Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Blood ; 139(21): 3194-3203, 2022 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-35358299

RESUMEN

Platelets are critical in hemostasis and a major contributor to arterial thrombosis (AT). (Pre)clinical studies suggest platelets also contribute to venous thrombosis (VT), but the mechanisms are largely unknown. We hypothesized that in VT, platelets use signaling machinery distinct from AT. Here we aimed to characterize the contributions of platelet G protein-coupled (GPCR) and immunoreceptor tyrosine-based activation motif (ITAM) receptor signaling to VT. Wild-type (WT) and transgenic mice were treated with inhibitors to selectively inhibit platelet-signaling pathways: ITAM-CLEC2 (Clec2mKO), glycoprotein VI (JAQ1 antibody), and Bruton's tyrosine kinase (ibrutinib); GPCR-cyclooxygenase 1 (aspirin); and P2Y12 (clopidogrel). VT was induced by inferior vena cava stenosis. Thrombin generation in platelet-rich plasma and whole-blood clot formation were studied ex vivo. Intravital microscopy was used to study platelet-leukocyte interactions after flow restriction. Thrombus weights were reduced in WT mice treated with high-dose aspirin + clopidogrel (dual antiplatelet therapy [DAPT]) but not in mice treated with either inhibitor alone or low-dose DAPT. Similarly, thrombus weights were reduced in mice with impaired ITAM signaling (Clec2mKO + JAQ1; WT + ibrutinib) but not in Clec2mKO or WT + JAQ1 mice. Both aspirin and clopidogrel, but not ibrutinib, protected mice from FeCl3-induced AT. Thrombin generation and clot formation were normal in blood from high-dose DAPT- or ibrutinib-treated mice; however, platelet adhesion and platelet-neutrophil aggregate formation at the vein wall were reduced in mice treated with high-dose DAPT or ibrutinib. In summary, VT initiation requires platelet activation via GPCRs and ITAM receptors. Strong inhibition of either signaling pathway reduces VT in mice.


Asunto(s)
Trombosis , Trombosis de la Vena , Animales , Aspirina , Plaquetas/metabolismo , Clopidogrel/metabolismo , Clopidogrel/farmacología , Proteínas de Unión al GTP , Motivo de Activación del Inmunorreceptor Basado en Tirosina , Ratones , Ratones Transgénicos , Activación Plaquetaria , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Trombina/metabolismo , Trombosis/metabolismo , Trombosis de la Vena/metabolismo
2.
Haematologica ; 105(1): 218-225, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31048354

RESUMEN

Pancreatic cancer is associated with a high incidence of venous thromboembolism. Neutrophils have been shown to contribute to thrombosis in part by releasing neutrophil extracellular traps (NET). A recent study showed that increased plasma levels of the NET biomarker, citrullinated histone H3 (H3Cit), are associated with venous thromboembolism in patients with pancreatic and lung cancer but not in those with other types of cancer, including breast cancer. In this study, we examined the contribution of neutrophils and NET to venous thrombosis in nude mice bearing human pancreatic tumors. We found that tumor-bearing mice had increased circulating neutrophil counts and levels of granulocyte-colony stimulating factor, neutrophil elastase, H3Cit and cell-free DNA compared with controls. In addition, thrombi from tumor-bearing mice contained increased levels of the neutrophil marker Ly6G, as well as higher levels of H3Cit and cell-free DNA. Thrombi from tumor-bearing mice also had denser fibrin with thinner fibers consistent with increased thrombin generation. Importantly, either neutrophil depletion or administration of DNase I reduced the thrombus size in tumor-bearing but not in control mice. Our results, together with clinical data, suggest that neutrophils and NET contribute to venous thrombosis in patients with pancreatic cancer.


Asunto(s)
Trampas Extracelulares , Neoplasias Pancreáticas , Trombosis de la Vena , Animales , Humanos , Ratones , Ratones Desnudos , Neutrófilos , Trombosis de la Vena/etiología
3.
J Surg Res ; 254: 165-169, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32445932

RESUMEN

BACKGROUND: Vein graft stenosis is a major complication of coronary artery bypass surgery and peripheral arterial bypass procedures. Experimental models of this clinical complication have used in vivo grafting procedures, relying on the relatively modest neointimal thickening in these models as a surrogate for clinical graft stenosis without regard to the donor site origin of the vein graft. MATERIALS AND METHODS: In a standard rat model of vein grafting, three different donor sites were used to supply veins used as interpositional grafts to the femoral artery: the superficial inferior epigastric vein, the common femoral vein, and the posterior facial vein (distal branch of the jugular vein). Grafts were harvested as 4 wk and histomorphometrically evaluated for the extent of neointimal formation and lumen narrowing. RESULTS: The posterior facial vein showed significantly thicker neointima and a greater extent of lumen narrowing than the other two graft sources, despite having a similar diameter to the femoral vein and nearly twice the initial diameter of the epigastric vein. CONCLUSIONS: The source of donor graft material can greatly influence the extent of neointimal response after interpositional vein grafting to arterial flow. These findings support use of the posterior facial vein graft over other more standard donor vein grafts in research directed at understanding the causes and prevention of vein graft stenosis.


