Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
1.
J Thromb Haemost ; 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38729577

RESUMEN

BACKGROUND: Direct oral factor (F)Xa inhibitors are widely used as alternatives to conventional vitamin K antagonists in managing venous thromboembolism and nonvalvular atrial fibrillation. Unfortunately, bleeding-related adverse events remain a major concern in clinical practice. In case of bleeding or emergency surgery, rapid-onset reversal agents may be required to counteract the anticoagulant activity. OBJECTIVES: The ability of FXa variants to bypass the direct oral FXa inhibitors was assessed. METHODS: Human FXa variants were generated through substitution of phenylalanine 174 (F174) for either alanine, isoleucine, or serine. FXa variants were stably expressed in HEK293 cells and purified to homogeneity using ion-exchange chromatography. RESULTS: F174-substituted human FX variants demonstrated efficacy in restoring thrombin generation in plasma containing direct FXa inhibitors (apixaban, rivaroxaban, edoxaban). Their ability to bypass the anticoagulant effects stems from a significantly reduced sensitivity for the direct FXa inhibitors due to a decrease in binding affinity determined using molecular dynamics simulations and free energy computation. Furthermore, F174 modification resulted in a partial loss of inhibition by tissue factor pathway inhibitor, enhancing the procoagulant effect of F174-substituted FX. Consequently, the F174A- and F174S-substituted FX variants effectively counteracted the effects of 2 widely used anticoagulants, apixaban and rivaroxaban, in plasma of atrial fibrillation and venous thromboembolism patients. CONCLUSION: These human FX variants have the potential to serve as a rescue reversal strategy to overcome the effect of direct FXa inhibitors in case of life-threatening bleeding events or emergency surgical interventions.

2.
Pharmacotherapy ; 44(6): 416-424, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38686648

RESUMEN

BACKGROUND: Major bleeding occurs annually in 1%-3% of patients on vitamin K antagonists (VKAs), despite close monitoring. Genetic variants in proteins involved in VKA response may affect this risk. AIM: To determine the association of genetic variants (cytochrome P450 enzymes 2C9 [CYP2C9] and 4F2 [CYP4F2], gamma-glutamyl carboxylase [GGCX]) with major bleeding in VKA users, separately and combined, including vitamin K epoxide reductase complex subunit-1 (VKORC1). METHODS: A case-cohort study was established within the BLEEDS cohort, which includes 16,570 patients who initiated VKAs between 2012 and 2014. We selected all 326 major bleeding cases that occurred during 17,613 years of follow-up and a random subcohort of 978 patients. We determined variants in CYP2C9, CYP4F2, GGCX, VKORC1 and evaluated the interaction between variant genotypes. Hazard ratios for major bleeding with 95% confidence intervals (95% CI) were estimated by weighted Cox regression. RESULTS: Genotype was determined in 256 cases and 783 subcohort members. Phenprocoumon was the most prescribed VKA for both cases and the subcohort (78% and 75%, respectively). Patients with major bleeding were slightly older than subcohort patients. CYP4F2-TT carriership was associated with a 1.6-fold (95% CI 0.9-2.8) increased risk of major bleeding compared with CC-alleles, albeit not statistically significant. For the CYP2C9 and GGCX variants instead, the major bleeding risk was around unity. Carrying at least two variant genotypes in CYP2C9 (poor metabolizer), CYP4F2-TT, and VKORC1-AA was associated with a 4.0-fold (95%CI 1.4-11.4) increased risk, while carriers of both CYP4F2-TT and VKORC1-AA had a particularly increased major bleeding risk (hazard ratio 6.7, 95% CI 1.5-29.8) compared with carriers of CC alleles in CYP4F2 and GG in VKORC1. However, the number of major bleeding cases in carriers of multiple variants was few (8 and 5 patients, respectively). CONCLUSIONS: CYP4F2 polymorphism was associated with major bleeding, especially in combination with VKORC1 genetic variants. These variants could be considered to further personalize anticoagulant treatment.


