Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
1.
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
2.
J Thromb Haemost ; 13(11): 2063-75, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26391536

ABSTRACT

BACKGROUND: Recently, conformational activation of ADAMTS-13 was identified. This mechanism showed the evolution from a condensed conformation, in which the proximal MDTCS and distal T2-CUB2 domains are in close contact with each other, to an activated, open structure due to binding with von Willebrand factor (VWF). OBJECTIVES: Identification of cryptic epitope/exosite exposure after conformational activation and of sites of flexibility in ADAMTS-13. METHODS: The activating effect of 25 anti-T2-CUB2 antibodies was studied in the FRETS-VWF73 and the vortex assay. Cryptic epitope/exosite exposure was determined with ELISA and VWF binding assay. The molecular basis for flexibility was hypothesized through rapid automatic detection and alignment of repeats (RADAR) analysis, tested with ELISA using deletion variants and visualized using electron microscopy. RESULTS: Eleven activating anti-ADAMTS-13 antibodies, directed against the T5-CUB2 domains, were identified in the FRETS-VWF73 assay. RADAR analysis identified three linker regions in the distal domains. Interestingly, identification of an antibody recognizing a cryptic epitope in the metalloprotease domain confirmed the contribution of these linker regions to conformational activation of the enzyme. The proof of flexibility around both the T2 and metalloprotease domains, as shown by by electron microscopy, further supported this contribution. In addition, cryptic epitope exposure was identified in the distal domains, because activating anti-T2-CUB2 antibodies increased the binding to folded VWF up to ~3-fold. CONCLUSION: Conformational activation of ADAMTS-13 leads to cryptic epitope/exosite exposure in both proximal and distal domains, subsequently inducing increased activity. Furthermore, three linker regions in the distal domains are responsible for flexibility and enable the interaction between the proximal and the T8-CUB2 domains.


Subject(s)
ADAM Proteins/chemistry , ADAM Proteins/immunology , ADAM Proteins/metabolism , ADAM Proteins/ultrastructure , ADAMTS13 Protein , Allosteric Regulation , Allosteric Site , Amino Acid Sequence , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions , Catalysis , Consensus Sequence , Enzyme Activation , Epitopes/chemistry , Epitopes/immunology , Humans , Microscopy, Electron , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Folding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Sequence Alignment , Sequence Homology, Amino Acid , Thrombospondin 1/chemistry , von Willebrand Factor/metabolism
3.
J Thromb Haemost ; 11(8): 1511-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23773695

ABSTRACT

BACKGROUND: Most ADAMTS13 assays use non-physiological conditions (low ionic strength, low pH, barium chloride), are subject to interference from plasma proteins, hemoglobin and bilirubin, and have limited sensitivity, especially for inhibitors. OBJECTIVES: We addressed these constraints by designing a substrate that can be used in undiluted plasma. METHODS: A polypeptide was expressed in E. coli that corresponds to von Willebrand factor Gln(1599) -Arg(1668) , with mutations N1610C and K1617R and an N-terminal Gly. Substrate FRETS-rVWF71 was prepared by modifying Cys(1610) with DyLight 633 (abs 638 nm, em 658 nm) and the N-terminus with IRDye QC-1 (abs 500-800 nm). Assays were performed at pH 7.4 in 150 mm NaCl, 10 mm CaCl2 . RESULTS: Serum and plasma anticoagulated with citrate or heparin had equivalent ADAMTS13 activity with FRETS-rVWF71. Neither bilirubin (≤ 20 mg dL(-1) ) nor hemoglobin (≤ 20 g L(-1) ) interfered with product detection. Assays with FRETS-rVWF71 and FRETS-VWF73 gave similar results (R(2 ) = 0.95) for plasma from 80 subjects with thrombotic microangiopathy, 22 subjects with other causes of thrombocytopenia, and 20 healthy controls. The limit of detection with FRETS-rVWF71 for ADAMTS13 activity was ≤ 0.3%. Inhibitor assays with FRETS-rVWF71 gave titers ~2.5-fold higher than with FRETS-VWF73 and clearly distinguished patients with and without inhibitors. CONCLUSIONS: FRETS-rVWF71 is suitable for ADAMTS13 assays in minimally diluted plasma or serum without interference from proteins, bilirubin or free hemoglobin in plasma. Optimized detection of ADAMTS13 inhibitors will facilitate the monitoring of antibody responses during the treatment of thrombotic thrombocytopenic purpura.