Asunto(s)
Neointima/etiología , Injerto Vascular , Venas/trasplante , Animales , Femenino , Ratas Endogámicas Lew
4.
Blood ; 129(21): 2917-2927, 2017 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-28336528

RESUMEN

Atherothrombosis is a process mediated by dysregulated platelet activation that can cause life-threatening complications and is the leading cause of death by cardiovascular disease. Platelet reactivity in hyperlipidemic conditions is enhanced when platelet scavenger receptor CD36 recognizes oxidized lipids in oxidized low-density lipoprotein (oxLDL) particles, a process that induces an overt prothrombotic phenotype. The mechanisms by which CD36 promotes platelet activation and thrombosis remain incompletely defined. In this study, we identify a mechanism for CD36 to promote thrombosis by increasing activation of MAPK extracellular signal-regulated kinase 5 (ERK5), a protein kinase known to be exquisitely sensitive to redox stress, through a signaling pathway requiring Src kinases, NADPH oxidase, superoxide radical anion, and hydrogen peroxide. Pharmacologic inhibitors of ERK5 blunted platelet activation and aggregation in response to oxLDL and targeted genetic deletion of ERK5 in murine platelets prevented oxLDL-induced platelet deposition on immobilized collagen in response to arterial shear. Importantly, in vivo thrombosis experiments after bone marrow transplantation from platelet-specific ERK5 null mice into hyperlipidemic apolipoprotein E null mice showed decreased platelet accumulation and increased thrombosis times compared with mice transplanted with ERK5 expressing control bone marrows. These findings suggest that atherogenic conditions critically regulate platelet CD36 signaling by increasing superoxide radical anion and hydrogen peroxide through a mechanism that promotes activation of MAPK ERK5.


Asunto(s)
Plaquetas/inmunología , Antígenos CD36/inmunología , Hiperlipidemias/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteína Quinasa 7 Activada por Mitógenos/inmunología , Activación Plaquetaria/inmunología , Trombosis/inmunología , Aloinjertos , Animales , Apolipoproteínas E/genética , Apolipoproteínas E/inmunología , Plaquetas/patología , Trasplante de Médula Ósea , Antígenos CD36/genética , Humanos , Hiperlipidemias/genética , Hiperlipidemias/patología , Lipoproteínas LDL/genética , Lipoproteínas LDL/inmunología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Mutantes , Proteína Quinasa 7 Activada por Mitógenos/genética , NADPH Oxidasas/genética , NADPH Oxidasas/inmunología , Activación Plaquetaria/genética , Trombosis/genética , Trombosis/patología
5.
Blood ; 129(18): 2537-2546, 2017 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-28251913

RESUMEN

Red blood cells (RBCs) demonstrate procoagulant properties in vitro, and elevated hematocrit is associated with reduced bleeding and increased thrombosis risk in humans. These observations suggest RBCs contribute to thrombus formation. However, effects of RBCs on thrombosis are difficult to assess because humans and mice with elevated hematocrit typically have coexisting pathologies. Using an experimental model of elevated hematocrit in healthy mice, we measured effects of hematocrit in 2 in vivo clot formation models. We also assessed thrombin generation, platelet-thrombus interactions, and platelet accumulation in thrombi ex vivo, in vitro, and in silico. Compared with controls, mice with elevated hematocrit (RBCHIGH) formed thrombi at a faster rate and had a shortened vessel occlusion time. Thrombi in control and RBCHIGH mice did not differ in size or fibrin content, and there was no difference in levels of circulating thrombin-antithrombin complexes. In vitro, increasing the hematocrit increased thrombin generation in the absence of platelets; however, this effect was reduced in the presence of platelets. In silico, direct numerical simulations of whole blood predicted elevated hematocrit increases the frequency and duration of interactions between platelets and a thrombus. When human whole blood was perfused over collagen at arterial shear rates, elevating the hematocrit increased the rate of platelet deposition and thrombus growth. These data suggest RBCs promote arterial thrombosis by enhancing platelet accumulation at the site of vessel injury. Maintaining a normal hematocrit may reduce arterial thrombosis risk in humans.


