Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hum Mutat ; 41(1): 277-290, 2020 01.
Article in English | MEDLINE | ID: mdl-31562665

ABSTRACT

The heterogeneous manifestations of MYH9-related disorder (MYH9-RD), characterized by macrothrombocytopenia, Döhle-like inclusion bodies in leukocytes, bleeding of variable severity with, in some cases, ear, eye, kidney, and liver involvement, make the diagnosis for these patients still challenging in clinical practice. We collected phenotypic data and analyzed the genetic variants in more than 3,000 patients with a bleeding or platelet disorder. Patients were enrolled in the BRIDGE-BPD and ThromboGenomics Projects and their samples processed by high throughput sequencing (HTS). We identified 50 patients with a rare variant in MYH9. All patients had macrothrombocytes and all except two had thrombocytopenia. Some degree of bleeding diathesis was reported in 41 of the 50 patients. Eleven patients presented hearing impairment, three renal failure and two elevated liver enzymes. Among the 28 rare variants identified in MYH9, 12 were novel. HTS was instrumental in diagnosing 23 patients (46%). Our results confirm the clinical heterogeneity of MYH9-RD and show that, in the presence of an unclassified platelet disorder with macrothrombocytes, MYH9-RD should always be considered. A HTS-based strategy is a reliable method to reach a conclusive diagnosis of MYH9-RD in clinical practice.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , High-Throughput Nucleotide Sequencing , Myosin Heavy Chains/genetics , Adolescent , Adult , Aged , Alleles , Child , Child, Preschool , Chromosome Mapping , Evolution, Molecular , Female , Fluorescent Antibody Technique , Gene Expression , Genetic Association Studies/methods , Genotype , High-Throughput Nucleotide Sequencing/methods , Humans , Infant , Male , Middle Aged , Mutation , Myosin Heavy Chains/metabolism , Phenotype , Young Adult
2.
J Biol Chem ; 294(13): 4878-4888, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30670593

ABSTRACT

ER protein 57 (ERp57), a thiol isomerase secreted from vascular cells, is essential for complete thrombus formation in vivo, but other extracellular ERp57 functions remain unexplored. Here, we employed a kinetic substrate-trapping approach to identify extracellular protein substrates of ERp57 in platelet-rich plasma. MS-based identification with immunochemical confirmation combined with gene ontology enrichment analysis revealed that ERp57 targets, among other substrates, components of the lectin pathway of complement activation: mannose-binding lectin, ficolin-2, ficolin-3, collectin-10, collectin-11, mannose-binding lectin-associated serine protease-1, and mannose-binding lectin-associated serine protease-2. Ficolin-3, the most abundant lectin pathway initiator in humans, circulates as disulfide-linked multimers of a monomer. ERp57 attenuated ficolin-3 ligand recognition and complement activation by cleaving intermolecular disulfide bonds in large ficolin-3 multimers, thereby reducing multimer size and ligand-binding affinity. We used MS to identify the disulfide-bonding pattern in ficolin-3 multimers and the disulfide bonds targeted by ERp57 and found that Cys6 and Cys23 in the N-terminal region of ficolin-3 form the intermolecular disulfide bonds in ficolin-3 multimers that are reduced by ERp57. Our results not only demonstrate that ERp57 can negatively regulate complement activation, but also identify a control mechanism for lectin pathway initiation in the vasculature. We conclude that extensive multimerization in large ficolin-3 multimers leads to a high affinity for ligands and strong complement-activating potential and that ERp57 suppresses complement activation by cleaving disulfide bonds in ficolin-3 and reducing its multimer size.


Subject(s)
Complement Pathway, Mannose-Binding Lectin , Glycoproteins/metabolism , Lectins/metabolism , Protein Disulfide-Isomerases/metabolism , Protein Multimerization , Proteolysis , Glycoproteins/genetics , Humans , Lectins/genetics , Protein Disulfide-Isomerases/genetics
3.
J Biol Chem ; 292(22): 9063-9074, 2017 06 02.
Article in English | MEDLINE | ID: mdl-28364042