Asunto(s)
Anticoagulantes , Hemorragia , Polimorfismo Genético , Vitamina K Epóxido Reductasas , Vitamina K , Humanos , Vitamina K/antagonistas & inhibidores , Hemorragia/inducido químicamente , Hemorragia/genética , Hemorragia/epidemiología , Femenino , Masculino , Anciano , Vitamina K Epóxido Reductasas/genética , Estudios de Cohortes , Anticoagulantes/efectos adversos , Persona de Mediana Edad , Citocromo P-450 CYP2C9/genética , Genotipo , Familia 4 del Citocromo P450/genética , Anciano de 80 o más Años , Ligasas de Carbono-Carbono/genética , Estudios de Casos y Controles
4.
Arterioscler Thromb Vasc Biol ; 41(8): 2263-2276, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34162230

RESUMEN

OBJECTIVE: The Australian snake venom ptFV (Pseudonaja textilis venom-derived factor V) variant retains cofactor function despite APC (activated protein C)-dependent proteolysis. Here, we aimed to unravel the mechanistic principles by determining the role of the absent Arg306 cleavage site that is required for the inactivation of FVa (mammalian factor Va). APPROACH AND RESULTS: Our findings show that in contrast to human FVa, APC-catalyzed proteolysis of ptFVa at Arg306 and Lys507 does not abrogate ptFVa cofactor function. Remarkably, the structural integrity of APC-proteolyzed ptFVa is maintained indicating that stable noncovalent interactions prevent A2-domain dissociation. Using Molecular Dynamics simulations, we uncovered key regions located in the A1 and A2 domain that may be at the basis of this remarkable characteristic. CONCLUSIONS: Taken together, we report a completely novel role for uniquely adapted regions in ptFVa that prevent A2 domain dissociation. As such, these results challenge our current understanding by which strict regulatory mechanisms control FVa activity.


Asunto(s)
Venenos Elapídicos/metabolismo , Factor Va/metabolismo , Proteína C/metabolismo , Animales , Línea Celular , Cricetinae , Venenos Elapídicos/química , Activación Enzimática , Factor Va/química , Factor Va/genética , Humanos , Enlace de Hidrógeno , Simulación de Dinámica Molecular , Dominios y Motivos de Interacción de Proteínas , Proteolisis , Relación Estructura-Actividad , Especificidad por Sustrato
5.
Thromb Res ; 198: 154-162, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33348189

RESUMEN

A wide variety of animal models on thrombosis and hemostasis are used in thrombosis and hemostasis research for the preclinical assessment of hemostatic agents. While the vertebrate coagulome is highly conserved, human and animal plasmas differ considerably when evaluated in coagulation assays such as prothrombin time (PT), activated partial thromboplastin time (APTT), and calibrated automated thrombography (CAT). Here, we have aimed to provide a reference framework for the evaluation of coagulation assays and inhibition of activated human FXa (hFXa) in various animal plasmas. To do so, a side-by-side evaluation of the extrinsic and intrinsic pathway of coagulation was performed by means of PT, APTT, and CAT measurements on (diluted) pooled plasmas from goats, pigs, rabbits, rats, mice, and humans. Plasma anti-FXa activity was assessed by determining the rate of recombinant hFXa inhibition through chromogenic activity analyses and immunoblotting. In general, rabbit, rat, and mouse plasmas exhibited robust clotting upon stimulation of both the extrinsic and intrinsic pathway, produced more thrombin during CAT upon plasma dilution, and displayed relatively high hFXa inhibitory activities. By comparison, goat, porcine, and human plasma displayed a similar profile in PT and APTT assays, produced less thrombin during CAT upon plasma dilution, and displayed comparable hFXa inhibitory activities. In conclusion, the observed differences in clotting parameters and anti-hFXa activity point to a higher anticoagulant threshold in plasma from rabbits, rats, and particularly in mice relative to human, goat, and porcine plasma. Finally, rat plasma was found to be more relevant to the preclinical assessment of human FX(a) in comparison to murine plasma.