Asunto(s)
Antitrombina III/metabolismo , Arterias , Coagulación Sanguínea , Péptido Hidrolasas/metabolismo , Trombosis/metabolismo , Lesiones del Sistema Vascular/metabolismo , Animales , Arterias/lesiones , Arterias/metabolismo , Plaquetas , Femenino , Hematócrito , Humanos , Masculino , Ratones , Resistencia al Corte
6.
Arterioscler Thromb Vasc Biol ; 38(4): 816-828, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29419409

RESUMEN

OBJECTIVE: PS (protein S) is a plasma protein that directly inhibits the coagulation FIXa (factor IXa) in vitro. Because elevated FIXa is associated with increased risk of venous thromboembolism, it is important to establish how PS inhibits FIXa function in vivo. The goal of this study is to confirm direct binding of PS with FIXa in vivo, identify FIXa amino acid residues required for binding PS in vivo, and use an enzymatically active FIXa mutant that is unable to bind PS to measure the significance of PS-FIXa interaction in hemostasis. APPROACH AND RESULTS: We demonstrate that PS inhibits FIXa in vivo by associating with the FIXa heparin-binding exosite. We used fluorescence tagging, immunohistochemistry, and protein-protein crosslinking to show in vivo interaction between FIXa and PS. Importantly, platelet colocalization required a direct interaction between the 2 proteins. FIXa and PS also coimmunoprecipitated from plasma, substantiating their interaction in a physiological milieu. PS binding to FIXa and PS inhibition of the intrinsic Xase complex required residues K132, K126, and R170 in the FIXa heparin-binding exosite. A double mutant, K132A/R170A, retained full activity but could not bind to PS. Crucially, Hemophilia B mice infused with FIXa K132A/R170A displayed an accelerated rate of fibrin clot formation compared with wild-type FIXa. CONCLUSIONS: Our findings establish PS as an important in vivo inhibitor of FIXa. Disruption of the interaction between PS and FIXa causes an increased rate of thrombus formation in mice. This newly discovered function of PS implies an unexploited target for antithrombotic therapeutics.


Asunto(s)
Plaquetas/metabolismo , Factor IXa/metabolismo , Hemofilia B/sangre , Hemostasis , Heparina/metabolismo , Proteína S/metabolismo , Trombosis de la Vena/prevención & control , Animales , Sitios de Unión , Unión Competitiva , Coagulantes/administración & dosificación , Modelos Animales de Enfermedad , Factor IX/genética , Factor IX/metabolismo , Factor IXa/administración & dosificación , Factor IXa/genética , Hemofilia B/tratamiento farmacológico , Hemofilia B/genética , Hemostasis/efectos de los fármacos , Humanos , Infusiones Intravenosas , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Trombosis de la Vena/sangre , Trombosis de la Vena/genética
7.
Circ Res ; 118(9): 1363-79, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-27126647

RESUMEN

Thrombosis is a leading cause of morbidity and mortality worldwide. Animal models are used to understand the pathological pathways involved in thrombosis and to test the efficacy and safety of new antithrombotic drugs. In this review, we will first describe the central role a variety of animal models of thrombosis and hemostasis has played in the development of new antiplatelet and anticoagulant drugs. These include the widely used P2Y12 antagonists and the recently developed orally available anticoagulants that directly target factor Xa or thrombin. Next, we will describe the new players, such as polyphosphate, neutrophil extracellular traps, and microparticles, which have been shown to contribute to thrombosis in mouse models, particularly venous thrombosis models. Other mouse studies have demonstrated roles for the factor XIIa and factor XIa in thrombosis. This has spurred the development of strategies to reduce their levels or activities as a new approach for preventing thrombosis. Finally, we will discuss the emergence of zebrafish as a model to study thrombosis and its potential use in the discovery of novel factors involved in thrombosis and hemostasis. Animal models of thrombosis from zebrafish to nonhuman primates are vital in identifying pathological pathways of thrombosis that can be safely targeted with a minimal effect on hemostasis. Future studies should focus on understanding the different triggers of thrombosis and the best drugs to prevent each type of thrombotic event.


Asunto(s)
Anticoagulantes/uso terapéutico , Fibrinolíticos/uso terapéutico , Inhibidores de Agregación Plaquetaria/uso terapéutico , Trombosis/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Ratones , Primates , Trombosis/genética , Trombosis/metabolismo , Trombosis/patología , Pez Cebra
8.
Blood ; 125(26): 4078-84, 2015 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-25954015