ABSTRACT

Thiol isomerases such as protein-disulfide isomerase (PDI) direct disulfide rearrangements required for proper folding of nascent proteins synthesized in the endoplasmic reticulum. Identifying PDI substrates is challenging because PDI catalyzes conformational changes that cannot be easily monitored (e.g. compared with proteolytic cleavage or amino acid phosphorylation); PDI has multiple substrates; and it can catalyze either oxidation, reduction, or isomerization of substrates. Kinetic-based substrate trapping wherein the active site motif CGHC is modified to CGHA to stabilize a PDI-substrate intermediate is effective in identifying some substrates. A limitation of this approach, however, is that it captures only substrates that are reduced by PDI, whereas many substrates are oxidized by PDI. By manipulating the highly conserved -GH- residues in the CGHC active site of PDI, we created PDI variants with a slowed reaction rate toward substrates. The prolonged intermediate state allowed us to identify protein substrates that have biased affinities for either oxidation or reduction by PDI. Because extracellular PDI is critical for thrombus formation but its extracellular substrates are not known, we evaluated the ability of these bidirectional trapping PDI variants to trap proteins released from platelets and on the platelet surface. Trapped proteins were identified by mass spectroscopy. Of the trapped substrate proteins identified by mass spectroscopy, five proteins, cathepsin G, glutaredoxin-1, thioredoxin, GP1b, and fibrinogen, showed a bias for oxidation, whereas annexin V, heparanase, ERp57, kallekrein-14, serpin B6, tetranectin, and collagen VI showed a bias for reduction. These bidirectional trapping variants will enable more comprehensive identification of thiol isomerase substrates and better elucidation of their cellular functions.


Subject(s)
Blood Platelets/enzymology , Protein Disulfide-Isomerases/chemistry , Catalytic Domain , Humans , Kinetics , Protein Disulfide-Isomerases/metabolism , Substrate Specificity
4.
Blood ; 128(7): 893-901, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27357699

ABSTRACT

Thiol isomerases are multifunctional enzymes that influence protein structure via their oxidoreductase, isomerase, and chaperone activities. These enzymes localize at high concentrations in the endoplasmic reticulum of all eukaryotic cells where they serve an essential function in folding nascent proteins. However, thiol isomerases can escape endoplasmic retention and be secreted and localized on plasma membranes. Several thiol isomerases including protein disulfide isomerase, ERp57, and ERp5 are secreted by and localize to the membranes of platelets and endothelial cells. These vascular thiol isomerases are released following vessel injury and participate in thrombus formation. Although most of the activities of vascular thiol isomerases that contribute to thrombus formation are yet to be defined at the molecular level, allosteric disulfide bonds that are modified by thiol isomerases have been described in substrates such as αIIbß3, αvß3, GPIbα, tissue factor, and thrombospondin. Vascular thiol isomerases also act as redox sensors. They respond to the local redox environment and influence S-nitrosylation of surface proteins on platelets and endothelial cells. Despite our rudimentary understanding of the mechanisms by which thiol isomerases control vascular function, the clinical utility of targeting them in thrombotic disorders is already being explored in clinical trials.


Subject(s)
Blood Vessels/enzymology , Protein Disulfide-Isomerases/metabolism , Animals , Blood Vessels/pathology , Hemostasis , Humans , Models, Biological , Oxidation-Reduction , Thrombosis/enzymology , Thrombosis/pathology
5.
Blood ; 127(23): 2791-803, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27084890

ABSTRACT

Inherited bleeding, thrombotic, and platelet disorders (BPDs) are diseases that affect ∼300 individuals per million births. With the exception of hemophilia and von Willebrand disease patients, a molecular analysis for patients with a BPD is often unavailable. Many specialized tests are usually required to reach a putative diagnosis and they are typically performed in a step-wise manner to control costs. This approach causes delays and a conclusive molecular diagnosis is often never reached, which can compromise treatment and impede rapid identification of affected relatives. To address this unmet diagnostic need, we designed a high-throughput sequencing platform targeting 63 genes relevant for BPDs. The platform can call single nucleotide variants, short insertions/deletions, and large copy number variants (though not inversions) which are subjected to automated filtering for diagnostic prioritization, resulting in an average of 5.34 candidate variants per individual. We sequenced 159 and 137 samples, respectively, from cases with and without previously known causal variants. Among the latter group, 61 cases had clinical and laboratory phenotypes indicative of a particular molecular etiology, whereas the remainder had an a priori highly uncertain etiology. All previously detected variants were recapitulated and, when the etiology was suspected but unknown or uncertain, a molecular diagnosis was reached in 56 of 61 and only 8 of 76 cases, respectively. The latter category highlights the need for further research into novel causes of BPDs. The ThromboGenomics platform thus provides an affordable DNA-based test to diagnose patients suspected of having a known inherited BPD.


Subject(s)
Blood Platelet Disorders/genetics , Genetic Predisposition to Disease , Hemorrhage/genetics , High-Throughput Nucleotide Sequencing/methods , Thrombosis/genetics , Case-Control Studies , DNA Copy Number Variations , Female , Genetic Association Studies/methods , Humans , Male , Mutation , Polymorphism, Single Nucleotide , Sequence Analysis, DNA/methods
6.
Blood ; 125(3): 438-42, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25377783

ABSTRACT

Severe deficiency of vitamin K-dependent proteins in patients not maintained on vitamin K antagonists is most commonly associated with poisoning by or surreptitious ingestion of warfarin, warfarin-like anticoagulants, or potent rodenticides ("superwarfarins"), such as brodifacoum. Serious bleeding manifestations are common. Superwarfarins are 2 orders of magnitude more potent than warfarin and have a half-life measured in weeks. These rodenticides are readily available household environmental hazards and are sometimes consumed accidentally or as manifestations of psychiatric disease. Immediate diagnosis and proper therapy is critically important to minimize morbidity and mortality because this condition, affecting thousands of patients annually, is reversible. Treatment with large doses of oral vitamin K1, often over months to years, to maintain a near-normal prothrombin time can reverse the coagulopathy associated with superwarfarins. Although these patients initially present to various medical specialties, the hematologist is often consulted to offer the definitive diagnosis and proper therapy.


Subject(s)
Anticoagulants/poisoning , Blood Coagulation Disorders/drug therapy , Hemorrhage/drug therapy , Rodenticides/poisoning , Vitamin K/antagonists & inhibitors , Aged , Anticoagulants/blood , Anticoagulants/pharmacokinetics , Blood Coagulation Disorders/complications , Female , Hemorrhage/etiology , Humans , Male , Middle Aged , Prognosis , Rodenticides/blood , Rodenticides/pharmacokinetics
7.
Blood ; 125(10): 1633-42, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25593336

ABSTRACT

Protein disulfide isomerase (PDI), secreted from platelets and endothelial cells after injury, is required for thrombus formation. The effect of platelet and endothelial cell granule contents on PDI-mediated thrombus formation was studied by intravital microscopy using a mouse model of Hermansky-Pudlak syndrome in which platelet dense granules are absent. Platelet deposition and fibrin generation were nearly absent, and extracellular PDI was significantly reduced in HPS6(-/-) mice after vascular injury. HPS6(-/-) platelets displayed impaired PDI secretion and impaired exocytosis of α granules, lysosomes, and T granules due to decreased sensitivity to thrombin, but these defects could be corrected by addition of subthreshold amounts of adenosine 5'-diphosphate (ADP). Human Hermansky-Pudlak syndrome platelets demonstrated similar characteristics. Infusion of wild-type platelets rescued thrombus formation in HPS6(-/-) mice. Human umbilical vein endothelial cells in which the HPS6 gene was silenced displayed impaired PDI secretion and exocytosis of Weibel-Palade bodies. Defective thrombus formation in Hermansky-Pudlak syndrome, associated with impaired exocytosis of residual granules in endothelial cells and platelets, the latter due to deficiency of ADP, is characterized by a defect in T granule secretion, a deficiency in extracellular PDI secretion, and impaired fibrin generation and platelet aggregation. Hermansky-Pudlak syndrome is an example of a hereditary disease whereby impaired PDI secretion contributes to a bleeding phenotype.


Subject(s)
Blood Platelets/enzymology , Endothelial Cells/enzymology , Hermanski-Pudlak Syndrome/blood , Hermanski-Pudlak Syndrome/enzymology , Protein Disulfide-Isomerases/metabolism , Thrombosis/blood , Thrombosis/enzymology , Adenosine Diphosphate/deficiency , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/pharmacology , Animals , Apyrase/metabolism , Apyrase/pharmacology , Blood Platelets/drug effects , Cell Degranulation , Disease Models, Animal , Endothelial Cells/pathology , Exocytosis/drug effects , Female , Fibrin/biosynthesis , Hermanski-Pudlak Syndrome/genetics , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/blood , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Aggregation , Protein Disulfide-Isomerases/blood , RNA, Small Interfering/genetics , Thrombin/metabolism , Vesicular Transport Proteins/deficiency , Vesicular Transport Proteins/genetics
8.
Blood ; 125(14): 2276-85, 2015 Apr 02.
Article in English | MEDLINE | ID: mdl-25624318

ABSTRACT

Protein disulfide isomerase (PDI) and endoplasmic reticulum protein 57 (ERp57) are emerging as important regulators of thrombus formation. Another thiol isomerase, endoplasmic reticulum protein 5 (ERp5), is involved in platelet activation. We show here the involvement of ERp5 in thrombus formation using the mouse laser-injury model of thrombosis and a specific antibody raised against recombinant ERp5. Anti-ERp5 antibody inhibited ERp5-dependent platelet and endothelial cell disulfide reductase activity in vitro. ERp5 release at the thrombus site was detected after infusion of Alexa Fluor 488-labeled anti-ERp5 antibody at 0.05 µg/g body weight, a dose that does not inhibit thrombus formation. Anti-ERp5 at 3 µg/g body weight inhibited laser-induced thrombus formation in vivo by causing a 70% decrease in the deposition of platelets and a 62% decrease in fibrin accumulation compared to infusion of control antibody (P < .01). ERp5 binds to ß3 integrin with an equilibrium dissociation constant (KD) of 21 µM, measured by surface plasmon resonance. The cysteine residues in the ERp5 active sites are not required for binding to ß3 integrin. These results provide evidence for a novel role of ERp5 in thrombus formation, a function that may be mediated through its association with αIIbß3.


Subject(s)
Disease Models, Animal , Integrin beta3/metabolism , Lasers/adverse effects , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Disulfide-Isomerases/metabolism , Thrombosis/pathology , Animals , Blood Platelets/metabolism , Blood Platelets/pathology , Blotting, Western , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Enzyme-Linked Immunosorbent Assay , Fibrin/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Platelet Activation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface Plasmon Resonance , Thrombosis/enzymology , Thrombosis/etiology
9.
Nat Chem Biol ; 11(4): 292-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25686372

ABSTRACT

In bacteria, disulfide bonds confer stability on many proteins exported to the cell envelope or beyond. These proteins include numerous bacterial virulence factors, and thus bacterial enzymes that promote disulfide bond formation represent targets for compounds inhibiting bacterial virulence. Here, we describe a new target- and cell-based screening methodology for identifying compounds that inhibit the disulfide bond-forming enzymes Escherichia coli DsbB (EcDsbB) or Mycobacterium tuberculosis VKOR (MtbVKOR), which can replace EcDsbB, although the two are not homologs. Initial screening of 51,487 compounds yielded six specifically inhibiting EcDsbB. These compounds share a structural motif and do not inhibit MtbVKOR. A medicinal chemistry approach led us to select related compounds, some of which are much more effective DsbB inhibitors than those found in the screen. These compounds inhibit purified DsbB and prevent anaerobic growth of E. coli. Furthermore, these compounds inhibit all but one of the DsbBs of nine other Gram-negative pathogenic bacteria tested.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Escherichia coli/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Mycobacterium tuberculosis/metabolism , Agar/chemistry , Anti-Bacterial Agents/chemistry , Catalytic Domain , Chemistry, Pharmaceutical/methods , Combinatorial Chemistry Techniques , Disulfides , Dose-Response Relationship, Drug , Drug Design , Electron Transport , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli Proteins/chemistry , Mass Spectrometry , Microbial Sensitivity Tests , Mycobacterium smegmatis/metabolism , Protein Conformation , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/chemistry , Pseudomonas aeruginosa/metabolism
10.
J Cell Mol Med ; 20(10): 1851-60, 2016 10.
Article in English | MEDLINE | ID: mdl-27197780

ABSTRACT

Fibrinolysis is a process responsible for the dissolution of formed thrombi to re-establish blood flow after thrombus formation. Plasminogen activator inhibitor-1 (PAI-1) inhibits urokinase-type and tissue-type plasminogen activator (uPA and tPA) and is the major negative regulator of fibrinolysis. Inhibition of PAI-1 activity prevents thrombosis and accelerates fibrinolysis. However, a specific antagonist of PAI-1 is currently unavailable for therapeutic use. We screened a panel of uPA variants with mutations at and near the active site to maximize their binding to PAI-1 and identified a potent PAI-1 antagonist, PAItrap. PAItrap is the serine protease domain of urokinase containing active-site mutation (S195A) and four additional mutations (G37bR-R217L-C122A-N145Q). PAItrap inhibits human recombinant PAI-1 with high potency (Kd = 0.15 nM) and high specificity. In vitro using human plasma, PAItrap showed significant thrombolytic activity by inhibiting endogenous PAI-1. In addition, PAItrap inhibits both human and murine PAI-1, allowing the evaluation in murine models. In vivo, using a laser-induced thrombosis mouse model in which thrombus formation and fibrinolysis are monitored by intravital microscopy, PAItrap reduced fibrin generation and inhibited platelet accumulation following vascular injury. Therefore, this work demonstrates the feasibility to generate PAI-1 inhibitors using inactivated urokinase.


Subject(s)
Peptide Fragments/pharmacology , Plasminogen Activator Inhibitor 1/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Amino Acid Sequence , Animals , Catalytic Domain , Crystallography, X-Ray , Disease Models, Animal , Fibrinolysis , Humans , Inhibitory Concentration 50 , Kinetics , Mice, Inbred C57BL , Models, Molecular , Mutant Proteins/chemistry , Peptide Fragments/chemistry , Protein Binding , Thrombosis/pathology , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/pharmacology
11.
J Biol Chem ; 290(39): 23543-52, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26240139

ABSTRACT

Quercetin-3-rutinoside inhibits thrombus formation in a mouse model by inhibiting extracellular protein disulfide isomerase (PDI), an enzyme required for platelet thrombus formation and fibrin generation. Prior studies have identified PDI as a potential target for novel antithrombotic agents. Using a fluorescence enhancement-based assay and isothermal calorimetry, we show that quercetin-3-rutinoside directly binds to the b' domain of PDI with a 1:1 stoichiometry. The binding of quercetin-3-rutinoside to PDI induces a more compact conformation and restricts the conformational flexibility of PDI, as revealed by small angle x-ray scattering. The binding sites of quercetin-3-rutinoside to PDI were determined by studying its interaction with isolated fragments of PDI. Quercetin-3-rutinoside binds to the b'x domain of PDI. The infusion of the b'x fragment of PDI rescued thrombus formation that was inhibited by quercetin-3-rutinoside in a mouse thrombosis model. This b'x fragment does not possess reductase activity and, in the absence of quercetin-3-rutinoside, does not affect thrombus formation in vivo. The isolated b' domain of PDI has potential as an antidote to reverse the antithrombotic effect of quercetin-3-rutinoside by binding and neutralizing quercetin-3-rutinoside.


Subject(s)
Protein Disulfide-Isomerases/antagonists & inhibitors , Rutin/pharmacology , Animals , Binding Sites , Calorimetry , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Protein Disulfide-Isomerases/metabolism , Rutin/metabolism , Scattering, Small Angle , Thrombosis/prevention & control , X-Ray Diffraction
12.
Blood ; 124(4): 611-22, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-24825863

ABSTRACT

Antiphospholipid syndrome (APS) is defined by thrombosis, fetal loss, and the presence of antiphospholipid antibodies, including anti-ß2-glycoprotein-1 autoantibodies (anti-ß2GP1) that have a direct role in the pathogenesis of thrombosis in vivo. The cellular targets of the anti-ß2GP1 autoantibody/ß2GP1 complex in vivo were studied using a laser-induced thrombosis model of APS in a live mouse and human anti-ß2GP1 autoantibodies affinity-purified from APS patients. Cell binding of fluorescently labeled ß2GP1 and anti-ß2GP1 autoantibodies revealed their colocalization on the platelet thrombus but not the endothelium. Anti-ß2GP1 autoantibodies enhanced platelet activation, monitored by calcium mobilization, and endothelial activation, monitored by intercellular adhesion molecule-1 expression. When eptifibatide was infused to block platelet thrombus formation, enhanced fibrin generation and endothelial cell activation were eliminated. Thus, the anti-ß2GP1 autoantibody/ß2GP1 complex binds to the thrombus, enhancing platelet activation, and platelet secretion leads to enhanced endothelium activation and fibrin generation. These results lead to a paradigm shift away from the concept that binding of the anti-ß2GP1 autoantibody/ß2GP1 complex activates both endothelial cells and platelets toward one in which activation of platelets in response to anti-ß2GP1 autoantibody/ß2GP1 complex binding leads to subsequent enhanced endothelium activation and fibrin generation.


Subject(s)
Antibodies, Antiphospholipid/blood , Antiphospholipid Syndrome/immunology , Blood Platelets/immunology , Disease Models, Animal , Endothelium/immunology , Thrombosis/immunology , beta 2-Glycoprotein I/metabolism , Animals , Antiphospholipid Syndrome/metabolism , Antiphospholipid Syndrome/pathology , Autoantibodies/blood , Autoantibodies/immunology , Blood Platelets/cytology , Blood Platelets/metabolism , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , Endothelium/cytology , Endothelium/metabolism , Fibrin/metabolism , Fibrinogen/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Mice , Mice, Inbred C57BL , Platelet Activation , Thrombosis/metabolism , Thrombosis/pathology , beta 2-Glycoprotein I/immunology
13.
Circ Res ; 114(7): 1162-73, 2014 Mar 28.
Article in English | MEDLINE | ID: mdl-24677236

ABSTRACT

Protein disulfide isomerase (PDI), ERp5, and ERp57, among perhaps other thiol isomerases, are important for the initiation of thrombus formation. Using the laser injury thrombosis model in mice to induce in vivo arterial thrombus formation, it was shown that thrombus formation is associated with PDI secretion by platelets, that inhibition of PDI blocked platelet thrombus formation and fibrin generation, and that endothelial cell activation leads to PDI secretion. Similar results using this and other thrombosis models in mice have demonstrated the importance of ERp5 and ERp57 in the initiation of thrombus formation. The integrins, αIIbß3 and αVß3, play a key role in this process and interact directly with PDI, ERp5, and ERp57. The mechanism by which thiol isomerases participate in thrombus generation is being evaluated using trapping mutant forms to identify substrates of thiol isomerases that participate in the network pathways linking thiol isomerases, platelet receptor activation, and fibrin generation. PDI as an antithrombotic target is being explored using isoquercetin and quercetin 3-rutinoside, inhibitors of PDI identified by high throughput screening. Regulation of thiol isomerase expression, analysis of the storage, and secretion of thiol isomerases and determination of the electron transfer pathway are key issues to understanding this newly discovered mechanism of regulation of the initiation of thrombus formation.


Subject(s)
Protein Disulfide-Isomerases/metabolism , Thrombosis/enzymology , Animals , Blood Platelets/metabolism , Flavonoids/pharmacology , Flavonoids/therapeutic use , Humans , Protein Disulfide-Isomerases/antagonists & inhibitors , Quercetin/pharmacology , Quercetin/therapeutic use , Thrombosis/drug therapy , Thrombosis/metabolism
14.
Arterioscler Thromb Vasc Biol ; 35(1): 16-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25104801

ABSTRACT

The study of thrombus formation has increasingly applied in vivo tools such as genetically modified mice and intravital microscopy to the evaluation of molecular and cellular mechanisms of thrombosis. Among several unexpected findings of this approach was the discovery that protein disulfide isomerase serves an essential role in thrombus formation at sites of vascular injury. The observation that the commonly ingested quercetin flavonoid, quercetin-3-rutinoside, inhibits protein disulfide isomerase and blocks thrombus formation in preclinical studies has set the stage for clinical trials using protein disulfide isomerase antagonists as antithrombotics. Although the mechanisms by which protein disulfide isomerase facilitates platelet activation and fibrin formation have yet to be elucidated, protein disulfide isomerase antagonists are currently being developed as antithrombotics. This review will consider what is known about the role of protein disulfide isomerase in platelet accumulation and fibrin generation with a focus on pharmacological strategies for blocking protein disulfide isomerase activity in the context of thrombus formation. Potential indications and clinical trial design for testing the efficacy of protein disulfide isomerase inhibition to reduce the incidence of thrombosis will be considered.


Subject(s)
Blood Platelets/drug effects , Enzyme Inhibitors/therapeutic use , Fibrinolytic Agents/therapeutic use , Protein Disulfide-Isomerases/antagonists & inhibitors , Thrombosis/drug therapy , Animals , Blood Coagulation/drug effects , Blood Platelets/enzymology , Drug Design , Fibrin/metabolism , Humans , Platelet Activation/drug effects , Protein Disulfide-Isomerases/blood , Thrombosis/blood , Thrombosis/enzymology
15.
Blood ; 120(3): 647-55, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22653978

ABSTRACT

Extracellular protein disulfide isomerase (PDI) is required for platelet thrombus formation and fibrin generation after arteriolar wall injury in live mice. PDI is secreted from platelets and endothelial cells on cellular activation, but the mechanism of capture of secreted PDI within the injured vasculature is unknown. We establish that, like the endothelial ß3 integrin α(V)ß(3), the platelet integrin α(IIb)ß(3) binds PDI. PDI also binds to recombinant ß3. Using intravital microscopy, we demonstrate that PDI accumulation at the site of laser-induced arteriolar wall injury is markedly reduced in ß3-null (ß3(-/-)) mice, and neither a platelet thrombus nor fibrin is generated at the vessel injury site. The absence of fibrin after vascular injury in ß3(-/-) mice is because of the absence of extracellular PDI. To evaluate the relative importance of endothelial α(V)ß(3) versus platelet α(IIb)ß(3) or α(V)ß(3), we performed reciprocal bone marrow transplants on wild-type and ß3(-/-) mice. PDI accumulation and platelet thrombus formation were markedly decreased after vessel injury in wild-type mice transplanted with ß3(-/-) bone marrow or in ß3(-/-) mice transplanted with wild-type bone marrow. These results indicate that both endothelial and platelet ß3 integrins contribute to extracellular PDI binding at the vascular injury site.


Subject(s)
Blood Platelets/enzymology , Endothelial Cells/enzymology , Integrin beta3/metabolism , Protein Disulfide-Isomerases/metabolism , Thrombosis/metabolism , Animals , Arterioles/enzymology , Arterioles/injuries , Blood Coagulation/physiology , Blood Platelets/metabolism , Bone Marrow Transplantation , CHO Cells , Cricetinae , Extracellular Space/enzymology , Integrin alphaVbeta3/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Mutant Strains , Microscopy, Video , Muscle, Skeletal/blood supply , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Transplantation Chimera
16.
Br J Haematol ; 160(4): 530-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23240761

ABSTRACT

Elevated levels of circulating tissue factor-bearing microparticles (TFMP) have been associated with an increased risk of developing venous thromboembolism (VTE) in cancer patients. We performed a randomized phase II study to evaluate the cumulative incidence of VTE in advanced cancer patients with lower levels of TFMP not receiving thromboprophylaxis and those with higher levels of circulating TFMP randomized to enoxaparin or observation. The cumulative incidence of VTE at 2 months in the higher TFMP group randomized to enoxaparin (N = 23) was 5·6% while the higher TFMP group observation arm (N = 11) was 27·3% (Gray test P = 0·06). The cumulative incidence of VTE in the low TFMP was 7·2% (N = 32). No major haemorrhages were observed in the enoxaparin arm. The median survival for patients with higher levels of TFMP followed by observation was 11·8 months compared with 17·8 months on enoxaparin (P = 0·58). In a prospective randomized trial, increased numbers of circulating TFMP detected by impedance flow cytometry identified cancer patients with a high incidence of VTE. Enoxaparin demonstrated a clear trend towards reducing the rate of VTE in patients with elevated levels of TFMP, with an overall rate of VTE similar in magnitude to the lower TFMP group.


Subject(s)
Anticoagulants/therapeutic use , Enoxaparin/therapeutic use , Neoplasms/complications , Thromboembolism/prevention & control , Thromboplastin/metabolism , Aged , Aged, 80 and over , Cell-Derived Microparticles/metabolism , Female , Fibrin Fibrinogen Degradation Products/metabolism , Humans , Male , Middle Aged , Survival Analysis
17.
Blood ; 117(12): 3453-9, 2011 Mar 24.
Article in English | MEDLINE | ID: mdl-21245481

ABSTRACT

Antiphospholipid syndrome is characterized by thrombosis, recurrent fetal loss, and the presence of the lupus anticoagulant, anticardiolipin antibodies, or anti-ß(2)-glycoprotein-1 (anti-ß(2)-GP1) antibodies. Although anti-ß(2)-GP1 antibodies have been documented as a biomarker for diagnosis of antiphospholipid syndrome, their direct role in the pathogenesis of thrombosis is unknown. We have demonstrated using intravital microscopy that anti-ß(2)-GP1 autoantibodies purified from the sera of patients with antiphospholipid syndrome complicated by thrombosis greatly amplify thrombus size after laser-induced vessel wall injury in live mice. Anti-ß(2)-GP1 autoantibodies from 3 patients with antiphospholipid syndrome were affinity-purified using human ß(2)-GP1 bound to agarose. The effects of purified anti-ß(2)-GP1 IgG autoantibodies, of anti-ß(2)-GP1-depleted IgG, and of IgG from normal human sera on thrombus formation were measured in mice after arterial injury in the cremaster muscle. Before injury, purified anti-ß(2)-GP1 IgG autoantibodies, anti-ß(2)-GP1 antibody-depleted IgG, or IgG from normal human sera were infused. Increasing amounts of purified anti-ß(2)-GP1 autoantibodies increased thrombus size in a dose-dependent manner, whereas neither anti-ß(2)-GP1 antibody-depleted IgG nor IgG from normal serum affected thrombus size. These results indicate that anti-ß(2)-GP1 IgG autoantibodies in antiphospholipid syndrome patient sera are not only a marker of antiphospholipid syndrome but are directly involved in the pathogenesis of thrombosis.


Subject(s)
Antiphospholipid Syndrome/immunology , Autoantibodies/adverse effects , Disease Models, Animal , Thrombosis/etiology , beta 2-Glycoprotein I/immunology , Adult , Animals , Antiphospholipid Syndrome/blood , Arteries/pathology , Autoantibodies/isolation & purification , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Thrombosis/metabolism , Thrombosis/pathology , beta 2-Glycoprotein I/metabolism
18.
J Biol Chem ; 286(9): 7027-32, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21199867

ABSTRACT

Plasminogen activator inhibitor-1 (PAI-1), together with its physiological target urokinase-type plasminogen activator (uPA), plays a pivotal role in fibrinolysis, cell migration, and tissue remodeling and is currently recognized as being among the most extensively validated biological prognostic factors in several cancer types. PAI-1 specifically and rapidly inhibits uPA and tissue-type PA (tPA). Despite extensive structural/functional studies on these two reactions, the underlying structural mechanism has remained unknown due to the technical difficulties of obtaining the relevant structures. Here, we report a strategy to generate a PAI-1·uPA(S195A) Michaelis complex and present its crystal structure at 2.3-Å resolution. In this structure, the PAI-1 reactive center loop serves as a bait to attract uPA onto the top of the PAI-1 molecule. The P4-P3' residues of the reactive center loop interact extensively with the uPA catalytic site, accounting for about two-thirds of the total contact area. Besides the active site, almost all uPA exosite loops, including the 37-, 60-, 97-, 147-, and 217-loops, are involved in the interaction with PAI-1. The uPA 37-loop makes an extensive interaction with PAI-1 ß-sheet B, and the 147-loop directly contacts PAI-1 ß-sheet C. Both loops are important for initial Michaelis complex formation. This study lays down a foundation for understanding the specificity of PAI-1 for uPA and tPA and provides a structural basis for further functional studies.


Subject(s)
Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 1/metabolism , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/metabolism , Catalytic Domain , Crystallography , Enzyme Activation/physiology , Humans , Mutation , Pichia/genetics , Plasminogen Activator Inhibitor 1/genetics , Protein Structure, Tertiary , Structure-Activity Relationship , Urokinase-Type Plasminogen Activator/genetics
19.
J Biol Chem ; 286(26): 23345-56, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21531712

ABSTRACT

Mouse and human prothrombin (ProT) active site specifically labeled with D-Phe-Pro-Arg-CH(2)Cl (FPR-ProT) inhibited tissue factor-initiated thrombin generation in platelet-rich and platelet-poor mouse and human plasmas. FPR-prethrombin 1 (Pre 1), fragment 1 (F1), fragment 1.2 (F1.2), and FPR-thrombin produced no significant inhibition, demonstrating the requirement for all three ProT domains. Kinetics of inhibition of ProT activation by the inactive ProT(S195A) mutant were compatible with competitive inhibition as an alternate nonproductive substrate, although FPR-ProT deviated from this mechanism, implicating a more complex process. FPR-ProT exhibited ∼10-fold more potent anticoagulant activity compared with ProT(S195A) as a result of conformational changes in the ProT catalytic domain that induce a more proteinase-like conformation upon FPR labeling. Unlike ProT and ProT(S195A), the pathway of FPR-ProT cleavage by prothrombinase was redirected from meizothrombin toward formation of the FPR-prethrombin 2 (Pre 2)·F1.2 inhibitory intermediate. Localization of ProT labeled with Alexa Fluor® 660 tethered through FPR-CH(2)Cl ([AF660]FPR-ProT) during laser-induced thrombus formation in vivo in murine arterioles was examined in real time wide-field and confocal fluorescence microscopy. [AF660]FPR-ProT bound rapidly to the vessel wall at the site of injury, preceding platelet accumulation, and subsequently to the thrombus proximal, but not distal, to the vessel wall. [AF660]FPR-ProT inhibited thrombus growth, whereas [AF660]FPR-Pre 1, lacking the F1 membrane-binding domain did not bind or inhibit. Labeled F1.2 localized similarly to [AF660]FPR-ProT, indicating binding to phosphatidylserine-rich membranes, but did not inhibit thrombosis. The studies provide new insight into the mechanism of ProT activation in vivo and in vitro, and the properties of a unique exosite-directed prothrombinase inhibitor.


Subject(s)
Catalytic Domain , Prothrombin/metabolism , Thromboplastin/metabolism , Thrombosis/enzymology , Amino Acid Substitution , Animals , Blood Coagulation , Enzyme Activation/genetics , Humans , Kinetics , Mice , Mutation, Missense , Protein Structure, Tertiary , Prothrombin/chemistry , Prothrombin/genetics , Thromboplastin/chemistry , Thromboplastin/genetics , Thrombosis/genetics
20.
Blood ; 116(22): 4665-74, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20668226

ABSTRACT

Protein disulfide isomerase (PDI) catalyzes the oxidation reduction and isomerization of disulfide bonds. We have previously identified an important role for extracellular PDI during thrombus formation in vivo. Here, we show that endothelial cells are a critical cellular source of secreted PDI, important for fibrin generation and platelet accumulation in vivo. Functional PDI is rapidly secreted from human umbilical vein endothelial cells in culture upon activation with thrombin or after laser-induced stimulation. PDI is localized in different cellular compartments in activated and quiescent endothelial cells, and is redistributed to the plasma membrane after cell activation. In vivo studies using intravital microscopy show that PDI appears rapidly after laser-induced vessel wall injury, before the appearance of the platelet thrombus. If platelet thrombus formation is inhibited by the infusion of eptifibatide into the circulation, PDI is detected after vessel wall injury, and fibrin deposition is normal. Treatment of mice with a function blocking anti-PDI antibody completely inhibits fibrin generation in eptifibatide-treated mice. These results indicate that, although both platelets and endothelial cells secrete PDI after laser-induced injury, PDI from endothelial cells is required for fibrin generation in vivo.


Subject(s)
Blood Platelets/metabolism , Endothelial Cells/metabolism , Protein Disulfide-Isomerases/metabolism , Thrombosis/metabolism , Animals , Cell Line , Cytosol/ultrastructure , Endothelial Cells/cytology , Endothelial Cells/ultrastructure , Endothelium/metabolism , Fibrin/metabolism , Humans , Mice , Mice, Inbred C57BL , Protein Disulfide-Isomerases/analysis
SELECTION OF CITATIONS
SEARCH DETAIL