinogen brought about dose-dependent decline in aggregatory response of washed human platelets elicited by Aß25-35, which could be reversed by increasing doses of Aß. Fibrinogen also attenuated Aß-induced platelet responses like secretion, clot retraction, rise in cytosolic Ca+2 and reactive oxygen species (ROS). Fibrinogen prevented intracellular accumulation of full length amyloid beta peptide (Aß42) in platelets as well as neuronal cells. We conclude that fibrinogen serves as a physiological check against the adverse effects of Aß by preventing its interaction with cells.

17.
Blood ; 125(20): 3164-72, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25700433

RESUMEN

Cellular fibronectin containing extra domain A (Fn-EDA+), which is produced in response to tissue injury in several disease states, has prothrombotic activity and is known to interact with Toll-like-receptor 4 (TLR4). The underlying mechanism and cell types involved in mediating the prothrombotic effect of Fn-EDA+ still remain unknown. Using intravital microscopy, we evaluated susceptibility to carotid artery thrombosis after FeCl3-induced injury in mice expressing Fn lacking EDA (Fn-EDA(-/-) mice) or Fn containing EDA (Fn-EDA(+/+) mice). Fn-EDA(-/-) mice exhibited prolonged times to first thrombus formation and complete occlusion and a significant decrease in the rate of thrombus growth (P < .05 vs Fn-EDA(+/+) mice). Genetic deletion of TLR4 reversed the accelerated thrombosis in Fn-EDA(+/+) mice (P < .05) but had no effect in Fn-EDA(-/-) mice. Bone marrow transplantation experiments revealed that TLR4 expressed on hematopoietic cells contributes to accelerated thrombosis in Fn-EDA(+/+) mice. In vitro studies showed that cellular Fn-EDA+ interacts with platelet TLR4 and promotes agonist-induced platelet aggregation. Finally, Fn-EDA(+/+) mice specifically lacking platelet TLR4 exhibited prolonged times to first thrombus formation and complete occlusion (P < .05 vs Fn-EDA(+/+) mice containing platelet TLR4). We conclude that platelet TLR4 contributes to the prothrombotic effect of cellular Fn-EDA+, suggesting another link between thrombosis and innate immunity.


Asunto(s)
Plaquetas/metabolismo , Fibronectinas/metabolismo , Trombosis/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Modelos Animales de Enfermedad , Femenino , Fibronectinas/genética , Inmunidad Innata , Masculino , Ratones , Ratones Noqueados , Agregación Plaquetaria , Trombosis/genética , Trombosis/inmunología
18.
Blood ; 125(2): 399-406, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25343959

RESUMEN

Thrombospondin 1 (TSP1) has been suggested as a counter receptor to platelet glycoprotein Ibα that supports initial platelet adhesion in absence of von Willebrand factor (VWF). Conversely, several other studies have shown that TSP1 interacts with VWF and may play a mechanistic role in modulating thrombosis. However, the in vivo evidence to support this mechanism remains unclear. Using intravital microscopy, in a 10% FeCl3-induced thrombosis model, we report similar platelet adhesion in Tsp1(-/-)/Vwf(-/-) mice compared with littermate Vwf(-/-) mice, suggesting that TSP1 does not mediate initial platelet adhesion in the absence of VWF. Tsp1(-/-) mice exhibited prolonged occlusion time and a significant decrease in the rate of thrombus growth (P < .05 vs wild-type), but not in the initial platelet adhesion. Complete deficiency of VWF abrogated the rate of thrombus growth in Tsp1(-/-) mice; therefore, we generated Tsp1(-/-)/Vwf(+/-) mice to determine whether TSP1 modulates thrombus growth under conditions of partial VWF deficiency. Tsp1(-/-)/Vwf(+/-) mice exhibited delayed thrombus growth kinetics and prolonged occlusion time (P < .05 vs Vwf(+/-)). Finally, we demonstrate that platelet-derived TSP1 modulates arterial thrombosis in vivo. We conclude that TSP1 released from platelets plays a mechanistic role in modulating thrombosis in the presence of VWF.


Asunto(s)
Trombosis/metabolismo , Trombospondina 1/metabolismo , Factor de von Willebrand/metabolismo , Animales , Arteriolas/patología , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Agregación Plaquetaria/fisiología
19.
FASEB J ; 28(4): 1819-29, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24421399

RESUMEN

Platelets contribute to 95% of circulating amyloid precursor protein in the body and have widely been employed as a "peripheral" model of neurons in Alzheimer's disease. We sought to analyze the effects of amyloid ß (Aß) on platelets and to understand the underlying molecular mechanism. The Aß active fragment containing amino acid sequence 25-35 (Aß(25-35); 10-20 µM) was found to induce strong aggregation of human platelets, granule release, and integrin activation, similar to that elicited by physiological agonists. Platelets exposed to Aß(25-35) retracted fibrin clot and displayed augmented adhesion to collagen under arterial shear, reflective of a switch to prothrombotic phenotype. Exposure of platelets to Aß peptide (20 µM) resulted in a 4.2- and 2.3-fold increase in phosphorylation of myosin light chain (MLC) and MLC phosphatase, respectively, which was reversed by Y27632, an inhibitor of Rho-associated coiled-coil protein kinase (ROCK). Aß(25-35)-induced platelet aggregation and clot retraction were also significantly attenuated by Y27632. Consistent with these findings, Aß(25-35) elicited a significant rise in the level of RhoA-GTP in platelets. Platelets pretreated with reverse-sequenced Aß fragment (Aß(35-25)) and untreated resting platelets served as controls. We conclude that Aß induces cellular activation through RhoA-dependent modulation of actomyosin, and hence, RhoA could be a potential therapeutic target in Alzheimer's disease and cerebral amyloid angiopathy.


Asunto(s)
Actomiosina/metabolismo , Péptidos beta-Amiloides/farmacología , Plaquetas/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo , Adulto , Enfermedad de Alzheimer/metabolismo , Amidas/farmacología , Secuencia de Aminoácidos , Péptidos beta-Amiloides/toxicidad , Animales , Plaquetas/metabolismo , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Femenino , Humanos , Immunoblotting , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Fragmentos de Péptidos/farmacología , Fragmentos de Péptidos/toxicidad , Agregación Plaquetaria/efectos de los fármacos , Embolia Pulmonar/inducido químicamente , Piridinas/farmacología , Adulto Joven , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo
20.
J Biol Chem ; 288(10): 6826-34, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23329846

RESUMEN

Limit of platelet life span (8-10 days) is determined by the activity of a putative "internal clock" composed of Bcl-2 family proteins, whereas the role of other molecular players in this process remains obscure. Here, we sought to establish a central role of proteasome in platelet life span regulation. Administration of mice with inhibitors of proteasome peptidase activity induced significant thrombocytopenia. This was associated with enhanced clearance of biotin-labeled platelets from circulation and reduction in average platelet half-life from 66 to 37 h. Cells pretreated in vitro with proteasome inhibitors exhibited augmented annexin V binding and a drop in mitochondrial transmembrane potential indicative of apoptotic cell death and decreased platelet life span. These cells were preferentially phagocytosed by monocyte-derived macrophages, thus linking proteasome activity with platelet survival. The decisive role of proteasome in this process was underscored from enhanced expression of conformationally active Bax in platelets with attenuated proteasome activity, which was consistent with pro-apoptotic phenotype of these cells. The present study establishes a critical role of proteasome in delimiting platelet life span ostensibly through constitutive elimination of the conformationally active Bax. These findings bear potential implications in clinical settings where proteasome peptidase activities are therapeutically targeted.


Asunto(s)
Plaquetas/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Proteína X Asociada a bcl-2/metabolismo , Animales , Apoptosis/efectos de los fármacos , Plaquetas/enzimología , Plaquetas/metabolismo , Western Blotting , Ácidos Borónicos/farmacología , Bortezomib , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Microscopía Fluorescente , Oligopéptidos/farmacología , Fagocitosis/efectos de los fármacos , Recuento de Plaquetas , Conformación Proteica , Pirazinas/farmacología , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Proteína X Asociada a bcl-2/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...