Subject(s)
ADAM Proteins/analysis , Purpura, Thrombotic Thrombocytopenic/diagnosis , ADAMTS13 Protein , Anticoagulants/chemistry , Case-Control Studies , Female , Fluorescent Dyes/chemistry , Humans , Male , Mutation , Peptides/chemistry , Plasmids/metabolism , Purpura, Thrombotic Thrombocytopenic/blood , Recombinant Fusion Proteins/metabolism , Reproducibility of Results , Sensitivity and Specificity , Substrate Specificity , Thrombocytopenia/blood , Thrombocytopenia/diagnosis , Thrombotic Microangiopathies/blood , Thrombotic Microangiopathies/diagnosis , von Willebrand Factor/chemistry
4.
J Thromb Haemost ; 8(6): 1273-80, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20088930

ABSTRACT

BACKGROUND: von Willebrand disease (VWD) Vicenza is characterized by low plasma von Willebrand factor (VWF) levels, the presence of ultra-large (UL) VWF multimers and less prominent satellite bands on multimer gels, and the heterozygous amino acid substitution R1205H in the VWF gene. The pathogenesis of VWD Vicenza has been elusive. Accelerated clearance is implicated as a cause of low VWF level. OBJECTIVES: We addressed the question, whether the presence of ultra-large multimers is a cause, or a result of accelerated VWF clearance, or whether it is an unrelated phenomenon. PATIENTS/METHODS: We studied the detailed phenotype of three Hungarian patients with VWD Vicenza, expressed the mutant VWF-R1205H in 293T cells and developed a mathematical model to simulate VWF synthesis and catabolism. RESULTS: We found that the half-life of VWF after DDAVP was approximately one-tenth of that after the administration of Haemate P, a source of exogenous wild-type (WT) VWF (0.81 + or - 0.2 vs. 7.25 + or - 2.38 h). An analysis of recombinant mutant VWF-R1205H showed that the biosynthesis and multimer structure of WT and mutant VWF were indistinguishable. A mathematical model of the complex interplay of VWF synthesis, clearance and cleavage showed that decreasing VWF half-life to one-tenth of normal reproduced all features of VWD Vicenza including low VWF level, ultra-large multimers and a decrease of satellite band intensity. CONCLUSION: We conclude that accelerated clearance alone may explain all features of VWD Vicenza.


Subject(s)
von Willebrand Diseases/metabolism , Amino Acid Substitution , Cell Line , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Female , Humans , Male , Pedigree , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism
5.
J Thromb Haemost ; 7(12): 2088-95, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19765212

ABSTRACT

BACKGROUND: ADAMTS-13 proteolytic activity is controlled by the conformation of its substrate, von Willebrand factor (VWF), and changes in the secondary structure of VWF are essential for efficient cleavage. Substrate recognition is mediated through several non-catalytic domains in ADAMTS-13 distant from the active site. OBJECTIVES: We hypothesized that not all binding sites for ADAMTS-13 in VWF are cryptic and analyzed binding of native VWF to ADAMTS-13. METHODS: Immunoprecipiation of VWF-ADAMTS-13 complexes using anti-VWF antibodies and magnetic beads was used. Binding was assessed by Western blotting and immunosorbent assays. RESULTS: Co-immunoprecipitation demonstrated that ADAMTS-13 binds to native multimeric VWF (K(d) of 79 +/- 11 nmol L(-1)) with no measurable proteolysis. Upon shear-induced unfolding of VWF, binding increased 3-fold and VWF was cleaved. Binding to native VWF was saturable, time dependent, reversible and did not vary with ionic strength (I of 50-200). Moreover, results with ADAMTS-13 deletion mutants indicated that binding to native VWF is mediated through domains distal to the ADAMTS-13 spacer, probably thrombospondin-1 repeats. Interestingly, this interaction occurs in normal human plasma with an ADAMTS-13 to VWF stoichiometry of 0.0040 +/- 0.0004 (mean +/- SEM, n = 10). CONCLUSIONS: ADAMTS-13 binds to circulating VWF and may therefore be incorporated into a platelet-rich thrombus, where it can immediately cleave VWF that is unfolded by fluid shear stress.


