Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
PLoS One ; 9(11): e114035, 2014.
Article in English | MEDLINE | ID: mdl-25415203

ABSTRACT

OBJECTIVE: Platelets express the α2ß1 integrin and the glycoprotein VI (GPVI)/FcRγ complex, both collagen receptors. Understanding platelet-collagen receptor function has been enhanced through use of genetically modified mouse models. Previous studies of GPVI/FcRγ-mediated collagen-induced platelet activation were perfomed with mice in which the FcRγ subunit was genetically deleted (FcRγ-/-) or the complex was depleted. The development of α2ß1-/- and GPVI-/- mice permits side-by-side comparison to address contributions of these collagen receptors in vivo and in vitro. APPROACH AND RESULTS: To understand the different roles played by the α2ß1 integrin, the GPVI receptor or FcRγ subunit in collagen-stimulated hemostasis and thrombosis, we compared α2ß1-/-, FcRγ-/-, and GPVI-/- mice in models of endothelial injury and intravascular thrombosis in vivo and their platelets in collagen-stimulated activation in vitro. We demonstrate that both the α2ß1 integrin and the GPVI receptor, but not the FcRγ subunit influence carotid artery occlusion in vivo. In contrast, the GPVI receptor and the FcRγ chain, but not the α2ß1 integrin, play similar roles in intravascular thrombosis in response to soluble Type I collagen. FcRγ-/- platelets showed less attenuation of tyrosine phosphorylation of several proteins including RhoGDI when compared to GPVI-/- and wild type platelets. The difference between FcRγ-/- and GPVI-/- platelet phosphotyrosine levels correlated with the in vivo thrombosis findings. CONCLUSION: Our data demonstrate that genetic deletion of GPVI receptor, FcRγ chain, or the α2ß1 integrin changes the thrombotic potentials of these platelets to collagen dependent on the stimulus mechanism. The data suggest that the FcRγ chain may provide a dominant negative effect through modulating signaling pathways in platelets involving several tyrosine phosphorylated proteins such as RhoGDI. In addition, these findings suggest a more complex signaling network downstream of the platelet collagen receptors than previously appreciated.


Subject(s)
Blood Platelets/metabolism , Collagen/pharmacology , Integrin alpha2beta1/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptors, IgG/metabolism , Thrombosis/metabolism , Animals , Collagen/adverse effects , Disease Models, Animal , Integrin alpha2beta1/genetics , Mice , Mice, Knockout , Platelet Activation/drug effects , Platelet Activation/genetics , Platelet Membrane Glycoproteins/genetics , Rats , Receptors, IgG/genetics , Thrombosis/chemically induced , Thrombosis/genetics
2.
J Immunol ; 190(12): 6303-10, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23656734

ABSTRACT

The abundant serine proteinase inhibitor heparin cofactor II (HCII) has been proposed to inhibit extravascular thrombin. However, the exact physiological role of this plasma protein remains enigmatic. In this study, we demonstrate a previously unknown role for HCII in host defense. Proteolytic cleavage of the molecule induced a conformational change, thereby inducing endotoxin-binding and antimicrobial properties. Analyses employing representative peptide epitopes mapped these effects to helices A and D. Mice deficient in HCII showed increased susceptibility to invasive infection by Pseudomonas aeruginosa, along with a significantly increased cytokine response. Correspondingly, decreased levels of HCII were observed in wild-type animals challenged with bacteria or endotoxin. In humans, proteolytically cleaved HCII forms were detected during wounding and in association with bacteria. Thus, the protease-induced uncovering of cryptic epitopes in HCII, which transforms the molecule into a host defense factor, represents a previously unknown regulatory mechanism in HCII biology and innate immunity.