Asunto(s)
Factor X , Factor Xa , Animales , Pruebas de Coagulación Sanguínea , Inhibidores del Factor Xa , Hemostasis , Humanos , Ratones , Modelos Animales , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina , Conejos , Ratas , Roedores , Porcinos
6.
Thromb Haemost ; 121(1): 70-75, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32854121

RESUMEN

BACKGROUND: Major bleeding occurs in 1 to 3% of patients treated with oral anticoagulants per year. Biomarkers may help to identify high-risk patients. A proposed marker for major bleeding while using anticoagulants is soluble thrombomodulin (sTM). METHODS: Plasma was available from 16,570 patients of the BLEEDS cohort that consisted of patients who started treatment with vitamin K antagonists between 2012 and 2014. A case-cohort study was performed including all patients with a major bleed (n = 326) during follow-up and a random sample of individuals selected at baseline (n = 652). Plasma sTM levels were measured and stratified by percentiles. Patients were also categorized by international normalized ratio (INR). Adjusted hazard ratios (for age, sex, hypertension, and diabetes) with 95% confidence intervals (CIs) were estimated by means of Cox regression. RESULTS: Plasma sTM levels were available for 263 patients with a major bleed and 538 control subjects. sTM levels were dose-dependently associated with risk of major bleeding, with a 1.9-fold increased risk (95% CI: 1.1-3.1) for levels above the 85th percentile versus the <25th percentile. A high INR (≥4) in the presence of high (≥70th percentile) sTM levels was associated with a 7.1-fold (95% CI: 4.1-12.3) increased risk of major bleeding, corresponding with a bleeding rate of 14.1 per 100 patient-years. CONCLUSION: High sTM levels at the start of treatment are associated with major bleeding during vitamin K antagonist treatment, particularly in the presence of a high INR.


Asunto(s)
Anticoagulantes/efectos adversos , Hemorragia/inducido químicamente , Trombomodulina/sangre , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Femenino , Hemorragia/sangre , Humanos , Relación Normalizada Internacional , Masculino , Persona de Mediana Edad , Factores de Riesgo , Vitamina K/antagonistas & inhibidores
7.
Thromb Haemost ; 120(11): 1512-1523, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32820486

RESUMEN

The venom of the Australian snake Pseudonaja textilis comprises powerful prothrombin activators consisting of factor X (v-ptFX)- and factor V-like proteins. While all vertebrate liver-expressed factor X (FX) homologs, including that of P. textilis, comprise an activation peptide of approximately 45 to 65 residues, the activation peptide of v-ptFX is significantly shortened to 27 residues. In this study, we demonstrate that exchanging the human FX activation peptide for the snake venom ortholog impedes proteolytic cleavage by the intrinsic factor VIIIa-factor IXa tenase complex. Furthermore, our findings indicate that the human FX activation peptide comprises an essential binding site for the intrinsic tenase complex. Conversely, incorporation of FX into the extrinsic tissue factor-factor VIIa tenase complex is completely dependent on exosite-mediated interactions. Remarkably, the shortened activation peptide allows for factor V-dependent prothrombin conversion while in the zymogen state. This indicates that the active site of FX molecules comprising the v-ptFX activation peptide partially matures upon assembly into a premature prothrombinase complex. Taken together, the shortened activation peptide is one of the remarkable characteristics of v-ptFX that has been modified from its original form, thereby transforming FX into a powerful procoagulant protein. Moreover, these results shed new light on the structural requirements for serine protease activation and indicate that catalytic activity can be obtained without formation of the characteristic Ile16-Asp194 salt bridge via modification of the activation peptide.


Asunto(s)
Venenos Elapídicos/metabolismo , Elapidae/metabolismo , Factor X/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Sitios de Unión , Unión Competitiva , Catálisis , Dominio Catalítico , Cisteína Endopeptidasas , Venenos Elapídicos/genética , Activación Enzimática , Evolución Molecular , Factor VIIIa/metabolismo , Factor VIIa/metabolismo , Factor X/antagonistas & inhibidores , Factor X/genética , Humanos , Complejos Multiproteicos , Fragmentos de Péptidos/farmacología , Pirazoles/farmacología , Piridonas/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Tromboplastina/metabolismo
8.
Semin Thromb Hemost ; 46(8): 986-998, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32688432