RESUMEN

Tissue factor pathway inhibitor (TFPI) is a critical anticoagulant protein present in endothelium and platelets. Mice lacking TFPI (Tfpi(-/-)) die in utero from disseminated intravascular coagulation. They are rescued by concomitant tissue factor (TF) deficiency, demonstrating that TFPI modulates TF function in vivo. Recent studies have found TFPI inhibits prothrombinase activity during the initiation of coagulation and limits platelet accumulation during thrombus formation, implicating TFPI in modulating platelet procoagulant activity. To examine whether altered platelet function would compensate for the lack of TFPI and rescue TFPI-null embryonic lethality, Tfpi(+/-) mice lacking the platelet thrombin receptor, protease activated receptor 4 (PAR4; Par4(-/-)), or its coreceptor, PAR3, were mated. PAR3 deficiency did not rescue Tfpi(-/-) embryos, but >40% of expected Tfpi(-/-):Par4(-/-) offspring survived to adulthood. Adult Tfpi(-/-):Par4(-/-) mice did not exhibit overt thrombosis. However, they had focal sterile inflammation with fibrin(ogen) deposition in the liver and elevated plasma thrombin-antithrombin complexes, indicating activation of coagulation at baseline. Tfpi(-/-):Par4(-/-) mice have platelet and fibrin accumulation similar to Par4(-/-) mice following venous electrolytic injury but were more susceptible than Par4(-/-) mice to TF-induced pulmonary embolism. In addition, ∼30% of the Tfpi(-/-):Par4(-/-) mice were born with short tails. Tfpi(-/-):Par4(-/-) mice are the first adult mice described that lack TFPI with unaltered TF. They demonstrate that TFPI physiologically modulates thrombin-dependent platelet activation in a manner that is required for successful embryonic development and identify a role for TFPI in dampening intravascular procoagulant stimuli that lead to thrombin generation, even in the absence of thrombin-mediated platelet activation.


Asunto(s)
Desarrollo Embrionario/fisiología , Lipoproteínas/metabolismo , Ratones/embriología , Activación Plaquetaria/fisiología , Trombina/metabolismo , Animales , Ratones Noqueados
9.
Arterioscler Thromb Vasc Biol ; 36(9): 1838-46, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27417588

RESUMEN

OBJECTIVE: The tight regulation of platelet adhesiveness, mediated by the αIIbß3 integrin, is critical for hemostasis and prevention of thrombosis. We recently demonstrated that integrin affinity in platelets is controlled by the guanine nucleotide exchange factor, CalDAG-GEFI (CD-GEFI), and its target, RAP1. In this study, we investigated whether low-level expression of CD-GEFI leads to protection from thrombosis without pathological bleeding in mice. APPROACH AND RESULTS: Cdg1(low) mice were generated by knockin of human CD-GEFI cDNA into the mouse Cdg1 locus. CD-GEFI expression in platelets from Cdg1(low) mice was reduced by ≈90% when compared with controls. Activation of RAP1 and αIIbß3 was abolished at low agonist concentrations and partially inhibited at high agonist concentrations in Cdg1(low) platelets. Consistently, the aggregation response of Cdg1(low) platelets was weaker than that of wild-type platelets, but more efficient than that observed in Cdg1(-/-) platelets. Importantly, Cdg1(low) mice were strongly protected from arterial and immune complex-mediated thrombosis, with only minimal impact on primary hemostasis. CONCLUSIONS: Together, our studies suggest the partial inhibition of CD-GEFI function as a powerful new approach to safely prevent thrombotic complications.


Asunto(s)
Plaquetas/metabolismo , Factores de Intercambio de Guanina Nucleótido/deficiencia , Hemostasis , Activación Plaquetaria , Trombosis/prevención & control , Animales , Modelos Animales de Enfermedad , Genotipo , Factores de Intercambio de Guanina Nucleótido/sangre , Factores de Intercambio de Guanina Nucleótido/genética , Humanos , Ratones Transgénicos , Mutación , Fenotipo , Agregación Plaquetaria , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Transducción de Señal , Trombosis/sangre , Trombosis/genética , Factores de Tiempo , Proteínas de Unión al GTP rap1/sangre
10.
Blood ; 121(10): 1858-67, 2013 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-23264598

RESUMEN

The integrin family is composed of a series of 24 αß heterodimer transmembrane adhesion receptors that mediate cell-cell and cell-extracellular matrix interactions. Adaptor molecules bearing immunoreceptor tyrosine-based activation motifs (ITAMs) have recently been shown to cooperate with specific integrins to increase the efficiency of transmitting ligand-binding-induced signals into cells. In human platelets, Fc receptor γ-chain IIa (FcγRIIa) has been identified as an ITAM-bearing transmembrane receptor responsible for mediating "outside-in" signaling through αIIbß3, the major adhesion receptor on the platelet surface. To explore the importance of FcγRIIa in thrombosis and hemostasis, we subjected FcγRIIa-negative and FcγRIIa-positive murine platelets to a number of well-accepted models of platelet function. Compared with their FcγRIIa-negative counterparts, FcγRIIa-positive platelets exhibited increased tyrosine phosphorylation of Syk and phospholipase Cγ2 and increased spreading upon interaction with immobilized fibrinogen, retracted a fibrin clot faster, and showed markedly enhanced thrombus formation when perfused over a collagen-coated flow chamber under conditions of arterial and venous shear. They also displayed increased thrombus formation and fibrin deposition in in vivo models of vascular injury. Taken together, these data establish FcγRIIa as a physiologically important functional conduit for αIIbß3-mediated outside-in signaling, and suggest that modulating the activity of this novel integrin/ITAM pair might be effective in controlling thrombosis.


Asunto(s)
Plaquetas/metabolismo , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Receptores de IgG/fisiología , Receptores Inmunológicos/metabolismo , Transducción de Señal , Trombosis/etiología , Tirosina/metabolismo , Animales , Arteriolas/metabolismo , Arteriolas/patología , Fibrina/metabolismo , Fibrinógeno/metabolismo , Hemostasis/fisiología , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Agregación Plaquetaria , Recuento de Plaquetas , Trombosis/metabolismo , Trombosis/patología , Venas/metabolismo , Venas/patología
11.
Proc Natl Acad Sci U S A ; 109(10): 3927-31, 2012 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-22355108

RESUMEN

Tissue factor pathway inhibitor (TFPI) blocks thrombin generation via the extrinsic blood coagulation pathway. Because the severe bleeding in patients with hemophilia occurs from deficiency of intrinsic blood coagulation pathway factor VIII or IX, pharmacological agents that inactivate TFPI and, therefore, restore thrombin generation via the extrinsic pathway, are being developed for treatment of hemophilia. Murine models of combined TFPI and factor VIII deficiency were used to examine the impact of TFPI deficiency on bleeding and clotting in hemophilia. In breeding studies, Factor VIII null (F8(-/-)) did not rescue the embryonic death of TFPI null (Tfpi(-/-)) mice. Tfpi(+/-) did not alter the bleeding phenotype of F8(-/-) mice. However, total inhibition of intravascular TFPI through injection of anti-TFPI antibody mitigated tail vein bleeding. Interestingly, tail blood loss progressively decreased at doses greater than needed to totally inhibit plasma TFPI, suggesting that inhibition of a sequestered pool of TFPI released at the injury site mitigates bleeding. Because TFPI is sequestered within platelets and released following their activation, the function of platelet TFPI was examined in F8(-/-) mice lacking hematopoietic cell TFPI that was generated by fetal liver transplantation. Blood loss after tail transection significantly decreased in Tfpi(+/-);F8(-/-) mice with hematopoietic Tfpi(-/-) cells compared with Tfpi(+/-);F8(-/-) mice with Tfpi(+/+) hematopoietic cells. Additionally, following femoral vein injury, Tfpi(+/-);F8(-/-) mice with Tfpi(-/-) hematopoietic cells had increased fibrin deposition compared with identical-genotype mice with Tfpi(+/+) hematopoietic cells. These findings implicate platelet TFPI as a primary physiological regulator of bleeding in hemophilia.


Asunto(s)
Hemofilia A/metabolismo , Lipoproteínas/genética , Lipoproteínas/metabolismo , Animales , Coagulación Sanguínea , Modelos Animales de Enfermedad , Femenino , Prueba de Complementación Genética , Genotipo , Células Madre Hematopoyéticas/citología , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Hemorragia , Hígado/embriología , Trasplante de Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo
12.
Blood ; 120(26): 5103-10, 2012 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22968458

RESUMEN

Inorganic polyphosphates are linear polymers of orthophosphate that modulate blood clotting and inflammation. Polyphosphate accumulates in infectious microorganisms and is secreted by activated platelets; long-chain polyphosphate in particular is an extremely potent initiator of the contact pathway, a limb of the clotting cascade important for thrombosis but dispensable for hemostasis. Polyphosphate inhibitors therefore might act as novel antithrombotic/anti-inflammatory agents with reduced bleeding side effects. Antipolyphosphate antibodies are unlikely because of polyphosphate's ubiquity and simple structure; and although phosphatases such as alkaline phosphatase can digest polyphosphate, they take time and may degrade other biologically active molecules. We now identify a panel of polyphosphate inhibitors, including cationic proteins, polymers, and small molecules, and report their effectiveness in vitro and in vivo. We also compare their effectiveness against the procoagulant activity of RNA. Polyphosphate inhibitors were antithrombotic in mouse models of venous and arterial thrombosis and blocked the inflammatory effect of polyphosphate injected intradermally in mice. This study provides proof of principle for polyphosphate inhibitors as antithrombotic/anti-inflammatory agents in vitro and in vivo, with a novel mode of action compared with conventional anticoagulants.