Subject(s)
Heparin Cofactor II/immunology , Heparin Cofactor II/metabolism , Animals , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Blotting, Western , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteolysis
3.
Bone ; 51(5): 902-12, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22985540

ABSTRACT

Osteoclasts are specialized secretory cells of the myeloid lineage important for normal skeletal homeostasis as well as pathologic conditions of bone including osteoporosis, inflammatory arthritis and cancer metastasis. Differentiation of these multinucleated giant cells from precursors is controlled by the cytokine RANKL, which through its receptor RANK initiates a signaling cascade culminating in the activation of transcriptional regulators which induce the expression of the bone degradation machinery. The transcription factor nuclear factor of activated T-cells c1 (NFATc1) is the master regulator of this process and in its absence osteoclast differentiation is aborted both in vitro and in vivo. Differential mRNA expression analysis by microarray is used to identify genes of potential physiologic relevance across nearly all biologic systems. We compared the gene expression profile of murine wild-type and NFATc1-deficient osteoclast precursors stimulated with RANKL and identified that the majority of the known genes important for osteoclastic bone resorption require NFATc1 for induction. Here, five novel RANKL-induced, NFATc1-dependent transcripts in the osteoclast are described: Nhedc2, Rhoc, Serpind1, Adcy3 and Rab38. Despite reasonable hypotheses for the importance of these molecules in the bone resorption pathway and their dramatic induction during differentiation, the analysis of mice with mutations in these genes failed to reveal a function in osteoclast biology. Compared to littermate controls, none of these mutants demonstrated a skeletal phenotype in vivo or alterations in osteoclast differentiation or function in vitro. These data highlight the need for rigorous validation studies to complement expression profiling results before functional importance can be assigned to highly regulated genes in any biologic process.


Subject(s)
Bone Resorption/genetics , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Mice , NFATC Transcription Factors/genetics , Oligonucleotide Array Sequence Analysis , Osteoclasts/cytology , Osteoclasts/drug effects , RANK Ligand/pharmacology , Real-Time Polymerase Chain Reaction , Sodium-Hydrogen Exchangers/genetics , Sodium-Hydrogen Exchangers/metabolism , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , ras Proteins/genetics , ras Proteins/metabolism , rhoC GTP-Binding Protein
4.
Thromb Res ; 130(4): 646-8, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22178578

ABSTRACT

INTRODUCTION: The thrombin mutant W215A/E217A (WE thrombin) has greatly reduced procoagulant activity, but it activates protein C in the presence of thrombomodulin and inhibits binding of platelet glycoprotein Ib to von Willebrand factor and collagen under flow conditions. Both thrombomodulin-dependent protein C activation and inhibition of platelet adhesion could contribute to the antithrombotic activity of WE thrombin. MATERIALS AND METHODS: To assess the role of thrombomodulin, we administered WE thrombin to thrombomodulin-deficient (TM(Pro/Pro)) mice and measured the time to occlusive thrombus formation in the carotid artery after photochemical injury of the endothelium. RESULTS AND CONCLUSIONS: Doses of WE thrombin ≥10µg/kg prolonged the thrombosis time of wild-type mice (>1.6-fold), while doses ≥100µg/kg only slightly prolonged the thrombosis time of TM(Pro/Pro) mice. We conclude that thrombomodulin plays a predominate role in mediating the antithrombotic effect of WE thrombin in the arterial circulation of mice after endothelial injury. Thrombomodulin-independent effects may occur only when high doses of WE thrombin are administered.


Subject(s)
Carotid Arteries/drug effects , Carotid Artery Thrombosis/drug therapy , Carotid Artery Thrombosis/pathology , Fibrinolytic Agents/therapeutic use , Thrombin/therapeutic use , Thrombomodulin/metabolism , Animals , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Thrombosis/metabolism , Fibrinolytic Agents/metabolism , Gene Deletion , Mice , Mice, Inbred C57BL , Point Mutation , Protein Engineering , Thrombin/genetics , Thrombomodulin/genetics
5.
Blood ; 117(23): 6347-54, 2011 Jun 09.
Article in English | MEDLINE | ID: mdl-21511958

ABSTRACT

Histidine-rich protein II (HRPII) is an abundant protein released into the bloodstream by Plasmodium falciparum, the parasite that causes the most severe form of human malaria. Here, we report that HRPII binds tightly and selectively to coagulation-active glycosaminoglycans (dermatan sulfate, heparan sulfate, and heparin) and inhibits antithrombin (AT). In purified systems, recombinant HRPII neutralized the heparin-catalyzed inhibition of factor Xa and thrombin by AT in a Zn(2+)-dependent manner. The observed 50% inhibitory concentration (IC(50)) for the HRPII neutralization of AT activity is approximately 30nM for factor Xa inhibition and 90nM for thrombin inhibition. Zn(2+) was required for these reactions with a distribution coefficient (K(d)) of approximately 7µM. Substituting Zn(2+) with Cu(2+), but not with Ca(2+), Mg(2+), or Fe(2+), maintained the HRPII effect. HRPII attenuated the prolongation in plasma clotting time induced by heparin, suggesting that HRPII inhibits AT activity by preventing its stimulation by heparin. In the microvasculature, where erythrocytes infected with P falciparum are sequestered, high levels of released HRPII may bind cellular glycosaminoglycans, prevent their interaction with AT, and thereby contribute to the procoagulant state associated with P falciparum infection.


Subject(s)
Antigens, Protozoan/metabolism , Antithrombin Proteins/metabolism , Malaria, Falciparum/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Anticoagulants/pharmacology , Antigens, Protozoan/chemistry , Antigens, Protozoan/genetics , Antithrombin Proteins/chemistry , Antithrombin Proteins/genetics , Blood Coagulation/drug effects , Blood Coagulation/genetics , Factor Xa/chemistry , Factor Xa/genetics , Factor Xa/metabolism , Heparin/pharmacology , Humans , Malaria, Falciparum/genetics , Metals/chemistry , Metals/metabolism , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
6.
Cytotherapy ; 13(6): 695-704, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21250866

ABSTRACT

BACKGROUND AIMS: Previously, we have demonstrated that administration of dermatan sulfate (DS) suppresses neointima formation in the mouse carotid artery by activating heparin co-factor II. A similar suppressive effect was observed by increasing the number of progenitor cells in circulation. In this study, we investigated the combination of DS and bone marrow mononuclear cells (MNC), which includes potential endothelial progenitors, in neointima formation after arterial injury. METHODS: Arterial injury was induced by mechanical dilation of the left common carotid artery. We analyzed the extension of endothelial lesion, thrombus formation, P-selectin expression and CD45(+) cell accumulation 1 and 3 days post-injury, and neointima formation 21 days post-injury. Animals were injected with MNC with or without DS during the first 48 h after injury. RESULTS: The extension of endothelial lesion was similar in all groups 1 day after surgery; however, in injured animals treated with MNC and DS the endothelium recovery seemed to be more efficient 21 days after lesion. Treatment with DS inhibited thrombosis, decreased CD45(+) cell accumulation and P-selectin expression at the site of injury, and reduced the neointimal area by 56%. Treatment with MNC reduced the neointimal area by 54%. The combination of DS and MNC reduced neointima formation by more than 91%. In addition, DS promoted a greater accumulation of MNC at the site of injury. CONCLUSIONS: DS inhibits the initial thrombotic and inflammatory processes after arterial injury and promotes migration of MNC to the site of the lesion, where they may assist in the recovery of the injured endothelium.


Subject(s)
Bone Marrow Cells/cytology , Carotid Arteries/drug effects , Dermatan Sulfate/therapeutic use , Neointima/prevention & control , Neointima/therapy , Animals , Anticoagulants/therapeutic use , Bone Marrow Cells/physiology , Carotid Arteries/metabolism , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , P-Selectin/metabolism , Thrombosis/prevention & control , Thrombosis/therapy
7.
Prog Mol Biol Transl Sci ; 93: 351-72, 2010.
Article in English | MEDLINE | ID: mdl-20807652

ABSTRACT

Heparin cofactor II (HCII) is a plasma protease inhibitor of the serpin family that inactivates thrombin by forming a covalent 1:1 complex. The rate of complex formation increases more than 1000-fold in the presence of dermatan sulfate (DS). Endothelial injury allows circulating HCII to enter the vessel wall, where it binds to DS and presumably becomes activated. Mice that lack HCII develop carotid artery thrombosis more rapidly than wild-type mice after oxidative damage to the endothelium. These mice also have increased arterial neointima formation following mechanical injury and develop more extensive atherosclerotic lesions when made hypercholesterolemic. Similarly, low plasma HCII levels appear to be a risk factor for atherosclerosis and in-stent restenosis in human subjects. These observations suggest that a major function of the HCII-DS system is to regulate the physiologic response to arterial injury.


Subject(s)
Anticoagulants/pharmacology , Blood Vessels/drug effects , Blood Vessels/metabolism , Dermatan Sulfate/pharmacology , Heparin Cofactor II/physiology , Serine Proteinase Inhibitors/physiology , Animals , Atherosclerosis/etiology , Atherosclerosis/pathology , Carotid Artery Thrombosis/etiology , Carotid Artery Thrombosis/pathology , Humans , Mice , Mice, Knockout
8.
Anal Biochem ; 406(2): 166-75, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20670608

ABSTRACT

Irreversible inactivation of alpha-thrombin (T) by the serpin, heparin cofactor II (HCII), is accelerated by ternary complex formation with the glycosaminoglycans (GAGs) heparin and dermatan sulfate (DS). Low expression of human HCII in Escherichia coli was optimized by silent mutation of 27 rare codons and five secondary Shine-Dalgarno sequences in the cDNA. The inhibitory activities of recombinant HCII, and native and deglycosylated plasma HCII, and their affinities for heparin and DS were compared. Recombinant and deglycosylated HCII bound heparin with dissociation constants (K(D)) of 6+/-1 and 7+/-1 microM, respectively, approximately 6-fold tighter than plasma HCII, with K(D) 40+/-4 microM. Binding of recombinant and deglycosylated HCII to DS, both with K(D) 4+/-1 microM, was approximately 4-fold tighter than for plasma HCII, with K(D) 15+/-4 microM. Recombinant HCII, lacking N-glycosylation and tyrosine sulfation, inactivated alpha-thrombin with a 1:1 stoichiometry, similar to plasma HCII. Second-order rate constants for thrombin inactivation by recombinant and deglycosylated HCII were comparable, at optimal GAG concentrations that were lower than those for plasma HCII, consistent with its weaker GAG binding. This weaker binding may be attributed to interference of the Asn(169)N-glycan with the HCII heparin-binding site.


Subject(s)
Escherichia coli/metabolism , Glycosaminoglycans/metabolism , Heparin Cofactor II/metabolism , Amino Acid Sequence , Dermatan Sulfate/metabolism , Enzyme Activation , Fluorescence , Glycosylation , Heparin Cofactor II/chemistry , Heparin Cofactor II/isolation & purification , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thrombin/metabolism
9.
Mol Ther ; 18(2): 334-42, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19844196

ABSTRACT

Mucopolysaccharidosis I (MPS I) is a lysosomal storage disease due to deficiency in alpha-L-iduronidase (IDUA) that results in accumulation of glycosaminoglycans (GAGs) throughout the body, causing numerous clinical defects. Intravenous administration of a gamma-retroviral vector (gamma-RV) with an intact long terminal repeat (LTR) reduced the clinical manifestations of MPS I, but could cause insertional mutagenesis. Although self-inactivating (SIN) gamma-RVs in which the enhancer and promoter elements in the viral LTR are absent after transduction reduces this risk, such vectors could be less effective. This report demonstrates that intravenous (i.v.) injection of a SIN gamma-RV expressing canine IDUA from the liver-specific human alpha(1)-antitrypsin promoter into adult or newborn MPS I mice completely prevents biochemical abnormalities in several organs, and improved bone disease, vision, hearing, and aorta to a similar extent as was seen with administration of the LTR-intact vector to adults. Improvements were less profound than when using an LTR-intact gamma-RV in newborns, which likely reflects a lower level of transduction and expression for the SIN vector-transduced mice, and might be overcome by using a higher dose of SIN vector. A SIN gamma-RV vector ameliorates clinical manifestations of MPS I in mice and should be safer than an LTR-intact gamma-RV.


Subject(s)
Genetic Vectors/genetics , Mucopolysaccharidosis I/therapy , Retroviridae/genetics , Animals , Dogs , Genetic Therapy/methods , Humans , Iduronidase/genetics , Iduronidase/physiology , Marmota , Mice , Promoter Regions, Genetic/genetics , Terminal Repeat Sequences/genetics , Terminal Repeat Sequences/physiology , alpha 1-Antitrypsin/genetics
10.
Blood ; 111(8): 4137-44, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18281502

ABSTRACT

Mice lacking the extracellular matrix protein microfibril-associated glycoprotein-1 (MAGP1) display delayed thrombotic occlusion of the carotid artery following injury as well as prolonged bleeding from a tail vein incision. Normal occlusion times were restored when recombinant MAGP1 was infused into deficient animals prior to vessel wounding. Blood coagulation was normal in these animals as assessed by activated partial thromboplastin time and prothrombin time. Platelet number was lower in MAGP1-deficient mice, but the platelets showed normal aggregation properties in response to various agonists. MAGP1 was not found in normal platelets or in the plasma of wild-type mice. In ligand blot assays, MAGP1 bound to fibronectin, fibrinogen, and von Willebrand factor, but von Willebrand factor was the only protein of the 3 that bound to MAGP1 in surface plasmon resonance studies. These findings show that MAGP1, a component of microfibrils and vascular elastic fibers, plays a role in hemostasis and thrombosis.


Subject(s)
Carotid Arteries/pathology , Contractile Proteins/deficiency , Extracellular Matrix Proteins/deficiency , Thrombosis/pathology , Animals , Bleeding Time , Blood Coagulation/drug effects , Blood Platelets/drug effects , Blood Pressure/drug effects , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Cattle , Contractile Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Fibrinogen/metabolism , Fibronectins/metabolism , Humans , Immunohistochemistry , Injections , Mice , Mice, Inbred C57BL , Platelet Function Tests , Protein Binding/drug effects , RNA Splicing Factors , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacology , Surface Plasmon Resonance , von Willebrand Factor/metabolism
11.
Blood ; 111(8): 4118-25, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18281504

ABSTRACT

Heparin cofactor II (HCII)-deficient mice form occlusive thrombi more rapidly than do wild-type mice following injury to the carotid arterial endothelium. Dermatan sulfate (DS) and heparan sulfate (HS) increase the rate of inhibition of thrombin by HCII in vitro, but it is unknown whether vascular glycosaminoglycans play a role in the antithrombotic effect of HCII in vivo. In this study, we found that intravenous injection of either wild-type recombinant HCII or a variant with low affinity for HS (K173H) corrected the abnormally short thrombosis time of HCII-deficient mice, while a variant with low affinity for DS (R189H) had no effect. When HCII was incubated with frozen sections of the mouse carotid artery, it bound specifically to DS in the adventitia. HCII was undetectable in the wall of the uninjured carotid artery, but it became concentrated in the adventitia following endothelial injury. These results support the hypothesis that HCII interacts with DS in the vessel wall after disruption of the endothelium and that this interaction regulates thrombus formation in vivo.


Subject(s)
Carotid Arteries/metabolism , Dermatan Sulfate/metabolism , Fibrinolytic Agents/metabolism , Heparin Cofactor II/metabolism , Animals , Carotid Arteries/drug effects , Carotid Arteries/pathology , Carotid Arteries/radiation effects , Chondroitin Lyases/metabolism , Female , Heparin Cofactor II/deficiency , Heparin Cofactor II/pharmacokinetics , Heparin Cofactor II/pharmacology , Heparitin Sulfate/metabolism , Humans , Light , Male , Mice , Mice, Inbred C57BL , Mutant Proteins/pharmacology , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Substrate Specificity/drug effects , Substrate Specificity/radiation effects , Swine , Thrombin/antagonists & inhibitors , Thrombosis/pathology , Time Factors
12.
Blood ; 110(13): 4261-7, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17878401

ABSTRACT

Heparin cofactor II (HCII) is a plasma protein that inhibits thrombin when bound to dermatan sulfate or heparin. HCII-deficient mice are viable and fertile but rapidly develop thrombosis of the carotid artery after endothelial injury. We now report the effects of HCII deficiency on atherogenesis and neointima formation. HCII-null or wild-type mice, both on an apolipoprotein E-null background, were fed an atherogenic diet for 12 weeks. HCII-null mice developed plaque areas in the aortic arch approximately 64% larger than wild-type mice despite having similar plasma lipid and glucose levels. Neointima formation was induced by mechanical dilation of the common carotid artery. Thrombin activity, determined by hirudin binding or chromogenic substrate hydrolysis within 1 hour after injury, was higher in the arterial walls of HCII-null mice than in wild-type mice. After 3 weeks, the median neointimal area was 2- to 3-fold greater in HCII-null than in wild-type mice. Dermatan sulfate administered intravenously within 48 hours after injury inhibited neointima formation in wild-type mice but had no effect in HCII-null mice. Heparin did not inhibit neointima formation. We conclude that HCII deficiency promotes atherogenesis and neointima formation and that treatment with dermatan sulfate reduces neointima formation in an HCII-dependent manner.


Subject(s)
Atherosclerosis/etiology , Heparin Cofactor II/deficiency , Tunica Intima/pathology , Animals , Aorta, Thoracic , Carotid Artery Thrombosis , Carotid Artery, Common , Dermatan Sulfate/administration & dosage , Dermatan Sulfate/pharmacology , Heparin Cofactor II/genetics , Mice , Mice, Mutant Strains , Thrombin/metabolism
13.
Arterioscler Thromb Vasc Biol ; 27(3): 454-60, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17194895

ABSTRACT

Heparin cofactor II (HCII) has several biochemical properties that distinguish it from other serpins: (1) it specifically inhibits thrombin; (2) the mechanism of inhibition involves binding of an acidic domain in HCII to thrombin exosite I; and (3) the rate of inhibition increases dramatically in the presence of dermatan sulfate molecules having specific structures. Human studies suggest that high plasma HCII levels are protective against in-stent restenosis and atherosclerosis. Studies with HCII knockout mice directly support the hypothesis that HCII interacts with dermatan sulfate in the arterial wall after endothelial injury and thereby exerts an antithrombotic effect. In addition, HCII deficiency appears to promote neointima formation and atherogenesis in mice. These results suggest that HCII plays a unique and important role in vascular homeostasis.


Subject(s)
Atherosclerosis/metabolism , Dermatan Sulfate/pharmacology , Heparin Cofactor II/metabolism , Thrombin/antagonists & inhibitors , Thrombosis/prevention & control , Animals , Coronary Restenosis/physiopathology , Coronary Restenosis/prevention & control , Disease Models, Animal , Female , Heparin Cofactor II/drug effects , Humans , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Pregnancy , Sensitivity and Specificity , Structure-Activity Relationship , Thrombin/drug effects , Thrombin/metabolism , Thrombosis/physiopathology , Tunica Intima/drug effects , Tunica Intima/metabolism
14.
Glycobiology ; 16(8): 693-701, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16624894

ABSTRACT

Dermatan sulfate (DS) accelerates the inhibition of thrombin by heparin cofactor II (HCII). A hexasaccharide consisting of three l-iduronic acid 2-O-sulfate (IdoA2SO3)-->N-acetyl-D-galactosamine 4-O-sulfate (GalNAc4SO3) subunits was previously isolated from porcine skin DS and shown to bind HCII with high affinity. DS from porcine intestinal mucosa has a much lower content of this disaccharide but activates HCII with potency similar to that of porcine skin DS. Therefore, we sought to characterize oligosaccharides from porcine mucosal DS that interact with HCII. DS was partially depolymerized with chondroitinase ABC, and oligosaccharides containing 2-12 monosaccharide units were isolated. The oligosaccharides were then fractionated by anion-exchange and affinity chromatography on HCII-Sepharose, and the disaccharide compositions of selected fractions were determined. We found that the smallest oligosaccharides able to bind HCII were hexasaccharides. Oligosaccharides 6-12 units long that lacked uronic acid (UA)2SO3 but contained one or two GalNAc4,6SO3 residues bound, and binding was proportional to both oligosaccharide size and number of GalNAc4,6SO3 residues. Intact DS and bound dodecasaccharides contained predominantly IdoA but little D-glucuronic acid. Decasaccharides and dodecasaccharides containing one or two GalNAc4,6SO3 residues stimulated thrombin inhibition by HCII and prolonged the clotting time of normal but not HCII-depleted human plasma. These data support the hypothesis that modification of IdoA-->GalNAc4SO3 subunits in the DS polymer by either 2-O-sulfation of IdoA or 6-O-sulfation of GalNAc can generate molecules with HCII-binding sites and anticoagulant activity.


Subject(s)
Acetylgalactosamine/chemistry , Dermatan Sulfate/metabolism , Heparin Cofactor II/metabolism , Mucous Membrane/chemistry , Sulfates/chemistry , Animals , Carbohydrate Sequence , Chondroitin Lyases/genetics , Chondroitin Lyases/metabolism , Dermatan Sulfate/chemistry , Heparin Cofactor II/isolation & purification , Humans , Monosaccharides/chemistry , Monosaccharides/isolation & purification , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Recombinant Proteins/metabolism , Swine , Thrombin/antagonists & inhibitors
15.
Blood ; 107(7): 2753-8, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16339402

ABSTRACT

Pregnancy is associated with hemostatic challenges that may lead to thrombosis. Heparin cofactor II (HCII) is a glycosaminoglycan-dependent thrombin inhibitor present in both maternal and fetal plasma. HCII activity increases during pregnancy, and HCII levels are significantly decreased in women with severe pre-eclampsia. Dermatan sulfate (DS) specifically activates HCII and is abundant in the placenta, but the locations of DS and HCII in the placenta have not been determined. We present evidence that DS is the major anticoagulant glycosaminoglycan in the human placenta at term. DS isolated from human placenta contains disaccharides implicated in activation of HCII and has anticoagulant activity similar to that of mucosal DS. Immunohistochemical studies revealed that DS is associated with fetal blood vessels and stromal regions of placental villi but is notably absent from the syncytiotrophoblast cells in contact with the maternal circulation. HCII colocalizes with DS in the walls of fetal blood vessels and is also present in syncytiotrophoblast cells. Our data suggest that DS is in a position to activate HCII in the fetal blood vessels or in the stroma of placental villi after injury to the syncytiotrophoblast layer and thereby inhibit fibrin generation in the placenta.


Subject(s)
Dermatan Sulfate/metabolism , Heparin Cofactor II/metabolism , Placenta/physiology , Anticoagulants/isolation & purification , Anticoagulants/pharmacology , Dermatan Sulfate/isolation & purification , Dermatan Sulfate/pharmacology , Female , Glycosaminoglycans/isolation & purification , Heparin Cofactor II/analysis , Humans , Immunohistochemistry , Placenta/cytology , Pre-Eclampsia/metabolism , Pregnancy , Substrate Specificity
17.
Blood ; 105(10): 3902-9, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15677561

ABSTRACT

The anionic phospholipid, phosphatidyl-L-serine (PS), is sequestered in the inner layer of the plasma membrane in normal cells. Upon injury, activation, and apoptosis, PS becomes exposed on the surfaces of cells and sheds microparticles, which are procoagulant. Coagulation is initiated by formation of a tissue factor/factor VIIa complex on PS-exposed membranes and propagated through the assembly of intrinsic tenase (factor VIIIa/factor IXa), prothrombinase (factor Va/factor Xa), and factor XIa complexes on PS-exposed activated platelets. We constructed a novel series of recombinant anticoagulant fusion proteins by linking annexin V (ANV), a PS-binding protein, to the Kunitz-type protease inhibitor (KPI) domain of tick anticoagulant protein, an aprotinin mutant (6L15), amyloid beta-protein precursor, or tissue factor pathway inhibitor. The resulting ANV-KPI fusion proteins were 6- to 86-fold more active than recombinant tissue factor pathway inhibitor and tick anticoagulant protein in an in vitro tissue factor-initiated clotting assay. The in vivo antithrombotic activities of the most active constructs were 3- to 10-fold higher than that of ANV in a mouse arterial thrombosis model. ANV-KPI fusion proteins represent a new class of anticoagulants that specifically target the anionic membrane-associated coagulation enzyme complexes present at sites of thrombogenesis and are potentially useful as antithrombotic agents.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Annexin A5/metabolism , Anticoagulants/metabolism , Blood Coagulation/drug effects , Protease Inhibitors/metabolism , Thrombosis/metabolism , Amyloid beta-Peptides/genetics , Animals , Annexin A5/genetics , Annexin A5/isolation & purification , Anticoagulants/chemistry , Anticoagulants/pharmacology , Cattle , Factor Xa/metabolism , Humans , Mice , Models, Animal , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protein Structure, Tertiary , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Swine , Time Factors , Trypsin/metabolism
18.
Glycobiology ; 15(4): 369-81, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15590773

ABSTRACT

A linear sulfated fucan with a regular repeating sequence of [3)-alpha-L-Fucp-(2SO4)-(1-->3)-alpha-L-Fucp-(4SO4)-(1-->3)-alpha-L-Fucp-(2,4SO4)-(1-->3)-alpha-L-Fucp-(2SO4)-(1-->]n is an anticoagulant polysaccharide mainly due to thrombin inhibition mediated by heparin cofactor II. No specific enzymatic or chemical method is available for the preparation of tailored oligosaccharides from sulfated fucans. We employ an apparently nonspecific approach to cleave this polysaccharide based on mild hydrolysis with acid. Surprisingly, the linear sulfated fucan was cleaved by mild acid hydrolysis on an ordered sequence. Initially a 2-sulfate ester of the first fucose unit is selectively removed. Thereafter the glycosidic linkage between the nonsulfated fucose residue and the subsequent 4-sulfated residue is preferentially cleaved by acid hydrolysis, forming oligosaccharides with well-defined size. The low-molecular-weight derivatives obtained from the sulfated fucan were employed to determine the requirement for interaction of this polysaccharide with heparin cofactor II and to achieve complete thrombin inhibition. The linear sulfated fucan requires significantly longer chains than mammalian glycosaminoglycans to achieve anticoagulant activity. A slight decrease in the molecular size of the sulfated fucan dramatically reduces its effect on thrombin inactivation mediated by heparin cofactor II. Sulfated fucan with approximately 45 tetrasaccharide repeating units binds to heparin cofactor II but is unable to link efficiently the plasma inhibitor and thrombin. This last effect requires chains with approximately 100 or more tetrasaccharide repeating units. We speculate that the template mechanism may predominate over the allosteric effect in the case of the linear sulfated fucan inactivation of thrombin in the presence of heparin cofactor II.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Laminaria/chemistry , Polysaccharides/pharmacology , Sea Urchins/chemistry , Animals , Anticoagulants/chemistry , Anticoagulants/isolation & purification , Heparin Cofactor II/chemistry , Heparin Cofactor II/metabolism , Hydrolysis , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Sulfuric Acid Esters/chemistry
19.
Blood ; 104(13): 3965-70, 2004 Dec 15.
Article in English | MEDLINE | ID: mdl-15315969

ABSTRACT

Heparin cofactor II (HCII) is a plasma protein that inhibits thrombin rapidly in the presence of dermatan sulfate or heparin. We previously reported that the time to thrombotic occlusion of the carotid artery after photochemical injury was shorter in HCII-deficient mice than in wild-type control animals. In this paper, we describe the antithrombotic activity of dermatan sulfate in wild-type and HCII-deficient mice. Intravenous administration of porcine skin dermatan sulfate induced a dose-dependent prolongation of the carotid artery occlusion time in HCII(+/+) mice that was not observed in HCII(-/-) animals. Pharmacokinetic studies suggested that porcine skin dermatan sulfate expresses antithrombotic activity after being transferred from the plasma to sites in the vessel wall. Using invertebrate dermatan sulfate preparations, we showed that N-acetylgalactosamine-4-O-sulfate residues are required for the HCII-dependent antithrombotic effect. Furthermore, the invertebrate dermatan sulfates, which have higher charge densities than mammalian dermatan sulfate, slightly prolonged the thrombotic occlusion time of HCII(-/-) mice. These results indicate that HCII mediates the antithrombotic effect of porcine skin dermatan sulfate after injury to the carotid arterial endothelium in mice, whereas more highly charged dermatan sulfates possess weak antithrombotic activity independent of HCII.


Subject(s)
Antithrombins/pharmacology , Dermatan Sulfate/pharmacology , Heparin Cofactor II/deficiency , Animals , Carotid Artery Thrombosis/drug therapy , Dermatan Sulfate/pharmacokinetics , Disease Models, Animal , Heparin Cofactor II/genetics , Heparin Cofactor II/therapeutic use , Humans , Mice , Mice, Knockout , Recombination, Genetic , Skin/metabolism , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...