RESUMEN

The direct oral anticoagulants targeting coagulation factor Xa or thrombin are widely used as alternatives to vitamin K antagonists in the management of venous thromboembolism and nonvalvular atrial fibrillation. In case of bleeding or emergency surgery, reversal agents are helpful to counteract the anticoagulant therapy and restore hemostasis. While idarucizumab has been established as an antidote for the direct thrombin inhibitor dabigatran, reversal strategies for the direct factor Xa inhibitors have been a focal point in clinical care over the past years. In the absence of specific reversal agents, the off-label use of (activated) prothrombin complex concentrate and recombinant factor VIIa have been suggested as effective treatment options during inhibitor-induced bleeding complications. Meanwhile, several specific reversal agents have been developed. In this review, an overview of the current state of nonspecific and specific reversal agents for the direct factor Xa inhibitors is provided, focusing on the biochemistry and mechanism of action and the preclinical assessment of newly emerging therapies.


Asunto(s)
Anticoagulantes/uso terapéutico , Inhibidores del Factor Xa/uso terapéutico , Humanos
9.
Blood ; 136(5): 533-541, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32457982

RESUMEN

Deep vein thrombosis and pulmonary embolism, collectively defined as venous thromboembolism (VTE), are the third leading cause of cardiovascular death in the United States. Common genetic variants conferring increased varying degrees of VTE risk have been identified by genome-wide association studies (GWAS). Rare mutations in the anticoagulant genes PROC, PROS1 and SERPINC1 result in perinatal lethal thrombosis in homozygotes and markedly increased VTE risk in heterozygotes. However, currently described VTE variants account for an insufficient portion of risk to be routinely used for clinical decision making. To identify new rare VTE risk variants, we performed a whole-exome study of 393 individuals with unprovoked VTE and 6114 controls. This study identified 4 genes harboring an excess number of rare damaging variants in patients with VTE: PROS1, STAB2, PROC, and SERPINC1. At STAB2, 7.8% of VTE cases and 2.4% of controls had a qualifying rare variant. In cell culture, VTE-associated variants of STAB2 had a reduced surface expression compared with reference STAB2. Common variants in STAB2 have been previously associated with plasma von Willebrand factor and coagulation factor VIII levels in GWAS, suggesting that haploinsufficiency of stabilin-2 may increase VTE risk through elevated levels of these procoagulants. In an independent cohort, we found higher von Willebrand factor levels and equivalent propeptide levels in individuals with rare STAB2 variants compared with controls. Taken together, this study demonstrates the utility of gene-based collapsing analyses to identify loci harboring an excess of rare variants with functional connections to a complex thrombotic disease.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Predisposición Genética a la Enfermedad/genética , Tromboembolia Venosa/genética , Adulto , Femenino , Genotipo , Humanos , Masculino , Mutación , Tromboembolia Venosa/sangre , Secuenciación del Exoma/métodos , Factor de von Willebrand/metabolismo
10.
Thromb Haemost ; 120(4): 538-564, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32289858

RESUMEN

Thrombo-inflammation describes the complex interplay between blood coagulation and inflammation that plays a critical role in cardiovascular diseases. The third Maastricht Consensus Conference on Thrombosis assembled basic, translational, and clinical scientists to discuss the origin and potential consequences of thrombo-inflammation in the etiology, diagnostics, and management of patients with cardiovascular disease, including myocardial infarction, stroke, and peripheral artery disease. This article presents a state-of-the-art reflection of expert opinions and consensus recommendations regarding the following topics: (1) challenges of the endothelial cell barrier; (2) circulating cells and thrombo-inflammation, focused on platelets, neutrophils, and neutrophil extracellular traps; (3) procoagulant mechanisms; (4) arterial vascular changes in atherogenesis; attenuating atherosclerosis and ischemia/reperfusion injury; (5) management of patients with arterial vascular disease; and (6) pathogenesis of venous thrombosis and late consequences of venous thromboembolism.