Asunto(s)
Antiinflamatorios/farmacología , Fibrinolíticos/farmacología , Inflamación/tratamiento farmacológico , Polifosfatos/antagonistas & inhibidores , Trombosis/tratamiento farmacológico , Animales , Antiinflamatorios/aislamiento & purificación , Coagulación Sanguínea/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos , Fibrinolíticos/aislamiento & purificación , Hemostasis/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Humanos , Inflamación/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Polifosfatos/sangre , Trombosis/sangre
13.
Arterioscler Thromb Vasc Biol ; 33(8): 1829-36, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23723374

RESUMEN

OBJECTIVE: Individuals with elevated prothrombin, including those with the prothrombin G20210A mutation, have increased risk of venous thrombosis. Although these individuals do not have increased circulating prothrombotic biomarkers, their plasma demonstrates increased tissue factor-dependent thrombin generation in vitro. The objectives of this study were to determine the pathological role of elevated prothrombin in venous and arterial thrombosis in vivo, and distinguish thrombogenic mechanisms in these vessels. APPROACH AND RESULTS: Prothrombin was infused into mice to raise circulating levels. Venous thrombosis was induced by electrolytic stimulus to the femoral vein or inferior vena cava ligation. Arterial thrombosis was induced by electrolytic stimulus or ferric chloride application to the carotid artery. Mice infused with prothrombin demonstrated increased tissue factor-triggered thrombin generation measured ex vivo, but did not have increased circulating prothrombotic biomarkers in the absence of vessel injury. After venous injury, elevated prothrombin increased thrombin generation and the fibrin accumulation rate and total amount of fibrin ≈ 3-fold, producing extended thrombi with increased mass. However, elevated prothrombin did not accelerate platelet accumulation, increase the fibrin accumulation rate, or shorten the vessel occlusion time after arterial injury. CONCLUSIONS: These findings reconcile previously discordant findings on thrombin generation in hyperprothrombinemic individuals measured ex vivo and in vitro, and show elevated prothrombin promotes venous, but not arterial, thrombosis in vivo.


Asunto(s)
Coagulación Sanguínea/fisiología , Protrombina/metabolismo , Trombofilia/metabolismo , Trombosis de la Vena/metabolismo , Animales , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/fisiología , Arterias Carótidas/fisiología , Cloruros/farmacología , Modelos Animales de Enfermedad , Vena Femoral/fisiología , Compuestos Férricos/farmacología , Fibrina/metabolismo , Humanos , Ratones , Noxas/farmacología , Protrombina/farmacología , Factores de Riesgo , Trombofilia/inducido químicamente , Trombofilia/epidemiología , Vena Cava Inferior/fisiología , Trombosis de la Vena/inducido químicamente , Trombosis de la Vena/epidemiología
14.
Eur Heart J ; 34(22): 1636-43, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23530023

RESUMEN

AIMS: The long-term failure of autologous saphenous vein bypass grafts due to neointimal thickening is a major clinical burden. Identifying novel strategies to prevent neointimal thickening is important. Thus, this study aimed to identify microRNAs (miRNAs) that are dysregulated during neointimal formation and determine their pathophysiological relevance following miRNA manipulation. METHODS AND RESULTS: We undertook a microarray approach to identify dysregulated miRNAs following engraftment in an interpositional porcine graft model. These profiling experiments identified a number of miRNAs which were dysregulated following engraftment. miR-21 levels were substantially elevated following engraftment and these results were confirmed by quantitative real-time PCR in mouse, pig, and human models of vein graft neointimal formation. Genetic ablation of miR-21 in mice or grafted veins dramatically reduced neointimal formation in a mouse model of vein grafting. Furthermore, pharmacological knockdown of miR-21 in human veins resulted in target gene de-repression and a significant reduction in neointimal formation. CONCLUSION: This is the first report demonstrating that miR-21 plays a pathological role in vein graft failure. Furthermore, we also provided evidence that knockdown of miR-21 has therapeutic potential for the prevention of pathological vein graft remodelling.


Asunto(s)
MicroARNs/genética , Neointima/genética , Vena Safena/metabolismo , Injerto Vascular , Animales , Arteria Carótida Común/metabolismo , Células Cultivadas , Técnicas de Silenciamiento del Gen , Rechazo de Injerto/genética , Rechazo de Injerto/metabolismo , Humanos , Ratones , Ratones Noqueados , MicroARNs/metabolismo , Análisis por Micromatrices , Vena Safena/trasplante , Porcinos , Venas Cavas/metabolismo
15.
Microsurgery ; 34(8): 653-6, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24848809

RESUMEN

Microvascular training models for vein grafting most often use the rat epigastric vein interpositioned to the femoral artery. We describe the rat posterior facial vein as an alternative vein graft model; it has at least a 2:1 diametric ratio to the femoral artery and a tougher connective tissue, making it more similar to clinical vein grafting for reconstructive microsurgery. A series of 24 grafts interpositioned to the femoral artery were done using 11-12 sutures per end-to-end anastomosis and yielded early patency rates of 96% at 20 min and 92% at 2 and 4 weeks for subsets of 12 grafts. As a training model the diametric disparity provides unique challenges with clinical relevance, for which a number of different techniques for matching arterial to venous circumferences can be done.