Subject(s)
ADAM Proteins/metabolism , von Willebrand Factor/metabolism , ADAM Proteins/chemistry , ADAMTS13 Protein , Binding Sites , Blood Circulation , Blotting, Western , Humans , Immunoprecipitation , Protein Binding , Protein Denaturation , Stress, Mechanical , von Willebrand Factor/chemistry
6.
J Thromb Haemost ; 7 Suppl 1: 24-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19630761

ABSTRACT

During its life history, von Willebrand factor (VWF) experiences a remarkable sequence of conformational changes that are triggered by differences in pH between the endoplasmic reticulum (ER), Golgi and extracellular environments. VWF subunits dimerize in the ER and assemble into disulfide-linked multimers in the trans-Golgi, which lacks known chaperones and has an acidic pH that inhibits disulfide rearrangement. VWF has circumvented these problems by evolving N-terminal domains that function as an oxidoreductase at the low pH of the Golgi. VWF multimers also condense into tightly packed, tubular arrays for storage in the Weibel-Palade bodies of endothelial cells. Like multimer assembly, tubular packing depends on low pH and Ca2+. Upon secretion, exposure to the neutral pH of the extracellular environment allows enormous VWF multimers to uncoil without tangling, which is crucial for hemostasis. Recent studies have identified some of the biochemical and structural properties that underlie these self-organizing behaviors.


Subject(s)
von Willebrand Factor/metabolism , Disulfides , Humans , Hydrogen-Ion Concentration , Protein Conformation , Protein Multimerization , Protein Transport , von Willebrand Factor/chemistry
7.
Haemophilia ; 14(2): 171-232, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18315614

ABSTRACT

von Willebrand disease (VWD) is a commonly encountered inherited bleeding disorder affecting both males and females, causing mucous membrane and skin bleeding symptoms, and bleeding with surgical or other haemostatic challenges. VWD may be disproportionately symptomatic in women of child-bearing age. It may also occur less frequently as an acquired disorder (acquired von Willebrand syndrome). VWD is caused by deficiency or dysfunction of von Willebrand factor (VWF), a plasma protein that mediates platelet haemostatic function and stabilizes blood coagulation factor VIII. The pathophysiology, classification, diagnosis and management of VWD are relatively complex, but understanding them is important for proper diagnosis and management of patients with VWD. These evidence-based guidelines for diagnosis and management of VWD from the National Heart, Lung, and Blood Institute (NHLBI) Expert Panel (USA) review relevant publications, summarize current understanding of VWD pathophysiology and classification, and present consensus diagnostic and management recommendations based on analysis of the literature and expert opinion. They also suggest an approach for clinical and laboratory evaluation of individuals with bleeding symptoms, history of bleeding or conditions associated with increased bleeding risk. This document summarizes needs for further research in VWF, VWD and bleeding disorders, including clinical research to obtain more objective information about bleeding symptoms, advancements in diagnostic and therapeutic tools, and enhancement in the education and training of clinicians and scientists in bleeding and thrombotic disorders. The NHLBI Web site (http://www.nhlbi.nih.gov/guidelines/vwd) has a more detailed document, a synopsis of these recommendations, and patient education information.


Subject(s)
von Willebrand Diseases/diagnosis , von Willebrand Diseases/drug therapy , Antifibrinolytic Agents/therapeutic use , Deamino Arginine Vasopressin/therapeutic use , Factor VIII/analysis , Female , Genetic Therapy/methods , Hemostatics/therapeutic use , Humans , Male , Pregnancy , von Willebrand Factor/administration & dosage , von Willebrand Factor/analysis
8.
J Thromb Haemost ; 4(10): 2103-14, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16889557

ABSTRACT

von Willebrand disease (VWD) is a bleeding disorder caused by inherited defects in the concentration, structure, or function of von Willebrand factor (VWF). VWD is classified into three primary categories. Type 1 includes partial quantitative deficiency, type 2 includes qualitative defects, and type 3 includes virtually complete deficiency of VWF. VWD type 2 is divided into four secondary categories. Type 2A includes variants with decreased platelet adhesion caused by selective deficiency of high-molecular-weight VWF multimers. Type 2B includes variants with increased affinity for platelet glycoprotein Ib. Type 2M includes variants with markedly defective platelet adhesion despite a relatively normal size distribution of VWF multimers. Type 2N includes variants with markedly decreased affinity for factor VIII. These six categories of VWD correlate with important clinical features and therapeutic requirements. Some VWF gene mutations, alone or in combination, have complex effects and give rise to mixed VWD phenotypes. Certain VWD types, especially type 1 and type 2A, encompass several pathophysiologic mechanisms that sometimes can be distinguished by appropriate laboratory studies. The clinical significance of this heterogeneity is under investigation, which may support further subdivision of VWD type 1 or type 2A in the future.


Subject(s)
von Willebrand Diseases/blood , von Willebrand Diseases/physiopathology , ADAM Proteins/physiology , ADAMTS13 Protein , Humans , Models, Biological , Phenotype , Protein Structure, Tertiary , von Willebrand Diseases/classification , von Willebrand Diseases/diagnosis , von Willebrand Factor/metabolism
9.
J Thromb Haemost ; 3(8): 1702-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16102036

ABSTRACT

Everyone experiences minor bleeding and clotting, and many illnesses feature extremes of hemorrhage or thrombosis. Recent advances have illuminated the ways in which von Willebrand factor (VWF) contributes to both kinds of hemostatic emergency, whether mundane or life threatening, often through disturbances in VWF synthesis or catabolism. von Willebrand factor multimer assembly depends on the ability of the propeptide to promote disulfide bond formation in the Golgi, possibly by acting as a pH-sensitive oxidoreductase. Once secreted into the blood, multimers are subject to competing processes of clearance and of proteolysis by ADAMTS-13. Defects in the secretion or intravascular clearance of VWF can cause exceptionally severe forms of von Willebrand disease (VWD) type 1. Defects in the assembly of VWF multimers, or exaggerated proteolytic degradation by ADAMTS-13, can cause VWD type 2A and contribute to VWD type 2B. Conversely, defects in the feedback proteolysis of VWF by ADAMTS-13 can cause thrombotic thrombocytopenic purpura (TTP). The pathophysiologic importance of VWF is not limited to the dramatic phenotypes of VWD and TTP. In fact, VWF level also correlates with thrombosis risk and inversely with bleeding risk within the apparently healthy population. More research is needed to understand how VWF function is regulated, and to enable physicians to use this knowledge for the benefit of their patients.


Subject(s)
von Willebrand Factor/chemistry , von Willebrand Factor/physiology , ADAM Proteins/metabolism , ADAMTS13 Protein , Animals , Disulfides , Golgi Apparatus/metabolism , Hemorrhage , Hemostasis , Humans , Hydrogen-Ion Concentration , Kinetics , Models, Biological , Models, Genetic , Mutation , Phenotype , Purpura, Thrombotic Thrombocytopenic/blood , Risk , Thrombosis
12.
Blood ; 98(10): 2973-9, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698279

ABSTRACT

Some families affected by von Willebrand disease type 1 show high penetrance with exceptionally low von Willebrand factor (VWF) levels. Previously, a mutation associated with this dominant phenotype, Cys1149Arg, was found to decrease the secretion of coexpressed normal VWF, and the mutation was proposed to cause intracellular retention of pro-VWF heterodimers. To demonstrate heterodimer formation, a model was developed in which subunits could be distinguished immunologically and by size. Recombinant VWF lacking domain A1 (dA1), A3 (dA3), or both (dA13) was secreted efficiently as a full range of multimers. Cotransfection of Cys1149Arg and dA13 resulted in the secretion of multimeric VWF containing about 250 kd (Cys1149Arg) and about 210 kd (dA13). Cell lysates contained pro-VWF forms of Cys1149Arg and dA13. Immunoprecipitation with an antidomain A1 antibody recovered both subunits in heterodimers, and subunit ratios were consistent with random dimerization. Similar results were obtained for cotransfection of Cys1149Arg and dA1. Normal VWF has a Cys1149-Cys1169 intrachain bond. When cotransfected with normal VWF, Cys1149Arg or the double mutant Cys1149Arg+Cys1169Ser caused a similar decrease in VWF secretion, suggesting that an unpaired Cys1169 does not explain the intracellular retention of Cys1149Arg. VWF Cys1149Arg was not secreted from BHK cells but was degraded intracellularly within about 4 hours, and the proteasome inhibitor lactacystin delayed its clearance more than 16 hours. Thus, dominant von Willebrand disease type 1 may be caused by heterodimerization of mutant and normal subunits in the endoplasmic reticulum followed by proteasomal degradation in the cytoplasm. A similar dominant negative mechanism could cause quantitative deficiencies of other multisubunit proteins.


Subject(s)
Amino Acid Substitution , Genes, Dominant , Mutation, Missense , von Willebrand Diseases/genetics , von Willebrand Factor/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cricetinae , Cysteine/chemistry , Cysteine Endopeptidases/metabolism , Cystine/chemistry , Dimerization , Endoplasmic Reticulum/metabolism , Humans , Intracellular Fluid/metabolism , Mesocricetus , Models, Genetic , Multienzyme Complexes/metabolism , Point Mutation , Proteasome Endopeptidase Complex , Protein Structure, Tertiary , Sequence Deletion , Transfection , von Willebrand Diseases/classification , von Willebrand Diseases/metabolism , von Willebrand Factor/chemistry , von Willebrand Factor/metabolism
14.
J Biol Chem ; 276(44): 41059-63, 2001 Nov 02.
Article in English | MEDLINE | ID: mdl-11557746

ABSTRACT

Thrombotic thrombocytopenic purpura is associated with acquired or congenital deficiency of a plasma von Willebrand factor-cleaving protease (VWFCP). Based on partial amino acid sequence, VWFCP was identified recently as a new member of the ADAMTS family of metalloproteases and designated ADAMTS13. The 4.6-kilobase pair cDNA sequence for VWFCP has now been determined. By Northern blotting, full-length VWFCP mRNA was detected only in liver. VWFCP consists of 1427 amino acid residues and has a signal peptide, a short propeptide terminating in the sequence RQRR, a reprolysin-like metalloprotease domain, a disintegrin-like domain, a thrombospondin-1 repeat, a Cys-rich domain, an ADAMTS spacer, seven additional thrombospondin-1 repeats, and two CUB domains. VWFCP apparently is made as a zymogen that requires proteolytic activation, possibly by furin intracellularly. Sites for Zn(2+) and Ca(2+) ions are conserved in the protease domain. The Cys-rich domain contains an RGDS sequence that could mediate integrin-dependent binding to platelets or other cells. Alternative splicing gives rise to at least seven potential variants that truncate the protein at different positions after the protease domain. Alternative splicing may have functional significance, producing proteins with distinct abilities to interact with cofactors, connective tissue, platelets, and von Willebrand factor.


Subject(s)
Metalloendopeptidases/chemistry , Purpura, Thrombotic Thrombocytopenic/enzymology , Amino Acid Sequence , Base Sequence , Blotting, Northern , Cloning, Molecular , DNA, Complementary , Humans , Liver/enzymology , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Molecular Sequence Data , Protein Conformation , RNA, Messenger/isolation & purification , Sequence Homology, Amino Acid
15.
Thromb Haemost ; 85(5): 929-31, 2001 May.
Article in English | MEDLINE | ID: mdl-11372690

ABSTRACT

Examination of the entire von Willebrand factor (VWF) gene for mutations, particularly in types 1 and 3 von Willebrand disease (VWD) is becoming more widely practised. The sequence of the entire VWF gene will soon be compiled as a single sequence. For these reasons, a clearly defined nomenclature to use for numbering the VWF nucleotide and amino acid sequence is required. The following recommendations are made for VWF numbering. VWF cDNA nucleotide sequence should be numbered from the A of the initiator ATG as the +1 position. Genomic DNA should be prefixed with a "g" and also numbered from this position. Amino acid (aa) numbering should be from the initiator methionine as the +1 position with sequential numbering of aa throughout VWF. To avoid confusion with previously used numbering schemes for mature VWF, which started from serine 764 of pre-pro VWF, the use of the single letter amino acid code is recommended.


Subject(s)
Terminology as Topic , von Willebrand Factor/genetics , Humans , Mutation , Polymorphism, Genetic
16.
J Biol Chem ; 276(9): 6161-8, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11024046

ABSTRACT

The glycoprotein (GP) Ib-IX complex is a platelet surface receptor that binds thrombin as one of its ligands, although the biological significance of thrombin interaction remains unclear. In this study we have used several approaches to investigate the GPIb alpha-thrombin interaction in more detail and to study its effect on the thrombin-induced elaboration of fibrin. We found that both glycocalicin and the amino-terminal fragment of GPIb alpha reduced the release of fibrinopeptide A from fibrinogen by about 50% by a noncompetitive allosteric mechanism. Similarly, GPIb alpha caused in thrombin an allosteric reduction in the rate of turnover of the small peptide substrate d-Phe-Pro-Arg-pNA. The K(d) for the glycocalicin-thrombin interaction was 1 microm at physiological ionic strength but was highly salt-dependent, decreasing to 0.19 microm at 100 mm NaCl (Gamma(salt) = -4.2). The salt dependence was characteristic of other thrombin ligands that bind to exosite II of this enzyme, and we confirmed this as the GPIb alpha-binding site on thrombin by using thrombin mutants and by competition binding studies. R68E or R70E mutations in exosite I of thrombin had little effect on its interaction with GPIb alpha. Both the allosteric inhibition of fibrinogen turnover caused by GPIb alpha binding to these mutants, and the K(d) values for their interactions with GPIb alpha were similar to those of wild-type thrombin. In contrast, R89E and K248E mutations in exosite II of thrombin markedly increased the K(d) values for the interactions of these thrombin mutants with GPIb alpha by 10- and 25-fold, respectively. Finally, we demonstrated that low molecular weight heparin (which binds to thrombin exosite II) but not hirugen (residues 54-65 of hirudin, which binds to exosite I of thrombin) inhibited thrombin binding to GPIb alpha. These data demonstrate that GPIb alpha binds to thrombin exosite II and in so doing causes a conformational change in the active site of thrombin by an allosteric mechanism that alters the accessibility of both its natural substrate, fibrinogen, and the small peptidyl substrate d-Phe-Pro-Arg-pNA.


Subject(s)
Hirudins/analogs & derivatives , Platelet Glycoprotein GPIb-IX Complex/metabolism , Thrombin/metabolism , Fibrinopeptide A/metabolism , Heparin, Low-Molecular-Weight/pharmacology , Hirudins/pharmacology , Humans , Peptide Fragments/pharmacology , Recombinant Proteins/metabolism , Sodium Chloride/pharmacology , Thrombin/chemistry
17.
EMBO J ; 19(21): 5650-60, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11060016

ABSTRACT

The serine proteinase alpha-thrombin plays a pivotal role in the regulation of blood fluidity, and therefore constitutes a primary target in the treatment of various haemostatic disorders. Haemadin is a slow tight- binding thrombin inhibitor from the land-living leech Haemadipsa sylvestris. Here we present the 3.1 A crystal structure of the human alpha-thrombin- haemadin complex. The N-terminal segment of haemadin binds to the active site of thrombin, forming a parallel beta-strand with residues Ser214-Gly216 of the proteinase. This mode of binding is similar to that observed in another leech-derived inhibitor, hirudin. In contrast to hirudin, however, the markedly acidic C-terminal peptide of haemadin does not bind the fibrinogen-recognition exosite, but interacts with the heparin-binding exosite of thrombin. Thus, haemadin binds to thrombin according to a novel mechanism, despite an overall structural similarity with hirudin. Haemadin inhibits both free and thrombomodulin-bound alpha-thrombin, but not intermediate activation forms such as meizothrombin. This specific anticoagulant ability of haemadin makes it an ideal candidate for an antithrombotic agent, as well as a starting point for the design of novel antithrombotics.


Subject(s)
Invertebrate Hormones/chemistry , Invertebrate Hormones/pharmacology , Thrombin/antagonists & inhibitors , Thrombin/chemistry , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Hirudins/metabolism , Hirudins/pharmacology , Humans , In Vitro Techniques , Invertebrate Hormones/metabolism , Leeches/chemistry , Macromolecular Substances , Models, Molecular , Molecular Sequence Data , Protein Conformation , Sequence Homology, Amino Acid , Solutions , Thrombin/metabolism
19.
Thromb Haemost ; 84(2): 345-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10959711

ABSTRACT

The acquired von Willebrand syndrome (AvWS) is a rare bleeding disorder with laboratory findings similar to those of congenital von Willebrand disease (vWD). Despite the numerous cases reported in the literature until 1999 (n = 266), large studies on AvWS are not available. Moreover, diagnosis of AvWS has been difficult and treatment empirical. These considerations prompted us to organize an international registry. A questionnaire, devised to collect specific information on AvWS, was sent to all the members of the International Society on Thrombosis and Haemostasis (ISTH), who were invited to respond if they had diagnosed cases with the AvWS cases. 156 members answered the questionnaire and 54 of them sent information on 211 AvWS cases from 50 centers. Data were compared with those already published in the literature and 25 cases already described or not correctly diagnosed were excluded. The 186 AvWS cases that qualified for the registry were associated with lymphoproliferative (48%) and myeloproliferative disorders (15%), neoplasia (5%), immunological (2%), cardiovascular (21%) and miscellaneous disorders (9%). Ristocetin cofactor activity (vWF:RCo) or collagen binding activity (vWF:CBA) were usually low in AvWS (median values 20 U/dL, range 3-150), while factor VIII coagulant activity was sometimes normal (median 25 U/dL, range 3-191). FVIII/vWF inhibiting activities were present in only a minority of cases (16%). Bleeding episodes in AvWS were mostly of mucocutaneous type (68%) and were managed by DDAVP (32%), FVIII/vWF concentrates (37%), intravenous immunoglobulins (33%), plasmapheresis (19%), corticosteroids (19%) and immunosuppressive or chemotherapic agents (35%). Based upon the data of this international registry, it appears that AvWS is especially frequent in lympho- or myeloproliferative and cardiovascular diseases. Therefore, AvWS should be suspected and searched with the appropriate laboratory tests especially when excessive bleeding occurs in patients with these disorders. On the basis of the information provided by this registry guidelines for diagnosis and management of the AvWS are given.


Subject(s)
Registries , von Willebrand Diseases , Adolescent , Adult , Aged , Aged, 80 and over , Blood Coagulation Tests , Child , Child, Preschool , Databases, Factual/statistics & numerical data , Female , Humans , Male , Middle Aged , Registries/statistics & numerical data , Surveys and Questionnaires , von Willebrand Diseases/complications , von Willebrand Diseases/diagnosis , von Willebrand Diseases/therapy
20.
J Biol Chem ; 275(33): 25585-94, 2000 Aug 18.
Article in English | MEDLINE | ID: mdl-10831592

ABSTRACT

von Willebrand factor (VWF) is a multimeric glycoprotein that is required for normal hemostasis. After translocation into the endoplasmic reticulum, proVWF subunits dimerize through disulfide bonds between their C-terminal cystine knot-like (CK) domains. CK domains are characterized by six conserved cysteines. Disulfide bonds between cysteines 2 and 5 and between cysteines 3 and 6 define a ring that is penetrated by a disulfide bond between cysteines 1 and 4. Dimerization often is mediated by additional cysteines that differ among CK domain subfamilies. When expressed in a baculovirus system, recombinant VWF CK domains (residues 1957-2050) were secreted as dimers that were converted to monomers by selective reduction and alkylation of three unconserved cysteine residues: Cys(2008), Cys(2010), and Cys(2048). By partial reduction and alkylation, chemical and proteolytic digestion, mass spectrometry, and amino acid sequencing, the remaining intrachain disulfide bonds were characterized: Cys(1961)-Cys(2011) (), Cys(1987)-Cys(2041) (), Cys(1991)-Cys(2043) (), and Cys(1976)-Cys(2025). The mutation C2008A or C2010A prevented dimerization, whereas the mutation C2048A did not. Symmetry considerations and molecular modeling based on the structure of transforming growth factor-beta suggest that one or three of residues Cys(2008), Cys(2010), and Cys(2048) in each subunit mediate the covalent dimerization of proVWF.


Subject(s)
Cystine/chemistry , von Willebrand Factor/chemistry , Alanine/chemistry , Alkylation , Amino Acid Sequence , Animals , COS Cells , Chromatography, High Pressure Liquid , Computer Simulation , Cyanogen Bromide/pharmacology , Dimerization , Disulfides , Dithiothreitol/pharmacology , Ethylmaleimide/pharmacology , Glycosylation , Humans , Indicators and Reagents/pharmacology , Mass Spectrometry , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphines/pharmacology , Plasmids/metabolism , Protein Structure, Tertiary , Pyridines/pharmacology , Recombinant Proteins/chemistry , Sequence Analysis, Protein , Sequence Homology, Amino Acid , Sulfhydryl Reagents/pharmacology , Thermolysin/pharmacology , Time Factors , von Willebrand Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...