Asunto(s)
Aterosclerosis/inmunología , Enfermedades Cardiovasculares/inmunología , Endotelio Vascular/fisiología , Inflamación/inmunología , Neutrófilos/inmunología , Tromboembolia Venosa/inmunología , Animales , Aterosclerosis/diagnóstico , Aterosclerosis/terapia , Coagulación Sanguínea , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/terapia , Testimonio de Experto , Humanos , Inmunidad Innata , Trombosis , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/terapia
12.
Nucleic Acid Ther ; 29(4): 218-223, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31063435

RESUMEN

Recently, our group reported that a small interfering RNA (siRNA) targeting coagulation factor XII (siF12) leads to an unexpected prothrombotic response in a mouse model where venous thrombosis follows inhibition of endogenous anticoagulants. In this study, we aimed to clarify this unexpected response by evaluating the effects of this siF12 (here, siF12-A) on plasma coagulation through thrombin generation (TG). Besides a routine negative control siRNA (siNEG), we included extra siRNA controls: one siRNA similar to siF12-A except for positions 9-11 of the siRNA that are replaced with its complementary base pairs (siF12-AC9/11), and a second siRNA against F12 (siF12-B). Three days after injection, a significant increase in TG peak height was observed solely for animals injected with siF12-A and siF12-AC9/11, which is considered prothrombotic. As this change in coagulation was unrelated to FXII we conclude that it was off-target. For siRNA studies we now recommend to include mismatch siRNA controls, such as the C9/11 mismatch control used in this study, and to consider plasma coagulation in off-target analysis.


Asunto(s)
Anticoagulantes/farmacología , Factor XII/genética , ARN Interferente Pequeño/genética , Trombosis de la Vena/tratamiento farmacológico , Animales , Anticoagulantes/química , Coagulación Sanguínea/efectos de los fármacos , Reparación de la Incompatibilidad de ADN/genética , Modelos Animales de Enfermedad , Factor XII/antagonistas & inhibidores , Humanos , Ratones , ARN Interferente Pequeño/farmacología , Trombina/genética , Trombosis de la Vena/genética , Trombosis de la Vena/prevención & control
13.
J Thromb Haemost ; 17(8): 1229-1239, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31102425

RESUMEN

Blood coagulation factor Va serves an indispensable role in hemostasis as cofactor for the serine protease factor Xa. In the presence of an anionic phospholipid membrane and calcium ions, factors Va and Xa assemble into the prothrombinase complex. Following formation of the ternary complex with the macromolecular zymogen substrate prothrombin, the latter is rapidly converted into thrombin, the key regulatory enzyme of coagulation. Over the years, multiple binding sites have been identified in factor Va that play a role in the interaction of the cofactor with factor Xa, prothrombin, or the anionic phospholipid membrane surface. In this review, an overview of the currently available information on these interactive sites in factor Va is provided, and data from biochemical approaches and 3D structural protein complex models are discussed. The structural models have been generated in recent years and provide novel insights into the molecular requirements for assembly of both the prothrombinase and the ternary prothrombinase-prothrombin complexes. Integrated knowledge of functionally important regions in factor Va will allow for a better understanding of factor Va cofactor activity.


Asunto(s)
Coagulación Sanguínea , Factor Va/metabolismo , Protrombina/metabolismo , Tromboplastina/metabolismo , Sitios de Unión , Membrana Celular/metabolismo , Factor Va/química , Factor Xa/metabolismo , Humanos , Modelos Moleculares , Fosfolípidos/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Protrombina/química , Relación Estructura-Actividad , Tromboplastina/química
14.
Blood ; 133(19): 2090-2099, 2019 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-30898865

RESUMEN

Tissue factor, coagulation factor XII, platelets, and neutrophils are implicated as important players in the pathophysiology of (experimental) venous thrombosis (VT). Their role became evident in mouse models in which surgical handlings were required to provoke VT. Combined inhibition of the natural anticoagulants antithrombin (Serpinc1) and protein C (Proc) using small interfering RNA without additional triggers also results in a venous thrombotic phenotype in mice, most notably with vessel occlusion in large veins of the head. VT is fatal but is fully rescued by thrombin inhibition. In the present study, we used this VT mouse model to investigate the involvement of tissue factor, coagulation factor XII, platelets, and neutrophils. Antibody-mediated inhibition of tissue factor reduced the clinical features of VT, the coagulopathy in the head, and fibrin deposition in the liver. In contrast, genetic deficiency in, and small interfering RNA-mediated depletion of, coagulation factor XII did not alter VT onset, severity, or thrombus morphology. Antibody-mediated depletion of platelets fully abrogated coagulopathy in the head and liver fibrin deposition. Although neutrophils were abundant in thrombotic lesions, depletion of circulating Ly6G-positive neutrophils did not affect onset, severity, thrombus morphology, or liver fibrin deposition. In conclusion, VT after inhibition of antithrombin and protein C is dependent on the presence of tissue factor and platelets but not on coagulation factor XII and circulating neutrophils. This study shows that distinct procoagulant pathways operate in mouse VT, dependent on the triggering stimulus.


Asunto(s)
Plaquetas/metabolismo , Factor XII/metabolismo , Neutrófilos/metabolismo , Tromboplastina/metabolismo , Trombosis de la Vena/sangre , Animales , Antitrombina III/antagonistas & inhibidores , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL , Proteína C/antagonistas & inhibidores
15.
Thromb Res ; 172: 104-109, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30408635

RESUMEN

INTRODUCTION: Altered levels of factor (F)VIII, prothrombin, or antithrombin have been associated with an increased risk for venous thromboembolism (VTE). However, the exact molecular mechanism by which these altered factor levels modulate the risk is incompletely understood. Here we hypothesize that elevated factor levels affect the pro- and anticoagulant balance in coagulation such that even minute amounts of tissue factor (TF) will initiate thrombin formation, thereby contributing to the VTE risk. MATERIALS AND METHODS: To test this so-called TF-threshold hypothesis, we monitored thrombin generation initiated by very low TF concentrations in FXII-deficient plasma, to avoid any contact pathway-mediated thrombin formation. Furthermore, similar experiments were performed in the presence of increasing concentrations of pro- and anticoagulant proteins. RESULTS: A TF-threshold was established in the FXII-deficient plasma, which is subject to inter-individual variation. Elevated plasma levels of procoagulant factors, such as FVIII or prothrombin, enhanced thrombin generation and reduced the amount of TF required for the initiation of thrombin formation. Conversely, elevated levels of the coagulation inhibitor antithrombin increased the TF-threshold. CONCLUSIONS: Our findings support a mediating role for the TF-threshold in the association between high procoagulant factor levels and the risk for VTE. Furthermore, elevated levels of anticoagulants may have a protective effect on the development of VTE.


Asunto(s)
Factores de Coagulación Sanguínea/metabolismo , Coagulación Sanguínea , Trombina/metabolismo , Tromboplastina/metabolismo , Tromboembolia Venosa/metabolismo , Factores de Coagulación Sanguínea/análisis , Factor VIII/análisis , Factor VIII/metabolismo , Humanos , Protrombina/análisis , Protrombina/metabolismo , Trombina/análisis , Tromboplastina/análisis , Tromboembolia Venosa/sangre
16.
Sci Rep ; 8(1): 15106, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30305662

RESUMEN

Silencing of anticoagulant protein C using RNA interference (siProc) evokes low incident but spontaneous atherothrombosis in the aortic root of apolipoprotein E-deficient (Apoe-/-) mice. The aims of the current study were (1) to analyze if plaque characteristics or circulating factors could be linked to atherothrombosis susceptibility, (2) to increase the incidence of atherothrombosis by transiently increasing blood pressure, and (3) to direct atherothrombosis to an additional predefined vascular site by applying a semi-constrictive collar around the carotid artery. siProc-driven spontaneous atherothrombosis in the aortic root of Apoe-/- mice was reproduced and occurred at an incidence of 23% (9 out of 39 mice), while the incidence of collar-induced atherothrombosis in the carotid artery was 2.6% (1 out of 39 mice). Treatment with phenylephrine, to transiently increase blood pressure, did not increase atherothrombosis in the aortic root of the Apoe-/- mice nor in the carotid arteries with collars. Plaques in the aortic root with an associated thrombus were lower in collagen and macrophage content, and mice with atherothrombosis had significantly more circulating platelets. Plasma protein C, white blood cell counts, total cholesterol, fibrinogen, serum amyloid A, and IL-6 were not different amongst siProc treated mice with or without thrombosis. Remarkably, our data revealed that thrombus formation preferably occurred on plaques in the right coronary sinus of the aortic root. In conclusion, there is a predilection of low protein C-induced spontaneous atherothrombosis in Apoe-/- mice for the right coronary sinus, a process that is associated with an increase in platelets and plaques lower in collagen and macrophage content.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/patología , Seno Coronario/patología , Proteína C/metabolismo , Trombosis/patología , Animales , Apolipoproteínas E/metabolismo , Aterosclerosis/complicaciones , Aterosclerosis/metabolismo , Peso Corporal , Femenino , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Placa Aterosclerótica/complicaciones , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Trombosis/complicaciones , Trombosis/metabolismo
17.
Res Pract Thromb Haemost ; 2(2): 320-326, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30046734

RESUMEN

BACKGROUND: Blood coagulation levels are associated with risk of venous thrombosis (VT). The role of factor (F)V is ambiguous since it plays a dual role in coagulation: it has a procoagulant role when it serves as a cofactor for the activation of thrombin and it has an anticoagulant role by enhancing the inactivation of activated FVIII. OBJECTIVES: To elucidate the association of FV levels with risk of VT. PATIENTS/METHODS: We analyzed FV antigen levels in 2377 patients with VT and 2943 controls from the MEGA study. FV levels were categorized according using the 1st, 2.5th, 5th, 10th, 25th, 50th, 75th, 90th, and 95th percentiles of FV levels in controls as cut-off points. Odds ratios (ORs) were estimated using logistic regression models and adjusted for age and sex, liver disease, FVIII levels, FV Leiden, and TFPI. RESULTS: The risk estimates were U-shaped with increased ORs for the lowest (<0.57 U/dL) levels (OR 1.46; 95% CI 0.87-2.43) as well as the highest (>1.22 U/dL) (OR 1.86; 95% CI 1.46-2.37) levels as compared with the reference group (25th-50th percentile). FVIII adjustment led to attenuation of the OR for high FV levels (OR 1.14; 95% CI 0.88-1.48), with little change for low FV levels (OR 1.68; 95% CI 0.97-2.91). Other adjustments had limited effects. CONCLUSIONS: We found high FV levels to be associated with increased risk for VT, which was explained by concurrently raised FVIII levels. For low levels of factor V, the increased risk for VT could not be explained by the mechanisms we explored.

18.
Nat Rev Dis Primers ; 4: 18028, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29770793

RESUMEN

Pulmonary embolism (PE) is caused by emboli, which have originated from venous thrombi, travelling to and occluding the arteries of the lung. PE is the most dangerous form of venous thromboembolism, and undiagnosed or untreated PE can be fatal. Acute PE is associated with right ventricular dysfunction, which can lead to arrhythmia, haemodynamic collapse and shock. Furthermore, individuals who survive PE can develop post-PE syndrome, which is characterized by chronic thrombotic remains in the pulmonary arteries, persistent right ventricular dysfunction, decreased quality of life and/or chronic functional limitations. Several important improvements have been made in the diagnostic and therapeutic management of acute PE in recent years, such as the introduction of a simplified diagnostic algorithm for suspected PE as well as phase III trials demonstrating the value of direct oral anticoagulants in acute and extended treatment of venous thromboembolism. Future research should aim to address novel treatment options (for example, fibrinolysis enhancers) and improved methods for predicting long-term complications and defining optimal anticoagulant therapy parameters in individual patients, and to gain a greater understanding of post-PE syndrome.


Asunto(s)
Embolia Pulmonar/diagnóstico , Trombosis de la Vena/complicaciones , Anticoagulantes/uso terapéutico , Técnicas de Apoyo para la Decisión , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Humanos , Embolia Pulmonar/epidemiología , Embolia Pulmonar/fisiopatología , Calidad de Vida/psicología , Factores de Riesgo , Factores Sexuales , Trombosis de la Vena/tratamiento farmacológico , Disfunción Ventricular Derecha/complicaciones
19.
Pharmacoepidemiol Drug Saf ; 27(1): 46-51, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29108127

RESUMEN

PURPOSE: Multi-dose drug dispensing (MDD) is a dosing aid that provides patients with disposable bags containing all drugs intended for 1 dosing moment. MDD is believed to increase medication adherence, but studies are based on self-reported data, and results may depend on socially desirable answers. Therefore, our purpose was to determine the effect of MDD on medication adherence in non-adherent patients taking vitamin K antagonists (VKAs), and to compare with instructing patients on medication use. METHODS: We conducted a before-after study in non-adherent patients where MDD was the exposure and change in adherence after MDD initiation was the outcome (within patient comparison). Time in therapeutic range (TTR) was selected as a measure for adherence, as this reflects stability of VKA treatment. To analyze whether MDD improved adherence as compared with standard care (ie, letters or calls from nurses of the anticoagulation clinic), non-adherent patients without MDD were also followed to estimate their TTR change over time (between patient comparison). RESULTS: Eighty-three non-adherent VKA patients started using MDD. The median TTR was 63% before MDD and 73% 6 months after MDD. The within patient TTR increased on average by 13% (95%CI 6% to 21%) within 1 month after starting MDD and remained stable during the next 5 months. The TTR of MDD-patients increased 10% (95%CI 2% to 19%) higher as compared with non-MDD patients within 1 month but was similar after 4 months (TTR difference 3%, 95%CI -2% to 9%). CONCLUSIONS: Adherence improved after initiation of MDD. Compared with instructing patients, MDD was associated with better adherence within 1 month but was associated with similar improvement after 4 months.


Asunto(s)
Anticoagulantes/uso terapéutico , Embalaje de Medicamentos/métodos , Cumplimiento de la Medicación/estadística & datos numéricos , Tromboembolia/prevención & control , Vitamina K/antagonistas & inhibidores , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Educación del Paciente como Asunto , Servicios Postales , Evaluación de Programas y Proyectos de Salud , Teléfono
20.
PLoS One ; 12(11): e0187699, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29117201

RESUMEN

BACKGROUND: Family studies have shown a strong heritability component for venous thromboembolism (VTE), but established genetic risk factors are present in only half of VTE patients. AIM: To identify genetic risk factors in two large families with unexplained hereditary VTE. METHODS: We performed whole exome sequencing in 10 affected relatives of two unrelated families with an unexplained tendency for VTE. We prioritized variants shared by all affected relatives from both families, and evaluated these in the remaining affected and unaffected individuals. We prioritized variants based on 3 different filter strategies: variants within candidate genes, rare variants across the exome, and SNPs present in patients with familial VTE and with low frequency in the general population. We used whole exome sequencing data available from 96 unrelated VTE cases with a positive family history of VTE from an affected sib study (the GIFT study) to identify additional carriers and compared the risk-allele frequencies with the general population. Variants found in only one individual were also retained for further analysis. Finally, we assessed the association of these variants with VTE in a population-based case-control study (the MEGA study) with 4,291 cases and 4,866 controls. RESULTS: Six variants remained as putative disease-risk candidates. These variants are located in 6 genes spread among 3 different loci: 2p21 (PLEKHH2 NM_172069:c.3105T>C, LRPPRC rs372371276, SRBD1 rs34959371), 5q35.2 (UNC5A NM_133369.2:c.1869+23C>A), and 17q25.1 (GPRC5C rs142232982, RAB37 rs556450784). In GIFT, additional carriers were identified only for the variants located in the 2p21 locus. In MEGA, additional carriers for several of these variants were identified in both cases and controls, without a difference in prevalence; no carrier of the UNC5A variant was present. CONCLUSION: Despite sequencing of several individuals from two thrombophilic families resulting in 6 candidate variants, we were unable to confirm their relevance as novel thrombophilic defects.


Asunto(s)
Exoma , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/genética , Adulto , Alelos , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Sitios Genéticos , Humanos , Masculino , Persona de Mediana Edad , Linaje , Factores de Riesgo , Análisis de Secuencia de ADN , Tromboembolia Venosa/patología , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...