Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Haemophilia ; 16(102): 6-15, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-18771423

RESUMEN

Factor VIII (FVIII) is a plasma protein critical to the haemostatic system. This notion is illustrated by the severe bleeding disorder that is associated with its functional absence, known as haemophilia A. In addition, several epidemiological studies have revealed an association between the presence of elevated levels of FVIII and thrombotic complications. In view of its relation to thrombotic and haemorrhagic disorders, it is not surprising that FVIII has gained wide attention from the research community in the previous decades. This research has led to a better understanding of not only the structural, functional and physiological aspects of this intriguing protein, but also of the pathogenesis of haemostatic defects associated with FVIII. In the present review, focus will be on the interaction between FVIII and surface receptors that are able to capture FVIII. These interactions are of importance for FVIII, as they may affect both function and survival of FVIII.


Asunto(s)
Factor VIII/fisiología , Hemofilia A/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/fisiología , Coagulación Sanguínea/fisiología , Factor VIII/metabolismo , Humanos , Factor de von Willebrand/fisiología
2.
Haemophilia ; 16 Suppl 5: 194-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20590881

RESUMEN

SUMMARY: Although factor VIII (FVIII) and von Willebrand factor (VWF) are products of two distinct genes, they circulate in plasma as a tight non-covalent complex. Moreover, they both play a critical role in the haemostatic process, a fact that is illustrated by the severe bleeding tendency associated with the functional absence of either protein. FVIII is an essential cofactor for coagulation factor IX, while VWF is pertinent to the recruitment of platelets to the injured vessel wall under conditions of rapid flow. FVIII and VWF have in common that they are heavily glycosylated: full-length FVIII contains 20 N-linked and at least seven O-linked glycans, while VWF contains 12 N-linked and 10 O-linked glycans. Three decades of research have revealed that the carbohydrate structures of FVIII and VWF contribute to many of the steps that can be distinguished in the life-cycle of these proteins, including biosynthesis/secretion, function and clearance. In this review, several of these aspects will be discussed. In addition, the interaction of the FVIII/VWF complex with two families of carbohydrate-binding proteins, i.e. Galectins and Siglecs, and their potential physiological relevance will be discussed.


Asunto(s)
Factor VIII/química , Polisacáridos/química , Factor de von Willebrand/química , Secuencia de Aminoácidos , Factor VIII/biosíntesis , Glicosilación , Humanos , Estructura Terciaria de Proteína , Enfermedades de von Willebrand/tratamiento farmacológico , Factor de von Willebrand/biosíntesis
3.
J Thromb Haemost ; 6(1): 90-6, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17922807

RESUMEN

BACKGROUND: The effect of exercise on von Willebrand factor (VWF) and ADAMTS-13 levels in individuals with von Willebrand disease (VWD) has never been reported. OBJECTIVES: The aim was to quantify the effect of a standardized exercise protocol on individuals with type 1 and type 2B VWD. PATIENTS/METHODS: Thirty individuals from three groups (10 controls, 11 with type 1 VWD and 9 with type 2B VWD) completed the Standard Bruce Protocol Treadmill Test. A bleeding questionnaire was administered and blood tests were performed pre- and immediately postexercise. The groups were well matched for age, gender and body mass index (BMI). RESULTS: There was a correlation in all groups between the metabolic equivalents (METS) achieved and the degree of change of VWF and FVIII:C levels (P < 0.002, Pearson's correlation). There was a significant postexercise increase in VWF:Ag, VWF:RCo, FVIII:C and activated VWF levels in both the control group and in the type 2B VWD group, but not in the type 1 VWD group. Specific to the type 2B VWD group was an increase in the percentage of high molecular weight multimers (P = 0.022), a decrease in the mean platelet count compared with the other groups (P < 0.001) and an increase in the ADAMTS-13 level (P = 0.001). CONCLUSIONS: There are significant differences in the effects of exercise on individuals with type 1 and type 2B VWD compared with controls. Further clinical studies are necessary to evaluate exercise as a therapeutic option in VWD.