Asunto(s)
Cara/irrigación sanguínea , Arteria Femoral/cirugía , Microcirugia/educación , Injerto Vascular/educación , Venas/trasplante , Anastomosis Quirúrgica/educación , Animales , Modelos Anatómicos , Modelos Animales , Ratas , Ratas Endogámicas Lew
16.
Blood Adv ; 8(2): 441-452, 2024 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-37773781

RESUMEN

ABSTRACT: Hemophilia B (HB) is caused by an inherited deficiency of plasma coagulation factor IX (FIX). Approximately 60% of pediatric patients with HB possess a severe form of FIX deficiency (<1% FIX activity). Treatment typically requires replacement therapy through the administration of FIX. However, exogenous FIX has a limited functional half-life, and the natural anticoagulant protein S (PS) inhibits activated FIX (FIXa). PS ultimately limits thrombin formation, which limits plasma coagulation. This regulation of FIXa activity by PS led us to test whether inhibiting PS would extend the functional half-life of FIX and thereby prolong FIX-based HB therapy. We assayed clotting times and thrombin generation to measure the efficacy of a PS antibody for increasing FIX activity in commercially obtained plasma and plasma from pediatric patients with HB. We included 11 pediatric patients who lacked additional comorbidities and coagulopathies. In vivo, we assessed thrombus formation in HB mice in the presence of the FIXa ± PS antibody. We found an accelerated rate of clotting in the presence of PS antibody. Similarly, the peak thrombin formed was significantly greater in the presence of the PS antibody, even in plasma from patients with severe HB. Furthermore, HB mice injected with PS antibody and FIX had a 4.5-fold higher accumulation of fibrin at the thrombus induction site compared with mice injected with FIX alone. Our findings imply that a PS antibody would be a valuable adjunct to increase the effectiveness of FIX replacement therapy in pediatric patients who have mild, moderate, and severe HB.


Asunto(s)
Hemofilia B , Trombosis , Humanos , Ratones , Niño , Animales , Hemofilia B/tratamiento farmacológico , Trombina/metabolismo , Factor IX/uso terapéutico , Factor IX/metabolismo , Factor IXa/metabolismo , Anticuerpos
17.
J Exp Med ; 204(10): 2439-48, 2007 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-17893198

RESUMEN

Activated protein C (APC) reduces mortality of severe sepsis patients but increases the risk of serious bleeding. APC exerts anticoagulant activity by proteolysis of factors Va/VIIIa. APC also exerts antiinflammatory and antiapoptotic effects and stabilizes endothelial barrier function by APC-initiated cell signaling that requires two receptors, endothelial cell protein C receptor (EPCR) and protease-activated receptor 1 (PAR1). The relative importance of APC's various activities for efficacy in sepsis is unknown. We used protein engineering of mouse APC and genetically altered mice to clarify mechanisms for the efficacy of APC in mouse sepsis models. Mortality reduction in LPS-induced endotoxemia required the enzymatic active site of APC, EPCR, and PAR-1, highlighting a key role for APC's cytoprotective actions. A recombinant APC variant with normal signaling but <10% anticoagulant activity (5A-APC) was as effective as wild-type APC in reducing mortality after LPS challenge, and enhanced the survival of mice subjected to peritonitis induced by gram-positive or -negative bacteria or to polymicrobial peritoneal sepsis triggered by colon ascendens stent implantation. Thus, APC's efficacy in severe sepsis is predominantly based on EPCR- and PAR1-dependent cell signaling, and APC variants with normal cell signaling but reduced anticoagulant activities retain efficacy while reducing the risk of bleeding.


Asunto(s)
Endotoxemia/metabolismo , Endotoxemia/patología , Proteína C/metabolismo , Ingeniería de Proteínas , Sepsis/metabolismo , Sepsis/patología , Animales , Apoptosis , Permeabilidad de la Membrana Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/enzimología , Endotoxemia/tratamiento farmacológico , Activación Enzimática/efectos de los fármacos , Lipopolisacáridos/farmacología , Linfocitos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína C/genética , Sepsis/tratamiento farmacológico , Transducción de Señal , Tasa de Supervivencia
18.
J Hand Surg Am ; 38(9): 1784-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23891176

RESUMEN

PURPOSE: To evaluate the hypothesis that platelets and fibrin differentially accrue at microvascular anastomoses in arteries versus veins and under different pharmacologic conditions. METHODS: We evaluated mouse arterial and venous anastomoses with intravital fluorescence imaging, using fluorophore-labeled platelets and anti-fibrin antibodies to measure the extent of thrombus component development in the intraluminal anastomotic site. We evaluated systemic heparin or eptifibatide (platelet aggregation inhibitor) to determine their relative influences on thrombus composition. RESULTS: Platelets accumulated rapidly in both arterial and venous repairs, and then fell in number after 10 to 30 minutes of reflow. Fibrin had a relatively steady development over 60 minutes in veins, with a more variable increase in arteries. Heparin reduced platelet accumulation in arteries and fibrin development in veins. Eptifibatide reduced platelets in both arteries and veins and had an apparent effect on lowering the amount of fibrin in veins. CONCLUSIONS: These findings show that platelets have a rapid, transient response, whereas fibrin has a slower, more sustained accrual in both arterial and venous anastomoses. Furthermore, inhibition of either coagulation or platelet aggregation can influence presumably non-targeted components of thrombosis in vascular repairs of both arteries and veins. CLINICAL RELEVANCE: Preventing replantation failure using antithrombotic therapies requires a better understanding of the effect of each pharmacologic compound on the various aspects of thrombogenesis.


Asunto(s)
Fibrina/efectos de los fármacos , Fibrinolíticos/farmacología , Péptidos/farmacología , Agregación Plaquetaria/efectos de los fármacos , Trombosis/fisiopatología , Anastomosis Quirúrgica , Animales , Eptifibatida , Fibrinolíticos/uso terapéutico , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirugia , Péptidos/uso terapéutico , Reimplantación , Trombosis/prevención & control , Grado de Desobstrucción Vascular/efectos de los fármacos
19.
Thromb Res ; 228: 163-171, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37331119

RESUMEN

INTRODUCTION: Arterial thrombosis is the main underlying mechanism of acute atherothrombosis. Combined antiplatelet and anticoagulant regimens prevent thrombosis but increase bleeding rates. Mast cell-derived heparin proteoglycans have local antithrombotic properties, and their semisynthetic dual AntiPlatelet and AntiCoagulant (APAC) mimetic may provide a new efficacious and safe tool for arterial thrombosis. We investigated the in vivo impact of intravenous APAC (0.3-0.5 mg/kg; doses chosen according to pharmacokinetic studies) in two mouse models of arterial thrombosis and the in vitro actions in mouse platelets and plasma. MATERIALS AND METHODS: Platelet function and coagulation were studied with light transmission aggregometry and clotting times. Carotid arterial thrombosis was induced either by photochemical injury or surgically exposing vascular collagen after infusion of APAC, UFH or vehicle. Time to occlusion, targeting of APAC to the vascular injury site and platelet deposition on these sites were assessed by intra-vital imaging. Tissue factor activity (TF) of the carotid artery and in plasma was captured. RESULTS: APAC inhibited platelet responsiveness to agonist stimulation (collagen and ADP) and prolonged APTT and thrombin time. After photochemical carotid injury, APAC-treatment prolonged times to occlusion in comparison with UFH or vehicle, and decreased TF both in carotid lysates and plasma. Upon binding from circulation to vascular collagen-exposing injury sites, APAC reduced the in situ platelet deposition. CONCLUSIONS: Intravenous APAC targets arterial injury sites to exert local dual antiplatelet and anticoagulant actions and attenuates thrombosis upon carotid injuries in mice. Systemic APAC provides local efficacy, highlighting APAC as a novel antithrombotic to reduce cardiovascular complications.


Asunto(s)
Trombosis de las Arterias Carótidas , Trombosis , Lesiones del Sistema Vascular , Animales , Ratones , Anticoagulantes/farmacología , Anticoagulantes/uso terapéutico , Anticoagulantes/química , Tromboplastina , Inhibidores de Agregación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Fibrinolíticos/uso terapéutico , Trombosis/etiología , Trombosis de las Arterias Carótidas/tratamiento farmacológico , Colágeno/farmacología , Agregación Plaquetaria
20.
Mol Ther Nucleic Acids ; 31: 440-451, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36817726

RESUMEN

Pathological blood clotting, or thrombosis, limits vital blood flow to organs; such deprivation can lead to catastrophic events including myocardial infarction, pulmonary embolism, and ischemic stroke. Prompt restoration of blood flow greatly improves outcomes. We explored whether aptamers could serve as molecular imaging probes to rapidly detect thrombi. An aptamer targeting thrombin, Tog25t, was found to rapidly localize to and visualize pre-existing clots in the femoral and jugular veins of mice using fluorescence imaging and, when circulating, was able to image clots as they form. Since free aptamer is quickly cleared from circulation, contrast is rapidly developed, allowing clot visualization within minutes. Moreover, administration of an antidote oligonucleotide further enhanced contrast development, causing the unbound aptamer to clear within 5min while impacting the clot-bound aptamer more slowly. These findings suggest that aptamers can serve as imaging agents for rapid detection of thrombi in acute care and perioperative settings.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA