Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Thromb Haemost ; 21(3): 652-666, 2023 03.
Article in English | MEDLINE | ID: mdl-36696211

ABSTRACT

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is a serious thrombotic disorder caused by ultralarge immune complexes (ULICs) containing platelet factor 4 (PF4) and heparin that form the HIT antigen, together with a subset of anti-PF4 antibodies. ULICs initiate prothrombotic responses by engaging Fcγ receptors on platelets, neutrophils, and monocytes. Contemporary anti-thrombotic therapy for HIT is neither entirely safe nor entirely successful and acts downstream of ULIC formation and Fcγ receptor-initiated generation of thrombin. OBJECTIVES: To determine whether HIT antigen and ULIC formation and stability could be modified favorably by inhibiting PF4-heparin interactions with fondaparinux, together with blocking formation of PF4 tetramers using a humanized monoclonal anti-PF4 antibody (hRTO). METHODS: Results: The combination of fondaparinux and hRTO inhibited HIT antigen formation, promoted antigen dissociation, inhibited ULIC formation, and promoted ULIC disassembly at concentrations below the effective concentration of either alone and blocked Fcγ receptor-dependent induction of factor Xa activity by monocytic THP1 cells and activation of human platelets in whole blood. Combined with hRTO, fondaparinux inhibited HIT antigen and immune complex formation and activation through Fcγ receptors at concentrations at or below those used clinically to inhibit FXa coagulant activity. CONCLUSIONS: HIT antigen and immune complexes are dynamic and amenable to modulation. Fondaparinux can be converted from an anticoagulant that acts at a downstream amplification step into a rationale, disease-specific intervention that blocks ULIC formation. Interventions that prevent ULIC formation and stability might increase the efficacy, permit use of lower doses, shorten the duration of antithrombotic therapy, and help prevent this serious thrombotic disorder.


Subject(s)
Thrombocytopenia , Thrombosis , Humans , Antibodies, Monoclonal, Humanized/adverse effects , Anticoagulants/adverse effects , Antigen-Antibody Complex , Fondaparinux/adverse effects , Heparin/adverse effects , Platelet Factor 4 , Receptors, IgG , Thrombosis/etiology
2.
Blood ; 138(21): 2106-2116, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34189574

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is a prothrombotic disorder mediated by ultra-large immune complexes (ULICs) containing immunoglobulin G (IgG) antibodies to a multivalent antigen composed of platelet factor 4 and heparin. The limitations of current antithrombotic therapy in HIT supports the need to identify additional pathways that may be targets for therapy. Activation of FcγRIIA by HIT ULICs initiates diverse procoagulant cellular effector functions. HIT ULICs are also known to activate complement, but the contribution of this pathway to the pathogenesis of HIT has not been studied in detail. We observed that HIT ULICs physically interact with C1q in buffer and plasma, activate complement via the classical pathway, promote codeposition of IgG and C3 complement fragments (C3c) on neutrophil and monocyte cell surfaces. Complement activation by ULICs, in turn, facilitates FcγR-independent monocyte tissue factor expression, enhances IgG binding to the cell surface FcγRs, and promotes platelet adhesion to injured endothelium. Inhibition of the proximal, but not terminal, steps in the complement pathway abrogates monocyte tissue factor expression by HIT ULICs. Together, these studies suggest a major role for complement activation in regulating Fc-dependent effector functions of HIT ULICs, identify potential non-anticoagulant targets for therapy, and provide insights into the broader roles of complement in immune complex-mediated thrombotic disorders.


Subject(s)
Anticoagulants/adverse effects , Antigen-Antibody Complex/immunology , Complement Activation , Heparin/adverse effects , Thrombocytopenia/chemically induced , Anticoagulants/immunology , Complement C3/immunology , Heparin/immunology , Humans , Immunoglobulin G/immunology , Platelet Factor 4/immunology , Receptors, IgG/immunology , Thrombocytopenia/complications , Thrombocytopenia/immunology , Thrombosis/etiology , Thrombosis/immunology
3.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L54-L68, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28860148

ABSTRACT

Elevated active plasminogen activator inhibitor-1 (PAI-1) has an adverse effect on the outcomes of intrapleural fibrinolytic therapy (IPFT) in tetracycline-induced pleural injury in rabbits. To enhance IPFT with prourokinase (scuPA), two mechanistically distinct approaches to targeting PAI-1 were tested: slowing its reaction with urokinase (uPA) and monoclonal antibody (mAb)-mediated PAI-1 inactivation. Removing positively charged residues at the "PAI-1 docking site" (179RHRGGS184→179AAAAAA184) of uPA results in a 60-fold decrease in the rate of inhibition by PAI-1. Mutant prourokinase (0.0625-0.5 mg/kg; n = 12) showed efficacy comparable to wild-type scuPA and did not change IPFT outcomes ( P > 0.05). Notably, the rate of PAI-1-independent intrapleural inactivation of mutant uPA was 2 times higher ( P < 0.05) than that of the wild-type enzyme. Trapping PAI-1 in a "molecular sandwich"-type complex with catalytically inactive two-chain urokinase with Ser195Ala substitution (S195A-tcuPA; 0.1 and 0.5 mg/kg) did not improve the efficacy of IPFT with scuPA (0.0625-0.5 mg/kg; n = 11). IPFT failed in the presence of MA-56A7C10 (0.5 mg/kg; n = 2), which forms a stable intrapleural molecular sandwich complex, allowing active PAI-1 to accumulate by blocking its transition to a latent form. In contrast, inactivation of PAI-1 by accelerating the active-to-latent transition mediated by mAb MA-33B8 (0.5 mg/kg; n = 2) improved the efficacy of IPFT with scuPA (0.25 mg/kg). Thus, under conditions of slow (4-8 h) fibrinolysis in tetracycline-induced pleural injury in rabbits, only the inactivation of PAI-1, but not a decrease in the rate of its reaction with uPA, enhances IPFT. Therefore the rate of fibrinolysis, which varies in different pathologic states, could affect the selection of PAI-1 inhibitors to enhance fibrinolytic therapy.


Subject(s)
Fibrinolysis/drug effects , Fibrinolytic Agents/pharmacology , Plasminogen Activator Inhibitor 1/chemistry , Pleural Diseases/drug therapy , Tetracycline/toxicity , Thrombolytic Therapy/methods , Animals , Disease Models, Animal , Female , Plasminogen Activator Inhibitor 1/metabolism , Pleural Diseases/chemically induced , Protein Synthesis Inhibitors/toxicity , Rabbits
4.
Blood Adv ; 1(1): 62-74, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-29296696

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is a thrombotic disorder initiated by antibodies to complexes between platelet factor 4 (PF4) and heparin. The risk of recurrent thromboembolism persists after heparin is cleared and platelet activation leading to release of PF4 has dissipated. We asked whether antigenic complexes between polyphosphates and PF4 released from activated platelets might intensify or sustain the prothrombotic phenotype of HIT. PF4 forms stable, ultralarge complexes with polyphosphates of various sizes, including those released from platelets, which are recognized by the HIT-like monoclonal KKO, an immunoglobulin G2bκ monoclonal heparin/PF4 binding antibody, and by human HIT antibodies. KKO helps to protect PF4/polyphosphate complexes from degradation by phosphatases. Complement is activated when HIT antibodies bind to PF4/polyphosphate complexes and PF4 reverses the inhibition of complement by polyphosphates. Polyphosphates and PF4 are stored primarily in separate granules in resting platelets, but they colocalize when the cells are activated. Platelets activated by subaggregating doses of thrombin receptor activating peptide release polyphosphates and PF4, which form antigenic complexes that allow KKO to further activate platelets in the absence of heparin and exogenous PF4. These studies suggest that thrombin- or immune complex-mediated release of endogenous antigenic PF4/polyphosphate complexes from platelets may augment the prothrombotic risk of HIT and perpetuate the risk of thrombosis after heparin has been discontinued.

5.
Nat Commun ; 6: 8277, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26391892

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is an autoimmune thrombotic disorder caused by immune complexes containing platelet factor 4 (PF4), antibodies to PF4 and heparin or cellular glycosaminoglycans (GAGs). Here we solve the crystal structures of the: (1) PF4 tetramer/fondaparinux complex, (2) PF4 tetramer/KKO-Fab complex (a murine monoclonal HIT-like antibody) and (3) PF4 monomer/RTO-Fab complex (a non-HIT anti-PF4 monoclonal antibody). Fondaparinux binds to the 'closed' end of the PF4 tetramer and stabilizes its conformation. This interaction in turn stabilizes the epitope for KKO on the 'open' end of the tetramer. Fondaparinux and KKO thereby collaborate to 'stabilize' the ternary pathogenic immune complex. Binding of RTO to PF4 monomers prevents PF4 tetramerization and inhibits KKO and human HIT IgG-induced platelet activation and platelet aggregation in vitro, and thrombus progression in vivo. The atomic structures provide a basis to develop new diagnostics and non-anticoagulant therapeutics for HIT.


Subject(s)
Anticoagulants/adverse effects , Autoimmune Diseases/chemically induced , Heparin/adverse effects , Thrombocytopenia/chemically induced , Animals , Antibodies/immunology , Antigen-Antibody Complex , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Glycosaminoglycans/immunology , Glycosaminoglycans/metabolism , Heparin/immunology , Humans , Immunoglobulin G/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Molecular , Platelet Activation , Platelet Factor 4/immunology , Platelet Factor 4/metabolism , Protein Conformation , Thrombocytopenia/immunology , Thrombocytopenia/pathology
6.
J Biol Chem ; 288(46): 33060-70, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24097975

ABSTRACT

Heparin-induced thrombocytopenia (HIT) is a thrombotic complication of heparin therapy mediated by antibodies to complexes between platelet factor 4 (PF4) and heparin or cellular glycosaminoglycans. However, only a fraction of patients with anti-PF4-heparin antibodies develop HIT, implying that only a subset of these antibodies is pathogenic. The basis for the pathogenic potential of anti-PF4-heparin antibodies remains unclear. To elucidate the intrinsic PF4-binding properties of HIT-like monoclonal antibody (KKO) versus non-pathogenic antibody (RTO) at the single-molecule level, we utilized optical trap-based force spectroscopy to measure the strength and probability of binding of surface-attached antibodies with oligomeric PF4 to simulate interactions on cells. To mimic the effect of heparin in bringing PF4 complexes into proximity, we chemically cross-linked PF4 tetramers using glutaraldehyde. Analysis of the force histograms revealed that KKO-PF4 interactions had ∼10-fold faster on-rates than RTO-PF4, and apparent equilibrium dissociation constants differed ∼10-fold with similar force-free off-rates (k(off) = 0.0031 and 0.0029 s(-1)). Qualitatively similar results were obtained for KKO and RTO interacting with PF4-heparin complexes. In contrast to WT PF4, KKO and RTO showed lower and similar binding probabilities to cross-linked PF4(K50E), which forms few if any oligomers. Thus, formation of stable PF4 polymers results in much stronger interactions with the pathogenic antibody without a significant effect on the binding of the non-pathogenic antibody. These results suggest a fundamental difference in the antigen-binding mechanisms between model pathogenic and non-pathogenic anti-PF4 antibodies that might underlie their distinct pathophysiological behaviors.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibody Specificity , Anticoagulants/chemistry , Autoantibodies/chemistry , Heparin/chemistry , Platelet Factor 4/chemistry , Antibodies, Monoclonal/immunology , Anticoagulants/adverse effects , Anticoagulants/immunology , Anticoagulants/therapeutic use , Autoantibodies/immunology , Binding Sites, Antibody , Heparin/adverse effects , Heparin/immunology , Heparin/therapeutic use , Humans , Kinetics , Platelet Factor 4/immunology , Thrombocytopenia/chemically induced , Thrombocytopenia/immunology
7.
Blood ; 121(18): 3727-32, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23446735

ABSTRACT

Laboratory testing for heparin-induced thrombocytopenia (HIT) has important shortcomings. Immunoassays fail to discriminate platelet-activating from nonpathogenic antibodies. Specific functional assays are impracticable due to the need for platelets and radioisotope. We describe 2 assays that may overcome these limitations. The KKO-inhibition test (KKO-I) measures the effect of plasma on binding of the HIT-like monoclonal antibody KKO to platelet factor 4 (PF4)/heparin. DT40-luciferase (DT40-luc) is a functional test comprised of a B-cell line expressing FcγRIIa coupled to a luciferase reporter. We compared these assays to polyspecific and immunoglobulin (Ig)G-specific PF4/heparin enzyme-linked immunosorbent assays (ELISAs) in samples from 58 patients with suspected HIT and circulating anti-PF4/heparin antibodies. HIT was defined as a 4Ts score ≥ 4 and positive (14)C-serotonin release assay. HIT-positive plasma demonstrated greater mean inhibition of KKO binding than HIT-negative plasma (78.9% vs 26.0%; P < .0001) and induced greater luciferase activity (3.14-fold basal vs 0.96-fold basal; P < .0001). The area under the receiver-operating characteristic curve was greater for KKO-I (0.93) than for the polyspecific (0.82; P = .020) and IgG-specific ELISA (0.76; P = .0044) and for DT40-luc (0.89) than for the IgG-specific ELISA (P = .046). KKO-I and DT40-luc showed better discrimination than 2 commercially available immunoassays, are simple to perform, and hold promise for improving the specificity and feasibility of HIT laboratory testing.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Heparin/adverse effects , Thrombocytopenia/chemically induced , Thrombocytopenia/diagnosis , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/immunology , Cells, Cultured , Female , Hematologic Tests , Humans , Male , Middle Aged
8.
Blood ; 120(5): 1137-42, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22577175

ABSTRACT

Rapid laboratory assessment of heparin-induced thrombocytopenia (HIT) is important for disease recognition and management. The utility of contemporary immunoassays to detect antiplatelet factor 4 (PF4)/heparin antibodies is hindered by detection of antibodies unassociated with disease. To begin to distinguish properties of pathogenic anti-PF4/heparin antibodies, we compared isotype-matched monoclonal antibodies that bind to different epitopes: KKO causes thrombocytopenia in an in vivo model of HIT, whereas RTO does not. KKO binding to PF4 and heparin is specifically inhibited by human HIT antibodies that activate platelets, whereas inhibition of RTO binding is not differentially affected. Heparin increased the avidity of KKO binding to PF4 without affecting RTO, but it did not increase total binding or binding to nontetrameric PF4(K50E). Single-molecule forced unbinding demonstrated KKO was 8-fold more reactive toward PF4 tetramers and formed stronger complexes than RTO, but not to PF4(K50E) dimers. KKO, but not RTO, promoted oligomerization of PF4 but not PF4(K50E). This study reveals differences in the properties of anti-PF4 antibodies that cause thrombocytopenia not revealed by ELISA that correlate with oligomerization of PF4 and sustained high-avidity interactions that may simulate transient antibody-antigen interactions in vivo. These differences suggest the potential importance of epitope specificity in the pathogenesis of HIT.


Subject(s)
Antibodies/metabolism , Antibody Affinity/physiology , Thrombocytopenia/etiology , Thrombocytopenia/immunology , Animals , Antibodies, Immobilized/metabolism , Antibodies, Monoclonal/metabolism , Antibody Affinity/genetics , Antigen-Antibody Reactions/genetics , Antigen-Antibody Reactions/physiology , Cell Line, Tumor , Drosophila , Enzyme-Linked Immunosorbent Assay , Heparin/adverse effects , Humans , Kinetics , Mice , Mice, Transgenic , Platelet Factor 4/genetics , Platelet Factor 4/immunology , Protein Binding/physiology , Thrombocytopenia/genetics , Thrombocytopenia/pathology
9.
Blood ; 119(25): 5955-62, 2012 Jun 21.
Article in English | MEDLINE | ID: mdl-22452981

ABSTRACT

Patients with heparin-induced thrombocytopenia (HIT) remain at risk for recurrent thromboembolic complications despite improvements in management. HIT is caused by antibodies that preferentially recognize ultralarge complexes (ULCs) of heparin and platelet factor 4 (PF4) tetramers. We demonstrated previously that a variant PF4(K50E) forms dimers but does not tetramerize or form ULCs. Here, we identified small molecules predicted to bind PF4 near the dimer-dimer interface and that interfere with PF4 tetramerization. Screening a library of small molecules in silico for binding at this site, we identified 4 compounds that inhibited tetramerization at micromolar concentrations, designated PF4 antagonists (PF4As). PF4As also inhibited formation of pathogenic ULCs, and 3 of these PF4As promoted the breakdown of preformed ULCs. To characterize the ability of PF4As to inhibit cellular activation, we developed a robust and reproducible assay that measures cellular activation by HIT antibodies via FcγRIIA using DT40 cells. PF4As inhibit FcγRIIA-dependent activation of DT40 cells by HIT antibodies as well as platelet activation, as measured by serotonin release. PF4As provide new tools to probe the pathophysiology of HIT. They also may provide insight into the development of novel, disease-specific therapeutics for the treatment of thromboembolic complications in HIT.


Subject(s)
Anticoagulants/isolation & purification , Anticoagulants/therapeutic use , Drug Discovery/methods , Platelet Factor 4/antagonists & inhibitors , Thrombocytopenia/drug therapy , Animals , Anticoagulants/chemistry , Cells, Cultured , Chickens , Computational Biology , Drosophila , Drug Evaluation, Preclinical , Heparin/adverse effects , Heparin/therapeutic use , Humans , Models, Biological , Models, Molecular , Rationalization , Research Design , Thrombocytopenia/chemically induced
10.
Thromb Haemost ; 95(3): 524-34, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16525582

ABSTRACT

It has been questioned whether there are receptors for urokinase-type plasminogen activator (uPA) that facilitate plasminogen activation other than the high affinity uPA receptor (uPAR/CD87) since studies of uPAR knockout mice did not support a major role of uPAR in plasminogen activation. uPA also promotes cell adhesion, chemotaxis, and proliferation besides plasminogen activation. These uPA-induced signaling events are not mediated by uPAR, but mediated by unidentified, lower-affinity receptors for the uPA kringle. We found that uPA binds specifically to integrin alpha v beta 3 on CHO cells depleted of uPAR. The binding of uPA to alpha v beta 3 required the uPA kringle domain. The isolated uPA kringle domain binds specifically to purified, recombinant soluble, and cell surface alpha v beta 3, and other integrins (alpha 4 beta 1 and alpha 9 beta 1), and induced migration of CHO cells in an alpha v beta 3-dependent manner. The binding of the uPA kringle to alpha v beta 3 and uPA kringle-induced alpha v beta 3-dependent cell migration were blocked by homologous plasminogen kringles 1-3 or 1-4 (angiostatin), a known integrin antagonist. We studied whether the binding of uPA to integrin alpha v beta 3 through the kringle domain plays a role in plasminogen activation. On CHO cell depleted of uPAR, uPA enhanced plasminogen activation in a kringle and alpha v beta 3-dependent manner. Endothelial cells bound to and migrated on uPA and uPA kringle in an alpha v beta 3-dependent manner. These results suggest that uPA binding to integrins through the kringle domain plays an important role in both plasminogen activation and uPA-induced intracellular signaling. The uPA kringle-integrin interaction may represent a novel therapeutic target for cancer, inflammation, and vascular remodeling.


Subject(s)
Integrin alphaVbeta3/metabolism , Kringles , Plasminogen/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Angiogenesis Inhibitors/pharmacology , Angiostatins/pharmacology , Animals , CHO Cells , Cell Adhesion , Cell Membrane/metabolism , Cell Movement , Cricetinae , Cricetulus , Endothelial Cells/drug effects , Endothelial Cells/ultrastructure , Integrin alphaVbeta3/drug effects , Integrin alphaVbeta3/genetics , Receptors, Cell Surface/deficiency , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...