Asunto(s)
Proteínas ADAM/sangre , Ejercicio Físico/fisiología , Enfermedades de von Willebrand/sangre , Enfermedades de von Willebrand/terapia , Factor de von Willebrand/análisis , Proteína ADAMTS13 , Estudios de Casos y Controles , Factor VIII/análisis , Humanos , Recuento de Plaquetas , Enfermedades de von Willebrand/clasificación
4.
J Thromb Haemost ; 5(9): 1956-62, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17723135

RESUMEN

BACKGROUND AND OBJECTIVE: Osteoprotegerin (OPG), a member of the tumor necrosis-factor receptor superfamily, plays an important role in bone remodeling and is also involved in vascular diseases. OPG is physically associated with von Willebrand factor (VWF), a glycoprotein involved in primary hemostasis, within the Weibel-Palade bodies (WPBs) of endothelial cells and in plasma. The present study aimed to elucidate the molecular mechanisms underlying the interaction between OPG and VWF. METHODS AND RESULTS: In a solid-phase binding assay, VWF was able to bind specifically to OPG in a calcium-dependent manner. This interaction displayed strong pH dependence with optimal binding occurring at pH 6.5 and was severely impaired by chloride-ion concentrations above 40 mm. Using a series of purified VWF derivatives the functional site that supports VWF interaction with OPG was localized on its Al domain. Fluorescence microscopy on human umbilical vein endothelial cells showed co-localization of VWF and OPG in WPBs. When secretion was induced, OPG remained associated with VWF in extracellular patches of release under biochemical conditions found in blood plasma. CONCLUSIONS: Our observations demonstrate the existence of an interactive site for OPG within the VWF A1-domain. This study established that the optimal biochemical parameters allowing a complex formation between VWF and OPG are those thought to prevail in the trans-Golgi network. These conditions would allow VWF to act as a cargo targeting OPG to WPBs. Finally, blood environments appear suitable to preserve the complex, which may participate in vascular injury, arterial calcification and inflammation.


Asunto(s)
Osteoprotegerina/metabolismo , Factor de von Willebrand/metabolismo , Secuencia de Aminoácidos , Células Cultivadas , Humanos , Osteoprotegerina/química , Unión Proteica , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie
5.
J Thromb Haemost ; 15(7): 1285-1294, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28671350

RESUMEN

Von Willebrand factor (VWF) is a plasma glycoprotein best known for its crucial hemostatic role in serving as a molecular bridge linking platelets to subendothelial components following vascular injury. In addition, VWF functions as chaperone for coagulation factor VIII. In pathological settings, VWF is recognized as a risk factor for both arterial and venous thrombosis, as well as a molecular player that directly promotes the thrombotic process. Recent years have seen the emergence of the concept of immuno-thrombosis by which inflammatory cells participate in thrombotic processes. In return, reports about the involvement of hemostatic proteins or cells (such as platelets) in inflammatory responses have become increasingly common, emphasizing the intricate link between hemostasis and inflammation. However, evidence of a link between VWF and inflammation arose much earlier than these recent developments. At first, VWF was considered only as a marker of inflammation in various pathologies, due to its acute release by the activated endothelium. Later on, a more complex role of VWF in inflammation was uncovered, owing to its capacity to direct the biogenesis of specific endothelial organelles, the Weibel-Palade bodies that contain known inflammation players such as P-selectin. Finally, a more direct link between VWF and inflammation has become apparent with the discovery that VWF is able to recruit leukocytes, either via direct leukocyte binding or by recruiting platelets which in turn will attract leukocytes. This review will focus on these different aspects of the connection between VWF and inflammation, with particular emphasis on VWF-leukocyte interactions.


Asunto(s)
Inflamación , Trombosis de la Vena/metabolismo , Factor de von Willebrand/metabolismo , Proteína ADAMTS13/metabolismo , Animales , Plaquetas/metabolismo , Células Endoteliales/metabolismo , Endotelio Vascular/metabolismo , Glicoproteínas/metabolismo , Hemostasis , Humanos , Leucocitos/citología , Ligandos , Ratones , Neutrófilos/metabolismo , Selectina-P/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Factores de Riesgo , Cuerpos de Weibel-Palade
6.
J Thromb Haemost ; 4(12): 2569-75, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16968329

RESUMEN

BACKGROUND: HELLP (hemolysis, elevated liver enzymes and low platelets) syndrome is a severe complication of pre-eclampsia in pregnancy, characterized by microvascular platelet thrombi. Activation of the endothelium is thought to play a key role in pre-eclampsia and HELLP syndrome. Activation of endothelial cells may lead to release of von Willebrand factor (VWF) multimers, which are highly reactive with platelets. Normally, newly released multimers are cleaved by ADAMTS13, resulting in less reactive derivatives. OBJECTIVE: We hypothesized that HELLP syndrome is characterized by increased amounts of active VWF compared with healthy pregnancy and pre-eclampsia, due to acute activation of endothelial cells. This might contribute to thrombocytopenia and thrombotic microangiopathy. METHODS: Active VWF and ADAMTS13 activity were measured in healthy pregnant volunteers (n = 9), patients with pre-eclampsia (n = 6) and patients with HELLP syndrome (n = 14) at similar gestational ages. To study the role of endothelial cell activation, the propeptide/mature VWF ratio was determined, and VWF released by cultured endothelial cells was analyzed. RESULTS: Active VWF levels were increased 2.1-fold in HELLP syndrome compared with healthy pregnant volunteers (P < 0.001) and 1.6-fold compared with patients with pre-eclampsia (P = 0.001). ADAMTS13 activity was moderately decreased in patients with HELLP syndrome compared with healthy pregnant volunteers (P < 0.004), but not compared with patients with pre-eclampsia. The propeptide/mature VWF ratio was increased 1.7-fold compared with healthy pregnant volunteers (P < 0.001) and 1.5-fold compared with patients with pre-eclampsia (P < 0.05). A significant correlation was found between this ratio and the activation factor of VWF (r = 0.68, P < 0.001). The amount of active VWF was increased 1.4-fold in medium of stimulated endothelial cells when compared with non-stimulated cells (P < 0.05). CONCLUSION: Acute endothelial cell activation in HELLP syndrome and decreased ADAMTS13 activity result in increased amounts of active VWF. This might explain the consumptive thrombocytopenia and thrombotic microangiopathy associated with HELLP syndrome. Inhibition of circulating active VWF could be a potential new approach in the treatment of patients with HELLP syndrome.


Asunto(s)
Células Endoteliales/metabolismo , Síndrome HELLP/metabolismo , Preeclampsia/metabolismo , Precursores de Proteínas/metabolismo , Factor de von Willebrand/metabolismo , Proteínas ADAM/sangre , Proteínas ADAM/metabolismo , Proteína ADAMTS13 , Adulto , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Femenino , Edad Gestacional , Síndrome HELLP/sangre , Humanos , Glicoproteínas de Membrana , Proteínas de la Membrana/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria , Preeclampsia/sangre , Embarazo , Unión Proteica , Acetato de Tetradecanoilforbol/farmacología , Venas Umbilicales/citología , Venas Umbilicales/efectos de los fármacos , Venas Umbilicales/metabolismo , Enfermedades de von Willebrand/metabolismo
7.
J Thromb Haemost ; 4(8): 1680-90, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16879209

RESUMEN

The antiphospholipid syndrome (APS) is a non-inflammatory autoimmune disease characterized by arterial and/or venous thrombosis and/or pregnancy morbidity in the presence of autoantibodies that recognize beta2-glycoprotein I (beta2GPI) bound to phospholipids. We have previously demonstrated that dimerization of beta2GPI by autoantibodies induces platelet activation, involving the platelet receptor apolipoprotein E receptor 2' (apoER2') a receptor belonging to the low-density lipoprotein receptor (LDL-R) family. Here, we show that dimeric beta2GPI, but not monomeric beta2GPI, interacts with four other LDL-R family members: the LDL-R related protein (LRP), megalin, the LDL-R and the very-low density lipoprotein receptor (VLDL-R). Interaction between dimeric beta2GPI and LDL-R, apoER2' and VLDL-R was best described with a one-site binding model (half-maximal binding; approximately 20 nm for apoER2' and VLDL-R and approximately 300 nm for LDL-R), whereas the interaction between dimeric beta2GPI and LRP or megalin was best described with a two-site binding model, representing a high- (approximately 3 nm) and a low-affinity site (approximately 0.2 microm). Binding to all receptors tested was unaffected by a tryptophane to serine (W316S) substitution in domain V of beta2GPI, which is known to disrupt the phospholipid binding site of beta2GPI. Also deletion of domain I or II left the interaction with the receptors unaffected. Deletion of domain V, however, significantly decreased the affinity for the receptors. In conclusion, our data show that dimeric beta2GPI can interact with different LDL-R family members. This interaction is dependent on a binding site within domain V of beta2GPI, which does not overlap with the phospholipid-binding site within domain V.


Asunto(s)
Receptores de LDL/metabolismo , beta 2 Glicoproteína I/sangre , beta 2 Glicoproteína I/metabolismo , Animales , Sitios de Unión , Línea Celular , Cricetinae , Dimerización , Relación Dosis-Respuesta a Droga , Heparina/química , Humanos , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie , Factores de Tiempo , beta 2 Glicoproteína I/química
8.
J Thromb Haemost ; 4(3): 519-26, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16405520

RESUMEN

BACKGROUND: The key role played by von Willebrand factor (VWF) in platelet adhesion suggests a potential implication in various pathologies, where this process is involved. In cancer metastasis development, tumor cells interact with platelets and the vessel wall to extravasate from the circulation. As a potential mediator of platelet-tumor cell interactions, VWF could influence this early step of tumor spread and therefore play a role in cancer metastasis. OBJECTIVES: To investigate whether VWF is involved in metastasis development. METHODS: In a first step, we characterized the interaction between murine melanoma cells B16-BL6 and VWF in vitro. In a second step, an experimental metastasis model was used to compare the formation of pulmonary metastatic foci in C57BL/6 wild-type and VWF-null mice following the injection of B16-BL6 cells or Lewis lung carcinoma cells. RESULTS: In vitro adhesion assays revealed that VWF is able to promote a dose-dependent adhesion of B16-BL6 cells via its Arg-Gly-Asp (RGD) sequence. In the experimental metastasis model, we found a significant increase in the number of pulmonary metastatic foci in VWF-null mice compared with the wild-type mice, a phenotype that could be corrected by restoring VWF plasma levels. We also showed that increased survival of the tumor cells in the lungs during the first 24 h in the absence of VWF was the cause of this increased metastasis. CONCLUSION: These findings suggest that VWF plays a protective role against tumor cell dissemination in vivo. Underlying mechanisms remain to be investigated.


Asunto(s)
Carcinoma Pulmonar de Lewis/patología , Neoplasias Pulmonares/prevención & control , Melanoma Experimental/patología , Factor de von Willebrand/genética , Animales , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Integrina alfaVbeta3/efectos de los fármacos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Trasplante de Neoplasias , Proteínas Recombinantes/farmacología , Factor de von Willebrand/farmacología
9.
J Thromb Haemost ; 14(12): 2458-2470, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27614059

RESUMEN

Essentials Factor VIIa is cleared principally as a complex with antithrombin. Enzyme/serpin complexes are preferred ligands for the scavenger-receptor LRP1. Factor VIIa/antithrombin but not factor VIIa alone is a ligand for LRP1. Macrophage-expressed LRP1 contributes to the clearance of factor VIIa/antithrombin. SUMMARY: Background Recent findings point to activated factor VII (FVIIa) being cleared predominantly (± 65% of the injected protein) as part of a complex with the serpin antithrombin. FVIIa-antithrombin complexes are targeted to hepatocytes and liver macrophages. Both cells lines abundantly express LDL receptor-related protein 1 (LRP1), a scavenger receptor mediating the clearance of protease-serpin complexes. Objectives To investigate whether FVIIa-antithrombin is a ligand for LRP1. Methods Binding of FVIIa and pre-formed FVIIa-antithrombin to purified LRP1 Fc-tagged cluster IV (rLRP1-cIV/Fc) and to human and murine macrophages was analyzed. FVIIa clearance was determined in macrophage LRP1 (macLRP1)-deficient mice. Results Solid-phase binding assays showed that FVIIa-antithrombin bound in a specific, dose-dependent and saturable manner to rLRP1-cIV/Fc. Competition experiments with human THP1 macrophages indicated that binding of FVIIa but not of FVIIa-antithrombin was reduced in the presence of annexin-V or anti-tissue factor antibodies, whereas binding of FVIIa-antithrombin but not FVIIa was inhibited by the LRP1-antagonist GST-RAP. Additional experiments revealed binding of both FVIIa and FVIIa-antithrombin to murine control macrophages. In contrast, no binding of FVIIa-antithrombin to macrophages derived from macLRP1-deficient mice could be detected. Clearance of FVIIa-antithrombin but not of active site-blocked FVIIa was delayed 1.5-fold (mean residence time of 3.3 ± 0.1 h versus 2.4 ± 0.2 h) in macLRP1-deficient mice. The circulatory presence of FVIIa was prolonged to a similar extent in macLRP1-deficient mice and in control mice. Conclusions Our data show that FVIIa-antithrombin but not FVIIa is a ligand for LRP1, and that LRP1 contributes to the clearance of FVIIa-antithrombin in vivo.


Asunto(s)
Antitrombinas/metabolismo , Factor VIIa/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas Portadoras/metabolismo , Dominio Catalítico , Línea Celular , Humanos , Ligandos , Macrófagos/metabolismo , Ratones , Unión Proteica , Proteínas Recombinantes/metabolismo , Serpinas/metabolismo , Tromboplastina/metabolismo , Factores de Tiempo
10.
J Thromb Haemost ; 3(3): 552-61, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15748246

RESUMEN

BACKGROUND: Platelet adhesion and aggregation at sites of vascular injury exposed to rapid blood flow require von Willebrand factor (VWF). VWF becomes immobilized by binding to subendothelial components or by a self-association at the interface of soluble and surface-bound VWF. OBJECTIVES: As this self-association has been demonstrated only under shear conditions, our first goal was to determine whether the same interaction could be observed under static conditions. Furthermore, we wanted to identify VWF domain(s) important for this self-association. RESULTS: Biotinylated VWF (b-VWF) interacted dose-dependently and specifically with immobilized VWF in an enzyme-linked immunosorbent assay (ELISA) assay, showing that shear is not necessary to induce the VWF self-association. Whereas anti-VWF monoclonal antibodies (mAbs) had no effect on the self-association, the proteolytic VWF-fragments SpII(1366-2050) and SpIII(1-1365) inhibited the b-VWF-VWF interaction by 70 and 80%, respectively. Moreover, a specific binding of b-VWF to immobilized Sp-fragments was demonstrated. Finally, both biotinylated SpII and SpIII were able to bind specifically to both immobilized SpII and SpIII. Similar results were observed under flow conditions, which confirmed the functional relevance of our ELISA system. CONCLUSION: We have developed an ELISA binding assay in which a specific VWF self-association under static conditions can be demonstrated. Our results suggest a multiple domain interaction between immobilized and soluble VWF.


Asunto(s)
Factor de von Willebrand/química , Factor de von Willebrand/metabolismo , Anticuerpos Monoclonales/farmacología , Biotinilación , Dimerización , Ensayo de Inmunoadsorción Enzimática , Heparina/farmacología , Humanos , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Factor de von Willebrand/inmunología
11.
J Thromb Haemost ; 3(10): 2228-37, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16194200

RESUMEN

BACKGROUND: von Willebrand disease (VWD) is a bleeding disorder caused by the decrease of functional von Willebrand factor (VWF). Low levels of VWF can result from decreased synthesis, impaired secretion, increased clearance or combinations thereof. Several mutations lead to impaired synthesis or secretion of VWF, however, little is known about the survival of VWF in the circulation. OBJECTIVES: To evaluate the effect of several VWF mutations on VWF clearance. PATIENTS/METHODS: The effect of three cysteine-mutations (C1130F, C1149R or C2671Y) on the in vivo survival of VWF was studied in patients carrying these mutations and in a VWF-deficient mice model. RESULTS: In patients carrying these mutations, we observed increased propeptide/mature VWF ratios and rapid disappearance of VWF from the circulation after desmopressin treatment. Detailed analysis of in vivo clearance of recombinant VWF in a VWF-deficient mice model revealed a fourfold increased clearance rate of the mutants. The mutations C1130F, C1149R and C2671Y are each associated with reduced survival of VWF in the circulation. Detailed analysis of the recombinant mutant VWF demonstrated that increased clearance was not due to increased proteolysis by ADAMTS-13. We did not identify functional or structural characteristics that the mutant proteins have in common and could be associated with the phenomenon of increased clearance. CONCLUSIONS: Cysteine-mutations in VWF may result in reduced in vivo survival. The observation that various mutations are associated with increased in vivo clearance may have major implications for the therapeutic strategies that rely on the rise of endogenous VWF after desmopressin administration.


Asunto(s)
Cisteína/genética , Mutación Missense , Enfermedades de von Willebrand/genética , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo , Proteína ADAMTS13 , Animales , Desamino Arginina Vasopresina/farmacología , Humanos , Metabolismo , Metaloendopeptidasas/metabolismo , Ratones , Ratones Noqueados , Proteínas Recombinantes/farmacocinética , Enfermedades de von Willebrand/sangre , Factor de von Willebrand/análisis
12.
Trends Cardiovasc Med ; 10(1): 8-14, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11150722

RESUMEN

Recent reports suggest that the multifunctional receptor low-density lipoprotein receptor-related protein (LRP) may contribute to the regulation of blood coagulation by mechanisms that differ from the simple removal of protease/inhibitor complexes from the circulation. This possibility became apparent from the observation that LRP is involved in down-regulation of Tissue Factor expression at the surface of monocytes and fibroblasts. Furthermore, coagulation Factor VIII and activated Factor IX (Factor IXa) have been identified as proteins that are able to bind to LRP. In the present review, the potential contribution of LRP to the regulation of the coagulation cascade through these novel pathways is discussed, with particular reference to the interaction between LRP and coagulation Factor VIII.


Asunto(s)
Coagulación Sanguínea/fisiología , Factor VIII/metabolismo , Receptores de LDL/sangre , Animales , Interacciones Farmacológicas , Factor VIIIa/metabolismo , Humanos , Ligandos , Factor de von Willebrand/metabolismo
13.
J Thromb Haemost ; 13(5): 815-20, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25728415

RESUMEN

BACKGROUND: von Willebrand factor (VWF) is cleared in a shear stress- and macrophage-dependent manner by LRP1. von Willebrand disease (VWD)-type 2B mutants are endocytosed more efficiently than wild-type (wt)-VWF by macrophages. OBJECTIVE: To investigate if VWD-type 2B mutations in the VWF A1-domain affect LRP1 binding and LRP1-dependent clearance. METHODS: Recombinant Fc-tagged A1 domain (A1-Fc, A2-Fc, A3-Fc) and full-length VWF (wt or mutants thereof) were tested for binding to LRP1 or a recombinant fragment thereof in a static immunosorbent assay. Mutant and wt-VWF were also compared for clearance in mice lacking macrophage LRP1 (macLRP1(-) ) and control mice (macLRP1(+) ). RESULTS: We found that A1-Fc but not A2-Fc or A3-Fc binds dose-dependently to LRP1. Binding of A1-Fc to LRP1 was markedly enhanced by the VWD-type 2B mutation p.V1316M. As expected, full-length wt-VWF was unable to bind LRP1 under static conditions unless ristocetin was added. In contrast, the presence of the p.V1316M or p.R1306Q mutation induced spontaneous binding to LRP1 without the need for ristocetin or shear stress. Both mutants were cleared more rapidly than wt-VWF in control macLRP1(+) mice. Surprisingly, deletion of macrophage LRP1 abrogated the increased clearance of the VWF/p.R1306Q and VWF/p.V1316M mutant. CONCLUSION: The VWF A1-domain contains a binding site for LRP1. Certain VWD-type 2B mutations relieve the need for shear stress to induce LRP1 binding. Enhanced LRP1 binding coincides with a reduced survival of VWF/p.R1306Q and VWF/p.V1316M. Our data provide a rationale for reduced VWF levels in at least some VWD-type 2B patients.


Asunto(s)
Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Mutación , Resistencia al Corte , Estrés Mecánico , Factor de von Willebrand/metabolismo , Animales , Femenino , Masculino , Ratones , Unión Proteica , Factor de von Willebrand/genética
14.
J Thromb Haemost ; 13(10): 1757-67, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26302994

RESUMEN

Circulatory support devices are increasingly being used to overcome cardiac or respiratory failure. Long-term devices are used either as a 'bridge to transplant' to support patients who are unable to wait any longer for a heart transplant, or, more recently, as 'destination therapy' for older patients suffering from end-stage heart failure and who have contraindications to heart transplantation. Short-term support devices for high-risk percutaneous coronary intervention, or as a 'bridge for decision' for patients suffering from refractory cardiogenic shock, have also been developed. The clinical benefit of such assist devices has been demonstrated in several important studies, but, unfortunately, thrombotic and bleeding complications are two major clinical issues in patients requiring these devices. Overcoming these issues is of major importance to allow the safe and broad use of these devices, and to consider them as true alternatives to heart transplantation. The present review focuses on thrombotic and bleeding complications, and describes how the risk of thrombosis and bleeding may vary according to the clinical indication, but also according to the type of device. We describe the current knowledge of the mechanisms underlying the occurrence of these complications, provide some guidance for choosing the most appropriate anticoagulation regimen to prevent their occurrence for each type of device and indication, and provide some recommendations for the management of patients when the complication occurs.


Asunto(s)
Anticoagulantes/uso terapéutico , Insuficiencia Cardíaca/terapia , Corazón Auxiliar/efectos adversos , Hemorragia/prevención & control , Trombosis/prevención & control , Animales , Anticoagulantes/efectos adversos , Coagulación Sanguínea/efectos de los fármacos , Técnicas de Apoyo para la Decisión , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/fisiopatología , Trasplante de Corazón , Hemorragia/sangre , Hemorragia/diagnóstico , Hemorragia/etiología , Hemorragia/mortalidad , Humanos , Selección de Paciente , Valor Predictivo de las Pruebas , Diseño de Prótesis , Medición de Riesgo , Factores de Riesgo , Trombosis/sangre , Trombosis/diagnóstico , Trombosis/etiología , Trombosis/mortalidad , Factores de Tiempo , Resultado del Tratamiento , Función Ventricular Izquierda
15.
J Thromb Haemost ; 11 Suppl 1: 202-11, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23809124

RESUMEN

Quantitative deficiencies in von Willebrand factor (VWF) are associated with abnormal hemostasis that can manifest in bleeding or thrombotic complications. Consequently, many studies have endeavored to elucidate the mechanisms underlying the regulation of VWF plasma levels. This review focuses on the role of VWF clearance pathways. A summary of recent developments are provided, including results from genetic studies, the relationship between glycosylation and VWF clearance, the contribution of increased VWF clearance to the pathogenesis of von Willebrand disease and the identification of VWF clearance receptors. These different studies converge in their conclusion that VWF clearance is a complex phenomenon that involves multiple mechanisms. Deciphering how such different mechanisms coordinate their role in this process is but one of the remaining challenges. Nevertheless, a better insight into the complex clearance pathways of VWF may help us to better understand the clinical implications of aberrant clearance in the pathogenesis of von Willebrand disease and perhaps other disorders as well as aid in developing alternative therapeutic approaches.


Asunto(s)
Factor de von Willebrand/metabolismo , Glicosilación , Humanos
20.
J Thromb Haemost ; 10(12): 2428-37, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23020315

RESUMEN

von Willebrand factor (VWF) is a protein best known from its critical role in hemostasis. Indeed, any dysfunction of VWF is associated with a severe bleeding tendency known as von Willebrand disease (VWD). Since the first description of the disease by Erich von Willebrand in 1926, remarkable progress has been made with regard to our understanding of the pathogenesis of this disease. The cloning of the gene encoding VWF has allowed numerous breakthroughs, and our knowledge of the epidemiology, genetics and molecular basis of VWD has been rapidly expanding since then. These studies have taught us that VWF is rather unique in terms of its multimeric structure and the unusual mechanisms regulating its participation in the hemostatic process. Moreover, it has become increasingly clear that VWF is a more all-round protein than originally thought, given its involvement in several pathologic processes beyond hemostasis. These include angiogenesis, cell proliferation, inflammation, and tumor cell survival. In the present article, an overview of advances concerning the various structural and functional aspects of VWF will be provided.


Asunto(s)
Factor de von Willebrand/fisiología , Apoptosis/fisiología , Proliferación Celular , Humanos , Conformación Proteica , Trombosis/fisiopatología , Factor de von Willebrand/química , Factor de von Willebrand/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA