Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Thromb Haemost ; 15(7): 1285-1294, 2017 07.
Article in English | MEDLINE | ID: mdl-28671350

ABSTRACT

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.


Subject(s)
Inflammation , Venous Thrombosis/metabolism , von Willebrand Factor/metabolism , ADAMTS13 Protein/metabolism , Animals , Blood Platelets/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Glycoproteins/metabolism , Hemostasis , Humans , Leukocytes/cytology , Ligands , Mice , Neutrophils/metabolism , P-Selectin/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , Risk Factors , Weibel-Palade Bodies
2.
Platelets ; 28(5): 518-520, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27885890

ABSTRACT

We present here a 63-year old woman with a long history of immune thrombocytopenia. She was hospitalized for a traumatic intracranial hemorrhage with thrombocytopenia. Following inefficient treatment of four platelet transfusions, immunoglobulins, and corticosteroids, we initiated treatment with a thrombopoietin (TPO) receptor agonist (eltrombopag 25 mg/d) with a good efficacy. Her mother and sister also had chronic thrombocytopenia. Clinical history, hemostasis results, and gene analysis revealed von Willebrand disease (VWD) type 2B with the mutation (c.3946G>A; p.V1316M), which combines a von Willebrand factor defect with severe thrombocytopenia, as well as a thrombocytopathy. The efficacy of TPO receptor agonists appears to counterbalance, at least to some extent, the thrombocytopathy associated with this mutation. As such, the use of TPO receptor agonists could represent an alternative therapeutic approach in cases of VWD type 2B with severe thrombocytopenia.


Subject(s)
Benzoates/administration & dosage , Hydrazines/administration & dosage , Intracranial Hemorrhages/drug therapy , Pyrazoles/administration & dosage , Receptors, Thrombopoietin/agonists , Thrombocytopenia/drug therapy , von Willebrand Disease, Type 2/drug therapy , Amino Acid Substitution , Female , Humans , Intracranial Hemorrhages/complications , Intracranial Hemorrhages/genetics , Middle Aged , Mutation, Missense , Thrombocytopenia/complications , Thrombocytopenia/genetics , von Willebrand Disease, Type 2/complications , von Willebrand Disease, Type 2/genetics , von Willebrand Factor/genetics
3.
J Thromb Haemost ; 14(12): 2458-2470, 2016 12.
Article in English | MEDLINE | ID: mdl-27614059

ABSTRACT

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.


Subject(s)
Antithrombins/metabolism , Factor VIIa/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Receptors, LDL/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Carrier Proteins/metabolism , Catalytic Domain , Cell Line , Humans , Ligands , Macrophages/metabolism , Mice , Protein Binding , Recombinant Proteins/metabolism , Serpins/metabolism , Thromboplastin/metabolism , Time Factors
4.
J Thromb Haemost ; 14(10): 1917-1930, 2016 10.
Article in English | MEDLINE | ID: mdl-27501520

ABSTRACT

Essentials Endothelial injury is thought to be a key event in thrombotic thrombocytopenic purpura (TTP). Endothelial and cardiac damages were assessed in a model of TTP using ADAMTS-13 knockout mice. Damages of cardiac perfusion and function were associated with nitric oxide pathway alteration. Endothelial dysfunction constitutes a critical event in TTP development and cardiac injury. SUMMARY: Background Cardiac alterations represent a major cause of mortality in patients with thrombotic thrombocytopenic purpura (TTP). Endothelial injury remains poorly defined, but seems to be a key initiating event leading to the formation of platelet-rich thrombi in TTP patients. Objectives To assess the changes in endothelial function and the induced cardiac damage in a mouse model of TTP. Patients/methods We used an animal model in which TTP-like symptoms are triggered by injection of 2000 units kg-1 of recombinant von Willebrand factor in ADAMTS-13 knockout mice. Results These mice developed TTP-like symptoms, i.e. severe thrombocytopenia, schistocytosis, and anemia. On day 2, magnetic resonance imaging demonstrated a decrease in left ventricular perfusion associated with alteration of left ventricular ejection fraction, fractional shortening, and cardiac output, suggesting early systolic dysfunction. This was associated with decrease in endothelium-mediated relaxation responses to acetylcholine in mesenteric and coronary arteries, demonstrating severe early endothelial dysfunction. In parallel, we showed decreased cardiac expression of endothelial nitric oxide (NO) synthase and increased expression of antioxidant enzymes, suggesting alteration of the NO pathway. At this time, cardiac immunohistochemistry revealed an increase in the expression of VCAM-1 and E-selectin. Conclusion This study provides evidence that the heart is a sensitive target organ in TTP, and shows, for the first time, strong mesenteric and coronary endothelial dysfunction in an induced-TTP model. The mechanisms incriminated are the occurrence of a pro-oxidant state, and proadhesive and proinflammatory phenotypes. This previously largely unrecognized vascular dysfunction may represent an important contributor to the systemic organ failure occurring in TTP.


Subject(s)
ADAMTS13 Protein/genetics , Endothelium, Vascular/pathology , Purpura, Thrombotic Thrombocytopenic/diagnosis , Animals , Antioxidants/metabolism , Disease Models, Animal , E-Selectin/metabolism , Female , Heart Ventricles/pathology , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/chemistry , Nitric Oxide Synthase Type III/metabolism , Oxidants/metabolism , Perfusion , Phenotype , Purpura, Thrombotic Thrombocytopenic/pathology , Reactive Oxygen Species/metabolism , Recombinant Proteins/pharmacology , Thrombosis/pathology , Vascular Cell Adhesion Molecule-1/metabolism , Ventricular Function, Left , von Willebrand Factor/pharmacology
5.
J Thromb Haemost ; 14(9): 1725-35, 2016 09.
Article in English | MEDLINE | ID: mdl-27344059

ABSTRACT

UNLABELLED: Essentials Von Willebrand disease IIC Miami features high von Willebrand factor (VWF) with reduced function. We aimed to identify and characterize the elusive underlying mutation in the original family. An inframe duplication of VWF exons 9-10 was identified and characterized. The mutation causes a defect in VWF multimerization and decreased VWF clearance from the circulation. SUMMARY: Background A variant of von Willebrand disease (VWD) type 2A, phenotype IIC (VWD2AIIC), is characterized by recessive inheritance, low von Willebrand factor antigen (VWF:Ag), lack of VWF high-molecular-weight multimers, absence of VWF proteolytic fragments and mutations in the VWF propeptide. A family with dominantly inherited VWD2AIIC but markedly elevated VWF:Ag of > 2 U L(-1) was described as VWD type IIC Miami (VWD2AIIC-Miami) in 1993; however, the molecular defect remained elusive. Objectives To identify the molecular mechanism underlying the phenotype of the original VWD2AIIC-Miami. Patients and Methods We studied the original family with VWD2AIIC-Miami phenotypically and by genotyping. The identified mutation was recombinantly expressed and characterized by standard techniques, confocal imaging and in a mouse model, respectively. Results By Multiplex ligation-dependent probe amplification we identified an in-frame duplication of VWF exons 9-10 (c.998_1156dup; p.Glu333_385dup) in all patients. Recombinant mutant (rm)VWF only presented as a dimer. Co-expressed with wild-type VWF, the multimer pattern was indistinguishable from patients' plasma VWF. Immunofluorescence studies indicated retention of rmVWF in unusually large intracellular granules in the endoplasmic reticulum. ADAMTS-13 proteolysis of rmVWF under denaturing conditions was normal; however, an aberrant proteolytic fragment was apparent. A decreased ratio of VWF propeptide to VWF:Ag and a 1-desamino-8-d-arginine vasopressin (DDAVP) test in one patient indicated delayed VWF clearance, which was supported by clearance data after infusion of rmVWF into VWF(-/-) mice. Conclusion The unique phenotype of VWD2 type IIC-Miami results from dominant impairment of multimer assembly, an aberrant structure of mutant mature VWF and reduced clearance in vivo.


Subject(s)
Mutation , von Willebrand Disease, Type 2/genetics , von Willebrand Factor/genetics , Adult , Aged , Animals , Deamino Arginine Vasopressin/chemistry , Endoplasmic Reticulum/metabolism , Female , Genes, Dominant , Genes, Recessive , Genotype , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Phenotype , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , von Willebrand Disease, Type 2/metabolism , von Willebrand Factor/metabolism
6.
J Thromb Haemost ; 13(5): 815-20, 2015 May.
Article in English | MEDLINE | ID: mdl-25728415

ABSTRACT

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.


Subject(s)
Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Mutation , Shear Strength , Stress, Mechanical , von Willebrand Factor/metabolism , Animals , Female , Male , Mice , Protein Binding , von Willebrand Factor/genetics
7.
J Thromb Haemost ; 11 Suppl 1: 202-11, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23809124

ABSTRACT

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.


Subject(s)
von Willebrand Factor/metabolism , Glycosylation , Humans
8.
J Thromb Haemost ; 10(12): 2428-37, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23020315

ABSTRACT

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.


Subject(s)
von Willebrand Factor/physiology , Apoptosis/physiology , Cell Proliferation , Humans , Protein Conformation , Thrombosis/physiopathology , von Willebrand Factor/chemistry , von Willebrand Factor/genetics
11.
Haemophilia ; 16 Suppl 5: 194-9, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20590881

ABSTRACT

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.


Subject(s)
Factor VIII/chemistry , Polysaccharides/chemistry , von Willebrand Factor/chemistry , Amino Acid Sequence , Factor VIII/biosynthesis , Glycosylation , Humans , Protein Structure, Tertiary , von Willebrand Diseases/drug therapy , von Willebrand Factor/biosynthesis
13.
J Thromb Haemost ; 8(1): 173-84, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19874458

ABSTRACT

BACKGROUND: We previously described a model of laser-induced thrombosis in mesenteric arterioles with superficial and deep levels of injury producing a transient thrombus resolving within 2 min and a larger almost occlusive thrombus, respectively. Both types of lesion were sensitive to platelet GPIIb-IIIa and P2Y(12) inhibition, whereas only deep injuries were sensitive to thrombin blockade. OBJECTIVE: The aim of the present study was to use histologic methods and electron and intravital microscopy to characterize the lesions and thrombi and to extend our knowledge of the sensitivity of this model to genetic and pharmacologic inhibition. RESULTS: A superficial injury was found to detach the endothelial cells and expose a collagen III- and IV-rich subendothelium where platelets could adhere. Tissue factor and fibrin were not detected. Deeper penetration of the external elastic lamina occurred in deep injuries, with exposure of collagen I, III and IV. Here the thrombus was composed of platelets exhibiting a decreasing gradient of degranulation from the deepest lesion area to the surface. Fibrin was found close to the most activated platelets. Consistently, glycoprotein VI (GPVI)-collagen and GPIb-von Willebrand factor (VWF) interactions were found to be critical in superficial injuries. After deep lesion, thrombus formation was modestly reduced in GPVI-immunodepleted mice and still strongly inhibited in VWF(-/-) mice. Combined hirudin infusion and GPVI depletion further inhibited thrombosis after deep injury. CONCLUSIONS: This study confirms the feasibility of inducing arterial thrombosis with distinct levels of severity and establishes the central roles of collagen and VWF in thrombus formation after superficial injury. Collagen, VWF and thrombin all appear to contribute to thrombosis after deep arterial lesion.


Subject(s)
Blood Platelets/ultrastructure , Endothelium, Vascular/ultrastructure , Mesenteric Arteries/ultrastructure , Mesenteric Vascular Occlusion/pathology , Platelet Adhesiveness , Thrombosis/pathology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Collagen Type I/metabolism , Collagen Type III/metabolism , Collagen Type IV/metabolism , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/injuries , Endothelium, Vascular/metabolism , Feasibility Studies , Fibrin/metabolism , Fibrinolytic Agents/administration & dosage , Hirudins/administration & dosage , Injections, Subcutaneous , Lasers, Gas , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/injuries , Mesenteric Arteries/metabolism , Mesenteric Vascular Occlusion/blood , Mesenteric Vascular Occlusion/etiology , Mesenteric Vascular Occlusion/prevention & control , Mice , Mice, Knockout , Platelet Adhesiveness/drug effects , Platelet Membrane Glycoproteins/deficiency , Platelet Membrane Glycoproteins/metabolism , Severity of Illness Index , Thrombosis/blood , Thrombosis/etiology , Thrombosis/prevention & control , Time Factors , von Willebrand Factor/genetics , von Willebrand Factor/metabolism
14.
J Thromb Haemost ; 7 Suppl 1: 61-4, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19630770

ABSTRACT

von Willebrand disease (VWD), caused by quantitative or qualitative abnormalities in von Willebrand factor (VWF) is considered the most common inherited bleeding disorder in humans. Mild and severe quantitative defects in VWF cause VWD types 1 and 3 respectively, whereas qualitative abnormalities induce VWD type 2. VWD has also been diagnosed in a number of animal species such as dogs, pigs, cats and horses, as a result of naturally occurring mutations. More recently, murine models have drawn a great deal of attention. Their small size along with their well-defined genetic background makes them ideal tools to study the in vivo function of VWF. The most commonly used model is the VWF-deficient mouse engineered through homologous recombination. However, models resulting from changes in modifier genes indirectly affecting VWF have also been described. These various models have proven very useful in elucidating some aspects of VWF biology not easily addressed through in vitro approaches.


Subject(s)
Disease Models, Animal , von Willebrand Diseases , Animals , Mice , Recombination, Genetic , von Willebrand Factor/genetics
15.
Haemophilia ; 15(5): 1058-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19500169

ABSTRACT

von Willebrand's disease (VWD) type 3 is a rare but severe autosomal-recessive inherited bleeding disorder with a prevalence higher in certain locations where consanguineous marriages are relatively frequent. The genetic defects causing recessive type 3 VWD in 10 unrelated families from Iran have been investigated and the genetic heterogeneity among these patients was evaluated. All exons and their flanking regions of von Willebrand factor gene were amplified by PCR and sequenced using specific primers. Eight patients were fully characterized at the molecular level. Six different gene alterations were identified. All the mutations caused null alleles, three being nonsense mutations (Q104X, Q793X and E1981X), two possible splice site mutations (2443-1G>C and 1110-1G>A) and one small deletion (3237delA). Three of them have not been described previously. Most patients were born from consanguineous marriages and all were homozygous for their mutations. The results confirm that molecular defects in type 3 VWD are heterogeneous with mutations arising randomly within the entire gene.


Subject(s)
Codon, Nonsense/genetics , Exons/genetics , Genes, Recessive/genetics , von Willebrand Disease, Type 3/genetics , Adult , Female , Genetic Heterogeneity , Genetic Testing , Genotype , Humans , Iran/epidemiology , Male , Middle Aged , Molecular Sequence Data , Pedigree , Phenotype , Polymerase Chain Reaction , Prevalence , Sequence Analysis, DNA , Severity of Illness Index , Young Adult , von Willebrand Disease, Type 3/epidemiology
16.
J Thromb Haemost ; 7(5): 843-50, 2009 May.
Article in English | MEDLINE | ID: mdl-19187073

ABSTRACT

SUMMARY BACKGROUND: During sepsis, von Willebrand factor (VWF) is abundantly secreted; the main mechanism regulating its size involves specific proteolysis by the metalloprotease ADAMTS-13. OBJECTIVES: To determine whether ADAMTS-13 consumption due to its binding to, and/or cleavage, of VWF contributes to its decrease during sepsis and whether abrogating or enhancing ADAMTS-13 activity influences sepsis outcome. METHODS: ADAMTS-13 activity was evaluated in a model of sepsis induced by cecum ligature and puncture (CLP) in wild-type and Vwf(-/-) mice. Sepsis outcome was studied in those mice and in Adamts-13(-/-) mice. Finally, survival was studied in wild-type mice injected hydrodynamically with the human ADAMTS-13 gene. RESULTS: In wild-type mice, CLP-induced sepsis elicited a significant ADAMTS-13 decrease, and a strong negative correlation existed between VWF and ADAMTS-13. In Vwf(-/-) mice, CLP also induced severe sepsis, but ADAMTS-13 was not significantly diminished. Notably, Vwf(-/-) mice lived significantly longer than wild-type mice. In contrast, Adamts-13(-/-) mice and wild-type mice were comparable with regard to thrombocytopenia, VWF concentrations, absence of thrombi, and survival. Hydrodynamic hADAMTS-13 gene transfer with the pLIVE expression vector resulted in high and stable ADAMTS13 activity in CLP mice; however, no impact on survival was observed. CONCLUSIONS: VWF secretion is a major determinant of ADAMTS-13 decrease in the CLP model, and plays an important role in sepsis-induced mortality, but the complete absence of its regulating protease, ADAMTS-13, had no detectable impact in this sepsis model. Furthermore, increasing ADAMTS-13 activity had no impact on survival.


Subject(s)
Cecum/pathology , Metalloendopeptidases/metabolism , Sepsis/metabolism , von Willebrand Factor/physiology , ADAMTS13 Protein , Animals , Disease Models, Animal , Kidney/pathology , Liver/pathology , Male , Mice , Mice, Knockout , von Willebrand Factor/genetics
17.
Hamostaseologie ; 29(1): 17-8, 20, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19151840

ABSTRACT

Von Willebrand factor (VWF) structure-function relationship has been studied only through in vitro approaches. The VWF-deficient mouse model has been extremely useful to examine the in vivo function of VWF but does not allow a more subtle analysis of the relative importance of its different domains. However, considering the large size of VWF and its capacity to interact with various ligands in order to support platelet adhesion and aggregation, the necessity to evaluate independently these interactions appeared increasingly crucial. A recently developed technique, known as hydrodynamic injection, which allows transient expression of a transgene by mouse hepatocytes, proved very useful in this regard. Indeed, transient expression of various VWF mutants in VWF-deficient mice contributed to improve our knowledge about the role of VWF interaction with subendothelial collagens and with platelets receptors in VWF roles in haemostasis and thrombosis. These findings can provide new leads in the development of anti-thrombotic therapies.


Subject(s)
von Willebrand Factor/chemistry , von Willebrand Factor/metabolism , Animals , Mice , Mice, Knockout , Models, Animal , Mutation , Thrombosis/blood , Thrombosis/genetics , von Willebrand Diseases/blood , von Willebrand Diseases/genetics , von Willebrand Factor/genetics
18.
J Thromb Haemost ; 5(9): 1956-62, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17723135

ABSTRACT

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.


Subject(s)
Osteoprotegerin/metabolism , von Willebrand Factor/metabolism , Amino Acid Sequence , Cells, Cultured , Humans , Osteoprotegerin/chemistry , Protein Binding , Recombinant Proteins/metabolism , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL
...