Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Thromb Res ; 221: 51-57, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36470070

RESUMEN

Local coagulation activation has been shown to impact both primary tumor growth and metastasis in mice. It is well known that components of the blood clotting cascade such as tissue factor and thrombin play a role in tumor progression by activating cellular receptors and local formation of fibrin. However, whether venous thromboembolism (VTE) or a hypercoagulable state has a direct impact on cancer progression is unknown. Here we have combined an orthotopic murine breast cancer model, using female Nod-SCID mice, with siRNA-mediated silencing of antithrombin (siAT) leading to the induction of a systemic hypercoagulable state. We show that, compared to control siRNA-treated (not experiencing a hypercoagulable state) tumor-bearing mice, siAT treated tumor-bearing mice do not show enhanced tumor growth nor enhanced metastasis. We conclude that, in this murine model for hypercoagulability, induction of a hypercoagulable state does not contribute to breast cancer progression.


Asunto(s)
Neoplasias de la Mama , Trombofilia , Humanos , Femenino , Animales , Ratones , Antitrombinas , Modelos Animales de Enfermedad , Xenoinjertos , Ratones SCID , Trombofilia/genética , Anticoagulantes , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/genética , Antitrombina III/genética , ARN Interferente Pequeño
2.
Orphanet J Rare Dis ; 17(1): 178, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35505430

RESUMEN

BACKGROUND: Cushing's syndrome (CS) is associated with an hypercoagulable state and an increased risk of venous thromboembolism (VTE). Evidence-based guidelines on thromboprophylaxis strategies in patients with CS are currently lacking. We aimed to map the current clinical practice for thromboprophylaxis management in patients with CS across reference centers (RCs) of the European Reference Network on Rare Endocrine Conditions (Endo-ERN), which are endorsed specifically for the diagnosis and treatment of CS. Using the EU survey tool, a primary screening survey, and subsequently a secondary, more in-depth survey were developed. RESULTS: The majority of the RCs provided thromboprophylaxis to patients with CS (n = 23/25), although only one center had a standardized thromboprophylaxis protocol (n = 1/23). RCs most frequently started thromboprophylaxis from CS diagnosis onwards (n = 11/23), and the majority stopped thromboprophylaxis based on individual patient characteristics, rather than standardized treatment duration (n = 15/23). Factors influencing the initiation of thromboprophylaxis were 'medical history of VTE' (n = 15/23) and 'severity of hypercortisolism' (n = 15/23). Low-Molecular-Weight-Heparin was selected as the first-choice anticoagulant drug for thromboprophylaxis by all RCs (n = 23/23). Postoperatively, the majority of RCs reported 'severe immobilization' as an indication to start thromboprophylaxis in patients with CS (n = 15/25). Most RCs (n = 19/25) did not provide standardized testing for variables of hemostasis in the postoperative care of CS. Furthermore, the majority of the RCs provided preoperative medical treatment to patients with CS (n = 23/25). About half of these RCs (n = 12/23) took a previous VTE into account when starting preoperative medical treatment, and about two-thirds (n = 15/23) included 'reduction of VTE risk' as a goal of treatment. CONCLUSIONS: There is a large practice variation regarding thromboprophylaxis management and perioperative medical treatment in patients with CS, even in Endo-ERN RCs. Randomized controlled trials are needed to establish the optimal prophylactic anticoagulant regimen, carefully balancing the increased risk of (perioperative) bleeding, and the presence of additional risk factors for thrombosis.


Asunto(s)
Síndrome de Cushing , Enfermedades del Sistema Endocrino , Endometriosis , Tromboembolia Venosa , Anticoagulantes/uso terapéutico , Síndrome de Cushing/complicaciones , Síndrome de Cushing/tratamiento farmacológico , Femenino , Humanos , Enfermedades Raras/tratamiento farmacológico , Tromboembolia Venosa/tratamiento farmacológico , Tromboembolia Venosa/prevención & control
3.
J Thromb Haemost ; 16(7): 1357-1368, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29734512

RESUMEN

Essentials Substitution therapy for von Willebrand (VW) disease leaves mutant VW factor (VWF) unhindered. Presence of mutant VWF may negatively affect phenotypes despite treatment. Inhibition of VWF by allele-specific siRNAs targeting single-nucleotide polymorphisms is effective. Allele-specific inhibition of VWF p.Cys2773Ser improves multimerization. SUMMARY: Background Treatment of the bleeding disorder von Willebrand disease (VWD) focuses on increasing von Willebrand factor (VWF) levels by administration of desmopressin or VWF-containing concentrates. Both therapies leave the production of mutant VWF unhindered, which may have additional consequences, such as thrombocytopenia in patients with VWD type 2B, competition between mutant and normal VWF for platelet receptors, and the potential development of intestinal angiodysplasia. Most cases of VWD are caused by dominant-negative mutations in VWF, and we hypothesize that diminishing expression of mutant VWF positively affects VWD phenotypes. Objectives To investigate allele-specific inhibition of VWF by applying small interfering RNAs (siRNAs) targeting common single-nucleotide polymorphisms (SNPs) in VWF. This approach allows allele-specific knockdown irrespective of the mutations causing VWD. Methods Four SNPs with a high predicted heterozygosity within VWF were selected, and siRNAs were designed against both alleles of the four SNPs. siRNA efficiency, allele specificity and siRNA-mediated phenotypic improvements were determined in VWF-expressing HEK293 cells. Results Twelve siRNAs were able to efficiently inhibit single VWF alleles in HEK293 cells that stably produce VWF. Transient cotransfections of these siRNAs with two VWF alleles resulted in a clear preference for the targeted allele over the untargeted allele for 11 siRNAs. We also demonstrated siRNA-mediated phenotypic improvement of the VWF multimerization pattern of the VWD type 2A mutation VWF p.Cys2773Ser. Conclusions Allele-specific siRNAs are able to distinguish VWF alleles on the basis of one nucleotide variation, and are able to improve a severe multimerization defect caused by VWF p.Cys2773Ser. This holds promise for the therapeutic application of allele-specific siRNAs in dominant-negative VWD.


Asunto(s)
Mutación , Polimorfismo de Nucleótido Simple , ARN Interferente Pequeño/genética , Tratamiento con ARN de Interferencia , Enfermedades de von Willebrand/terapia , Factor de von Willebrand/genética , Estudios de Factibilidad , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Fenotipo , Multimerización de Proteína , ARN Interferente Pequeño/metabolismo , Enfermedades de von Willebrand/sangre , Enfermedades de von Willebrand/genética , Factor de von Willebrand/metabolismo
4.
Cell Death Differ ; 13(12): 2089-98, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16729027

RESUMEN

p53 is a potent inhibitor of cell growth and an inducer of apoptosis. During embryonic development, Mdm2 and Mdm4 inhibit the growth suppressive activities of p53. However, whether tight surveillance of p53 activity is required in quiescent cells is unknown. To test this, conditional inactivation of mdm2 and mdm4 was carried out in smooth muscle cells (SMCs). Upon SMC-specific inactivation of mdm2, and not of mdm4, mice rapidly became ill and died. Necropsy showed small intestinal dilation, and histological analyses indicated a severe reduction in the number of intestinal SMCs. Increased p53 levels and activity were detected in the remaining SMCs, and the phenotype was completely rescued on a p53-null background. Interestingly, intestinal SMCs are caspase-3-negative and therefore did not undergo caspase-3-dependent apoptotic cell death. Together, Mdm2, but not Mdm4, prevents accumulation of active p53 in quiescent SMCs and thereby the induction of p53-mediated caspase-3-independent cell death.


Asunto(s)
Apoptosis/fisiología , Caspasa 3/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Apoptosis/genética , Caspasa 3/genética , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/fisiología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Ratones , Ratones Transgénicos , Miocitos del Músculo Liso/patología , Proteína p53 Supresora de Tumor/genética
5.
J Thromb Haemost ; 5(10): 2079-86, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17666016

RESUMEN

BACKGROUND: Mouse models have become increasingly important in thrombosis research. However, only a limited number of assays are available for assessment of the coagulation system in mouse plasma. OBJECTIVES: To quantify tissue factor-initiated thrombin generation in murine platelet-rich and platelet-free plasma and to develop a test for measurement of resistance to activated protein C (APC) in mouse plasma. METHODS: Thrombin generation was monitored with calibrated automated thrombography (CAT) using a low-affinity fluorogenic substrate for thrombin. RESULTS: To overcome the higher activity of coagulation inhibitors in mouse plasma as compared with human plasma, the reaction temperature was lowered to 33 degrees C and the assay was carried out at a 2-fold higher final plasma dilution (1:3) than commonly used for CAT in human plasma. This increased the endogenous thrombin potential (ETP) 4- to 5-fold and enabled reliable measurement of thrombin generation in both platelet-free and platelet-rich mouse plasma. For the APC resistance measurement, the reaction conditions were further optimized with respect to tissue factor, phospholipid, APC and CaCl(2) concentrations. The test was validated using plasma of mice with different genetic background with respect to the factor V Leiden mutation (FV Leiden). Mice homozygous for FV Leiden had higher APC sensitivity ratios (mean 5.46; 95% CI 4.88-6.03) than heterozygous FV Leiden mice (mean 4.21; 95% CI 3.53-4.89) and than wild-type mice (mean 2.71; 95%CI 2.15-3.27). CONCLUSIONS: We have established reaction conditions for measurement of thrombin generation and APC resistance in mouse plasma. This assay enables evaluation of the coagulation system and the function of the protein C system in mouse models.


Asunto(s)
Resistencia a la Proteína C Activada/genética , Pruebas de Coagulación Sanguínea/instrumentación , Pruebas de Coagulación Sanguínea/métodos , Trombina/química , Adulto , Animales , Automatización , Plaquetas/metabolismo , Proteínas Sanguíneas/metabolismo , Calibración , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Protrombina/metabolismo , Sensibilidad y Especificidad , Trombina/metabolismo
6.
Arterioscler Thromb Vasc Biol ; 26(12): 2710-5, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17038633

RESUMEN

OBJECTIVE: In vitro studies implicate that the low-density lipoprotein receptor (LDLR)-related protein (LRP) in macrophages has a pro-atherogenic potential. In the present study, we investigated the in vivo role of macrophage specific LRP in atherogenesis independent of its role in the uptake of lipoproteins. METHODS AND RESULTS: We generated macrophage-specific LRP-deficient mice on an apoE/LDLR double-deficient background. Macrophage LRP deletion did not affect plasma cholesterol and triglyceride levels, lipoprotein distribution, and blood monocyte counts. Nevertheless, macrophage LRP deficiency resulted in a 1.8-fold increase in total atherosclerotic lesion area in the aortic root of 18-week-old mice. Moreover, LRP deficiency also resulted in a relatively higher number of advanced lesions. Whereas macrophage and smooth muscle cell content did not differ between LRP-deficient mice and control littermates, a 1.7-fold increase in collagen content and 2.3-fold decrease in relative number of CD3+ T cells were observed in lesions from macrophage specific LRP-deficient mice. CONCLUSIONS: Our data demonstrate that independent of its role in lipoprotein uptake, absence of LRP in macrophages resulted in more advanced atherosclerosis and in lesions that contained more collagen and less CD3+ T cells. In contrast to previous in vitro studies, we conclude that macrophage LRP has an atheroprotective potential and may modulate the extracellular matrix in the atherosclerotic lesions.


Asunto(s)
Apolipoproteínas E/metabolismo , Aterosclerosis/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Macrófagos/metabolismo , Receptores de LDL/metabolismo , Animales , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Colágeno/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Lipoproteínas/sangre , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Macrófagos/patología , Ratones , Ratones Noqueados , Receptores de LDL/genética
7.
J Thromb Haemost ; 3(6): 1257-65, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15946216

RESUMEN

BACKGROUND: Coagulation factor VIII (FVIII) is a heavily glycosylated heterodimeric plasma protein that consists of a heavy (domains A1-A2-B) and light chain (domains A3-C1-C2). It has been well established that the clearance of FVIII from the circulation involves mechanisms that are sensitive to the low-density lipoprotein receptor (LDLR) family antagonist receptor-associated protein (RAP), including LDLR-related protein. Because FVIII clearance in the presence of a bolus injection of RAP still occurs fairly efficient, also RAP-independent mechanisms are likely to be involved. OBJECTIVES: In the present study, we investigated the interaction of FVIII with the endocytic lectin asialoglycoprotein receptor (ASGPR) and the physiological relevance thereof. METHODS AND RESULTS: Surface plasmon resonance studies demonstrated that FVIII dose-dependently bound to ASGPR with high affinity (Kd approximately 2 nM). FVIII subunits were different in that only the heavy chain displayed high-affinity binding to ASGPR. Studies employing a FVIII variant that lacks the B domain revealed that FVIII-ASGPR complex assembly is driven by structure elements within the B domain of the heavy chain. The FVIII heavy chain-ASGPR interaction required calcium ions and was inhibited by soluble D-galactose. Furthermore, deglycosylation of the FVIII heavy chain by endoglycosidase F completely abrogated the interaction with ASGPR. In clearance experiments in mice, the FVIII mean residence time was prolonged by the ASGPR-antagonist asialo-orosomucoid (ASOR). CONCLUSIONS: We conclude that asparagine-linked oligosaccharide structures of the FVIII B domain recognize the carbohydrate recognition domains of ASGPR and that an ASOR-sensitive mechanism, most likely ASGPR, contributes to the catabolism of coagulation FVIII in vivo.


Asunto(s)
Receptor de Asialoglicoproteína/metabolismo , Factor VIII/metabolismo , Animales , Sitios de Unión , Calcio/farmacología , Carbohidratos , Factor VIII/química , Factor VIII/farmacocinética , Galactosa/farmacología , Glicosilación , Humanos , Masculino , Tasa de Depuración Metabólica , Ratones , Ratones Endogámicos C57BL , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Subunidades de Proteína/metabolismo , Resonancia por Plasmón de Superficie
10.
J Thromb Haemost ; 8(8): 1838-46, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20524981

RESUMEN

BACKGROUND: Oral estrogen use is associated with changes in plasma levels of many coagulation proteins. OBJECTIVE: To gain more insight into the underlying mechanism of estrogen-induced changes in coagulation. METHODS: Ovariectomized female mice were used to study the impact of oral 17α-ethinylestradiol (EE) on plasma coagulation, hepatic coagulation gene transcript levels, and dependence on estrogen receptor (ER) α and ERß. RESULTS: Ten days of oral EE treatment resulted in significantly reduced plasma activity levels of factor (F)VIII, FXII, combined FII/FVII/FX and antithrombin, whereas FIX activity significantly increased. Regarding hepatic transcript levels, oral EE caused significant decreases in fibrinogen-γ, FII, FV, FVII, FX, FXII, antithrombin, protein C, protein Z, protein Z inhibitor and heparin cofactor II mRNA levels, whereas FXI levels significantly increased and transcript levels of FVIII, FIX, protein S and α(2) -antiplasmin remained unaffected. All EE-induced coagulation-related changes were neutralized by coadministration of the non-specific ER antagonist ICI182780. In addition, ERα-deficient mice lacked the EE-induced changes in plasma coagulation and hepatic transcript profile, whereas ERß-deficient mice responded similarly to non-deficient littermate controls. A crucial role for the ER was further demonstrated by its rapid effects on transcription, within 2.5-5 h after EE administration, suggesting a short chain of events leading to its final effects. CONCLUSIONS: Oral EE administration has a broad impact on the mouse coagulation profile at the level of both plasma and hepatic mRNA levels. The effects on transcription are rapidly induced, mostly downregulatory, and principally mediated by ERα.


Asunto(s)
Receptor alfa de Estrógeno/genética , Etinilestradiol/metabolismo , Administración Oral , Animales , Coagulación Sanguínea , Estradiol/análogos & derivados , Estradiol/farmacología , Receptor beta de Estrógeno/genética , Femenino , Fulvestrant , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/metabolismo , Factores de Tiempo
11.
J Thromb Haemost ; 7(2): 312-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19036061

RESUMEN

BACKGROUND: Pregnancy, oral contraceptive (OC)use and hormone replacement therapy (HRT) are established risk factors for venous thrombosis. Acquired resistance to activated protein C (APC) has been proposed to contribute to the increased thrombosis risk. Mouse models are often used for preclinical testing of newly developed hormone preparations. However, it is not known whether hormone-induced APC resistance is also observed in laboratory animals. OBJECTIVES: To investigate whether hormonal changes modulate APC resistance in mice, we used pregnant mice as a model of hormone-induced APC resistance. The effect of pregnancy on APC resistance was studied in wild-type and factor (F)V Leiden mice. METHODS: APC resistance was determined in mouse plasma using a thrombin generation-based APC resistance test. APC resistance determinants,i.e. prothrombin, FV, FX, antithrombin and protein S levels,and of tissue factor pathway inhibitor (TFPI) activity were evaluated in plasma from non-pregnant and pregnant mice. RESULTS: In contrast to humans, pregnancy induced a decrease in APC resistance in wild-type and in FV Leiden mice.Pregnant mice had higher levels of prothrombin, FV, FX,protein S and TFPI activity as compared with non-pregnant mice. CONCLUSIONS: Pregnancy causes a decrease in APC resistance in mice, which can be explained by the elevation of protein S levels and increased TFPI activity in plasma. Our findings show species specificity in the effects of pregnancy on the major determinants of the protein C system and suggest that protein S and TFPI play an important role in the development of pregnancy-induced APC resistance in humans.


Asunto(s)
Resistencia a la Proteína C Activada/etiología , Factor V , Hemostasis , Lipoproteínas/sangre , Proteína S/análisis , Resistencia a la Proteína C Activada/diagnóstico , Animales , Biomarcadores/sangre , Femenino , Ratones , Embarazo , Complicaciones Hematológicas del Embarazo/sangre , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA