Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 188
Filter
1.
Nat Commun ; 11(1): 2992, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32532974

ABSTRACT

Activated protein C (APC) is a plasma serine protease with antithrombotic and cytoprotective functions. Based on the hypothesis that specific inhibition of APC's anticoagulant but not its cytoprotective activity can be beneficial for hemophilia therapy, 2 types of inhibitory monoclonal antibodies (mAbs) are tested: A type I active-site binding mAb and a type II mAb binding to an exosite on APC (required for anticoagulant activity) as shown by X-ray crystallography. Both mAbs increase thrombin generation and promote plasma clotting. Type I blocks all APC activities, whereas type II preserves APC's cytoprotective function. In normal monkeys, type I causes many adverse effects including animal death. In contrast, type II is well-tolerated in normal monkeys and shows both acute and prophylactic dose-dependent efficacy in hemophilic monkeys. Our data show that the type II mAb can specifically inhibit APC's anticoagulant function without compromising its cytoprotective function and offers superior therapeutic opportunities for hemophilia.


Subject(s)
Antibodies, Monoclonal/pharmacology , Hemophilia A/prevention & control , Immunoglobulin Fab Fragments/immunology , Protein C Inhibitor/pharmacology , Protein C/antagonists & inhibitors , Animals , Antibodies, Monoclonal/classification , Antibodies, Monoclonal/immunology , Bleeding Time , Cell Membrane Permeability/drug effects , Cells, Cultured , Crystallography, X-Ray , Hemophilia A/blood , Hemorrhage/prevention & control , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Immunoglobulin Fab Fragments/metabolism , Macaca fascicularis , Male , Protein C/chemistry , Protein C/immunology , Protein C/metabolism , Protein C Inhibitor/blood , Protein C Inhibitor/pharmacokinetics
2.
Int Immunopharmacol ; 81: 106255, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32007797

ABSTRACT

BACKGROUND: Endothelial protein C receptor (EPCR) is a membranous protein that can be combined with a variety of ligands and plays important roles in anticoagulant and anti-inflammation. Recent reports have shown that surface EPCR expression on T cells is negatively associated with Th17 differentiation and is co-expressed with other immunosuppressive molecules, such as The programmed cell death 1 (PD-1) and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4). Hence, we hypothesized that EPCR may play a critical role in rheumatoid arthritis (RA) disease progression that is mediated by Th17 differentiation. In order to explore the role of EPCR on RA disease pathogenesis, we detected membranous EPCR (mEPCR) expression in CD4+ T cells and soluble EPCR (sEPCR) expression in the sera of RA patients. METHODS: The proportion of CD4+/EPCR+ T cells in the peripheral blood of RA patients was detected by flow cytometry, and the expression of sEPCR in the sera of RA patients was detected by enzyme-linked immunosorbent assay (ELISA). For in vitro experiments, protein C (PC) and EPCR recombinant proteins were used to block peripheral blood mononuclear cell (PBMC) activation and to detect Th17 differentiation. For in vivo experiments in DBA/1 mice with collagen-induced arthritis (CIA), we administered PC and EPCR recombinant proteins, monitored disease progression, and evaluated the role of EPCR in disease progression. RESULTS: The proportion of CD4+/EPCR+ T cells in the peripheral blood of RA patients was lower than that of osteoarthritis (OA) patients, while the expression level of sEPCR in the sera of RA patients was concomitantly higher than that in OA patients. Subsequent analysis revealed that sEPCR expression was positively correlated with rheumatoid factors (RF) and other inflammatory indicators in RA patients. Further studies confirmed that sEPCR administration alleviated the progression of collagen-induced arthritis and partially blocked the therapeutic effect of PC in CIA mice. CONCLUSION: Soluble EPCR is associated with RA disease progression and induces disease remission in CIA mice by inhibiting Th17 differentiation.


Subject(s)
Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Endothelial Protein C Receptor/metabolism , Osteoarthritis/metabolism , Th17 Cells/immunology , Adult , Aged , Animals , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/pathology , CTLA-4 Antigen/metabolism , Cell Differentiation , Endothelial Protein C Receptor/genetics , Female , Gene Expression Regulation , Humans , Male , Mice, Inbred DBA , Middle Aged , Osteoarthritis/pathology , Programmed Cell Death 1 Receptor/metabolism , Protein C/antagonists & inhibitors , Protein C/metabolism
3.
Blood ; 133(19): 2090-2099, 2019 05 09.
Article in English | MEDLINE | ID: mdl-30898865

ABSTRACT

Tissue factor, coagulation factor XII, platelets, and neutrophils are implicated as important players in the pathophysiology of (experimental) venous thrombosis (VT). Their role became evident in mouse models in which surgical handlings were required to provoke VT. Combined inhibition of the natural anticoagulants antithrombin (Serpinc1) and protein C (Proc) using small interfering RNA without additional triggers also results in a venous thrombotic phenotype in mice, most notably with vessel occlusion in large veins of the head. VT is fatal but is fully rescued by thrombin inhibition. In the present study, we used this VT mouse model to investigate the involvement of tissue factor, coagulation factor XII, platelets, and neutrophils. Antibody-mediated inhibition of tissue factor reduced the clinical features of VT, the coagulopathy in the head, and fibrin deposition in the liver. In contrast, genetic deficiency in, and small interfering RNA-mediated depletion of, coagulation factor XII did not alter VT onset, severity, or thrombus morphology. Antibody-mediated depletion of platelets fully abrogated coagulopathy in the head and liver fibrin deposition. Although neutrophils were abundant in thrombotic lesions, depletion of circulating Ly6G-positive neutrophils did not affect onset, severity, thrombus morphology, or liver fibrin deposition. In conclusion, VT after inhibition of antithrombin and protein C is dependent on the presence of tissue factor and platelets but not on coagulation factor XII and circulating neutrophils. This study shows that distinct procoagulant pathways operate in mouse VT, dependent on the triggering stimulus.


Subject(s)
Blood Platelets/metabolism , Factor XII/metabolism , Neutrophils/metabolism , Thromboplastin/metabolism , Venous Thrombosis/blood , Animals , Antithrombin III/antagonists & inhibitors , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Protein C/antagonists & inhibitors
4.
Sci Rep ; 8(1): 8793, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29884816

ABSTRACT

Activated protein C (APC) is a powerful anticoagulant enzyme that proteolytically inactivates the cofactors of the Xase and prothrombinase complexes, factors VIIIa and Va. A common mutation in factor V, fVLeiden, confers resistance to APC leading to an increased risk of thrombosis in the normal population. However, when coinherited with haemophilia, fVLeiden reduces bleeding severity, suggesting that inhibition of APC may be a useful strategy for treatment of haemophilia. We previously reported on serpins that were rationally designed for improved specificity for APC over other coagulation serine proteases. Based on structural differences in the substrate binding pockets to either side of the P1 Arg, we mutated the P2 and P1' residues to Lys. Although this approach achieved APC specificity, it resulted in a reduction in the rate of APC inhibition relative to the parent containing only the P1 Arg. Here we conduct site-specific random mutagenesis at the P2 and P1' positions to determine if improvements could be made in the rate of APC inhibition. In addition to our original Lys mutations, we found that Arg and Gln also confer specificity for APC. However, in all cases specificity for APC resulted in a reduction in inhibition rate.


Subject(s)
Mutagenesis, Site-Directed , Protein C/antagonists & inhibitors , Serpins/genetics , Serpins/pharmacology , Binding Sites , Blood Coagulation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Protein C/metabolism , Protein Conformation , Serpins/chemistry , alpha 1-Antitrypsin/chemistry , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/pharmacology
5.
Thromb Haemost ; 118(4): 630-638, 2018 04.
Article in English | MEDLINE | ID: mdl-29448296

ABSTRACT

BACKGROUND: Activated protein C (APC) is a major regulator of thrombin formation. Two major plasma inhibitors form complexes with APC, protein C inhibitor (PCI) and α1-antitrypsin (α1AT), and these complexes have been quantified by specific enzyme-linked immunosorbent assays (ELISAs). Also, complexes of APC with α2-macroglobulin (α2M) have been observed by immunoblotting. Here, we report an ELISA for APC:α2M complexes in plasma. METHODS: Plasma samples were pre-treated with dithiothreitol and then with iodoacetamide. The detection range of the newly developed APC:α2M assay was 0.031 to 8.0 ng/mL of complexed APC. Following infusions of APC in humans and baboons, complexes of APC with α2M, PCI and α1AT were quantified. These complexes as well as circulating APC were also measured in 121 patients with a history of venous thromboembolism (VTE) and 119 matched controls. RESULTS: In all the in vivo experiments, α2M was a significant APC inhibitor. The VTE case-control study showed that VTE patients had significantly lower APC:α2M and APC levels than the controls (p < 0.001). Individuals in the lowest quartile of APC:α2M or the lowest quartile of APC had approximately four times more VTE risk than those in the highest quartile of APC:α2M or of APC. The risk increased for individuals with low levels of both parameters. CONCLUSION: The APC:α2M assay reported here may be useful to help monitor the in vivo fate of APC in plasma. In addition, our results show that a low APC:α2M level is associated with increased VTE risk.


Subject(s)
Pregnancy-Associated alpha 2-Macroglobulins/metabolism , Protein C/antagonists & inhibitors , Protein C/metabolism , Venous Thromboembolism/blood , alpha 1-Antitrypsin/metabolism , Adult , Animals , Case-Control Studies , Dithiothreitol/therapeutic use , Enzyme Inhibitors/therapeutic use , Enzyme-Linked Immunosorbent Assay , Female , Humans , Iodoacetamide/therapeutic use , Limit of Detection , Lung , Male , Middle Aged , Papio , Protein C/therapeutic use , Protein C Inhibitor/therapeutic use , Recombinant Proteins/therapeutic use
6.
Curr Opin Hematol ; 24(5): 446-452, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28632502

ABSTRACT

PURPOSE OF REVIEW: Hemophilia is a debilitating disease, marked by frequent, painful bleeding events, joint deterioration and early death. All current treatments consist of i.v. infusions of replacement factor or other procoagulant factors, and are incompletely effective, due in part to the short half-lives of the proteins. An alternative approach is to rebalance hemostasis by inhibiting natural anticoagulant mechanisms. In this article, we explain why activated protein C (APC) is an appropriate and safe target for the treatment of hemophilia. RECENT FINDINGS: A serpin (serine protease inhibitor) was engineered to specifically inhibit APC and was found to rescue hemostasis in a hemophilia mouse model, even after a severe tail clip injury. However, APC is also anti-inflammatory and has cytoprotective activities, raising safety concerns over the use of an APC inhibitor to treat hemophilia. We summarize the molecular basis of the anticoagulant and signaling activities of APC to assess the potential impact of targeting APC. SUMMARY: We conclude that the signaling and anticoagulant functions of APC are in spatially and kinetically distinct compartments, and that it is possible to specifically inhibit the anticoagulant activity of APC. Targeting APC with a serpin is remarkably effective and may be safe for long-term prophylactic use in the treatment of hemophilia.


Subject(s)
Drug Delivery Systems/methods , Hemophilia A/drug therapy , Protein C/antagonists & inhibitors , Serpins/therapeutic use , Animals , Disease Models, Animal , Hemophilia A/blood , Humans , Mice , Protein C/metabolism
7.
Blood ; 129(1): 105-113, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27789479

ABSTRACT

Hemophilia is a bleeding disorder caused by deficiency in factors VIII or IX, the two components of the intrinsic Xase complex. Treatment with replacement factor can lead to the development of inhibitory antibodies, requiring the use of bypassing agents such as factor VIIa and factor concentrates. An alternative approach to bypass the Xase complex is to inhibit endogenous anticoagulant activities. Activated protein C (APC) breaks down the complex that produces thrombin by proteolytically inactivating factor Va. Defects in this mechanism (eg, factor V Leiden) are associated with thrombosis but result in less severe bleeding when co-inherited with hemophilia. Selective inhibition of APC might therefore be effective for the treatment of hemophilia. The endogenous inhibitors of APC are members of the serpin family: protein C inhibitor (PCI) and α1-antitrypsin (α1AT); however, both exhibit poor reactivity and selectivity for APC. We mutated residues in and around the scissile P1-P1' bond in PCI and α1AT, resulting in serpins with the desired specificity profile. The lead candidate was shown to promote thrombin generation in vitro and to restore fibrin and platelet deposition in an intravital laser injury model in hemophilia B mice. The power of targeting APC was further demonstrated by the complete normalization of bleeding after a severe tail clip injury in these mice. These results demonstrate that the protein C anticoagulant system can be successfully targeted by engineered serpins and that administration of such agents is effective at restoring hemostasis in vivo.


Subject(s)
Hemophilia B/drug therapy , Protein C Inhibitor/pharmacology , Protein C/antagonists & inhibitors , Serpins/pharmacology , Animals , Disease Models, Animal , Drug Design , Electrophoresis, Polyacrylamide Gel , Humans , Mice
8.
Nucleic Acid Ther ; 26(6): 355-362, 2016 12.
Article in English | MEDLINE | ID: mdl-27736370

ABSTRACT

Activated protein C (APC) is a critical regulator of thrombin formation and thereby protects against thrombosis. On the other hand, overwhelming formation of APC increases the risk of bleeding such as in trauma-induced coagulopathy. Thus, pharmacological inhibition of APC activity may improve blood clottability in certain clinical situations. In this study, we demonstrate that the DNA aptamer HS02-52G binds with fast onset (1.118 ± 0.013 × 105 M-1 s-1) to APC and possesses a long residence time of 13.5 min within the aptamer-APC complex. Functional analysis revealed HS02-52G as a highly potent and specific inhibitor of APC in plasma and whole blood with IC50 values ≤30 nM, whose activity can be readily neutralized by the short complementary DNA molecule AD22. These features qualify the novel aptamer-antidote pair as a candidate treatment option for acute APC-related bleedings.


Subject(s)
Anticoagulants/chemistry , Aptamers, Nucleotide/chemistry , Oligonucleotides, Antisense/chemistry , Protein C/antagonists & inhibitors , Thrombin/chemistry , Anticoagulants/chemical synthesis , Aptamers, Nucleotide/chemical synthesis , Base Pairing , Humans , Kinetics , Nucleic Acid Conformation , Oligonucleotides, Antisense/chemical synthesis , Partial Thromboplastin Time , Protein Binding , Protein C/chemistry , Recombinant Proteins/chemistry , Thermodynamics , Thrombin/agonists , Thrombin/antagonists & inhibitors , Whole Blood Coagulation Time
9.
Thromb Res ; 141: 77-83, 2016 May.
Article in English | MEDLINE | ID: mdl-26974491

ABSTRACT

We have demonstrated that antithrombin (AT)-independent thrombin inhibitors paradoxically increase thrombin generation (TG) in human plasma in a thrombomodulin (TM)- and protein C (PC)-dependent manner. We determined the effects of AT-independent thrombin inhibitors on the negative-feedback system, activation of PC and production and degradation of factor Va (FVa), as possible mechanisms underlying the paradoxical enhancement of TG. TG in human plasma containing 10nM TM was assayed by means of the calibrated automated thrombography. As an index of PC activation, plasma concentration of activated PC-PC inhibitor complex (aPC-PCI) was measured. The amounts of FVa heavy chain and its degradation product (FVa(307-506)) were examined by western blotting. AT-independent thrombin inhibitors, melagatran and dabigatran (both at 25-600nM) and 3-30µg/ml active site-blocked thrombin (IIai), increased peak levels of TG. Melagatran, dabigatran and IIai significantly decreased plasma concentration of aPC-PCI complex at 25nM or more, 75nM or more, and 10 and 30µg/ml, respectively. Melagatran (300nM) significantly increased FVa and decreased FVa(307-506). In contrast, a direct factor Xa inhibitor edoxaban preferentially inhibited thrombin generation (≥25nM), and higher concentrations were required to inhibit PC activation (≥150nM) and FVa degradation (300nM). The present study suggests that the inhibitions of protein C activation and subsequent degradation of FVa and increase in FVa by antithrombin-independent thrombin inhibitors may contribute to the paradoxical TG enhancement, and edoxaban may inhibit PC activation and FVa degradation as a result of TG suppression.


Subject(s)
Antithrombins/pharmacology , Azetidines/pharmacology , Benzylamines/pharmacology , Enzyme Activation/drug effects , Factor Va/metabolism , Protein C/antagonists & inhibitors , Proteolysis/drug effects , Thrombin/metabolism , Blood Coagulation/drug effects , Blood Coagulation Tests , Humans , Protein C/metabolism
10.
PLoS One ; 10(11): e0143137, 2015.
Article in English | MEDLINE | ID: mdl-26580551

ABSTRACT

Protein C inhibitor is a secreted, non-specific serine protease inhibitor with broad protease reactivity. It binds glycosaminoglycans and anionic phospholipids, which can modulate its activity. Anionic phospholipids, such as phosphatidylserine are normally localized to the inner leaflet of the plasma membrane, but are exposed on activated and apoptotic cells and on plasma membrane-derived microparticles. In this report we show by flow cytometry that microparticles derived from cultured cells and activated platelets incorporated protein C inhibitor during membrane blebbing. Moreover, protein C inhibitor is present in/on microparticles circulating in normal human plasma as judged from Western blots, ELISAs, flow cytometry, and mass spectrometry. These plasma microparticles are mainly derived from megakaryocytes. They seem to be saturated with protein C inhibitor, since they do not bind added fluorescence-labeled protein C inhibitor. Heparin partially removed microparticle-bound protein C inhibitor, supporting our assumption that protein C inhibitor is bound via phospholipids. To assess the biological role of microparticle-bound protein C inhibitor we performed protease inhibition assays and co-precipitated putative binding partners on microparticles with anti-protein C inhibitor IgG. As judged from amidolytic assays microparticle-bound protein C inhibitor did not inhibit activated protein C or thrombin, nor did microparticles modulate the activity of exogenous protein C inhibitor. Among the proteins co-precipitating with protein C inhibitor, complement factors, especially complement factor 3, were most striking. Taken together, our data do not support a major role of microparticle-associated protein C inhibitor in coagulation, but rather suggest an interaction with proteins of the complement system present on these phospholipid vesicles.


Subject(s)
Blood Platelets/chemistry , Cell Membrane/chemistry , Cell-Derived Microparticles/chemistry , Megakaryocytes/chemistry , Protein C Inhibitor/chemistry , Protein C/antagonists & inhibitors , Adult , Blood Platelets/cytology , Cell Membrane/metabolism , Cell-Derived Microparticles/metabolism , Female , Heparin/chemistry , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Jurkat Cells , Male , Megakaryocytes/cytology , Middle Aged , Phospholipids/chemistry , Phospholipids/metabolism , Platelet Factor 3/chemistry , Platelet Factor 3/metabolism , Protein Binding , Protein C/metabolism , Protein C Inhibitor/metabolism , Thrombin/chemistry , Thrombin/metabolism
11.
Thromb Res ; 135(6): 1203-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25887633

ABSTRACT

INTRODUCTION: Protein C inhibitor (PCI), a member of the serine protease inhibitor family, is expressed in various human tissues, including liver and kidneys. In the plasma, PCI physiologically inhibits an anticoagulant serine protease, activated protein C (APC). PCI expressed by cancer cells suppresses tumor invasion by inhibiting urokinase-type plasminogen activator, and inhibits tumor growth and metastasis, which are independent of its protease-inhibitory activity. In the present study, we clarified the effects of host PCI on growth and metastasis of B16 melanoma (B16) cells by comparing between wild-type mice and mice transgenic for human PCI gene (hPCI-TG), which have a tissue distribution of PCI similar to that observed in humans. MATERIALS AND METHODS: Growth of intracutaneously-injected B16 cells was evaluated by measuring the tumor volume, and metastatic behavior of intravenously-injected B16 cells by counting the number of metastatic lung nodules. RESULTS: Growth of intracutaneously injected B16 cells was significantly faster in wild-type mice than in hPCI-TG mice; however, hPCI-TG mice developed more metastatic nodules of B16 cells in the lungs. Immunohistochemical analysis using anti-mouse fibrinogen antibody revealed more fibrin deposition in the lung in hPCI-TG mice than in wild-type mice. Furthermore, the more invasive behavior observed in hPCI-TG mice was reduced by rabbit anti-human PCI IgG, APC, or soluble TM administration for 3 consecutive days including the day that B16 cells were injected. CONCLUSIONS: Our results suggest that like PCI expressed in tumor cells, host PCI also inhibits tumor growth, but host PCI promotes tumor metastasis via its procoagulant properties.


Subject(s)
Coagulants/chemistry , Lung Neoplasms/drug therapy , Protein C Inhibitor/blood , Protein C/antagonists & inhibitors , Thrombophilia/blood , Animals , Female , Humans , Immunoenzyme Techniques , Immunohistochemistry , Male , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Transplantation , Protein C Inhibitor/chemistry , RNA/analysis , Thrombomodulin/chemistry , Time Factors
12.
Thromb Res ; 133(6): 1105-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24513148

ABSTRACT

INTRODUCTION: Activated protein C (APC) is the central enzyme of the anticoagulant protein C pathway. Low concentrations of APC circulate in plasma and are believed to contribute to the maintenance of a normal haemostatic balance. MATERIALS AND METHODS: We have used a structure-based virtual screening approach to discover small drug-like molecules that inhibit the interaction between APC and its substrate FVa through inhibition of a predominant APC exosite, known to be involved in FVa substrate binding. We have combined in silico selection with functional screening and direct binding analysis to identify novel molecules and to ascertain and characterize the inhibition of the interaction between APC and FVa. RESULTS: We have identified a number of novel molecules that bind to APC and protein C with Kd values in the range of 10(-3)- 10(-5)M. Inhibition by these molecules is incomplete, which most likely reflects the extended surface that is involved in the interaction between APC and its substrates. Direct binding of hit molecules to variant APC molecules that were mutated in the targeted binding site revealed that several of the molecules presented a 100-500 fold lower affinity for the variant molecule, suggesting that these molecules indeed bind the exosite of APC. CONCLUSIONS: The protein-protein interaction inhibitors discovered here, could function as starting molecules for further development of small molecules with anti-APC properties. Such molecules may be of clinical interest, in particular in individuals where thrombin formation is compromised and the haemostatic balance is tipped towards bleeding tendencies, such as in haemophilia A.


Subject(s)
Protein C Inhibitor/pharmacology , Protein C/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Binding Sites , Drug Design , Drug Evaluation, Preclinical/methods , Humans , Models, Molecular , Protein C/chemistry , Protein C/metabolism , Protein C Inhibitor/chemistry , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Surface Plasmon Resonance
13.
Thromb Haemost ; 111(1): 41-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24136361

ABSTRACT

Protein C inhibitor (PCI, SerpinA5) is a heparin-binding serpin which can penetrate through cellular membranes. Selected negatively charged phospholipids like unsaturated phosphatidylserine and oxidised phosphatidylethanolamine bind to PCI and stimulate its inhibitory activity towards different proteases. The interaction of phospholipids with PCI might also alter the lipid distribution pattern of blood cells and influence the remodelling of cellular membranes. Here we showed that PCI is an additional binding partner of phosphatidic acid (PA), cardiolipin (CL), and phosphoinositides (PIPs). Protein lipid overlay assays exhibited a unique binding pattern of PCI towards different lipid species. In addition PA, CL, and unsaturated, monophosphorylated PIPs stimulated the inhibitory property of PCI towards activated protein C in a heparin like manner. As shown for kallistatin (SerpinA4) and vaspin (SerpinA12), the incubation of cells with PCI led to the activation of protein kinase B (AKT), which could be achieved through direct interaction of PCI with PIPs. This model is supported by the fact that PCI stimulated the PIP-dependent 5-phosphatase SHIP2 in vitro, which would result in AKT activation. Hence the interaction of PCI with different lipids might not only stimulate the inhibition of potential target protease by PCI, but could also alter intracellular lipid signalling.


Subject(s)
Lipids/chemistry , Phosphoric Monoester Hydrolases/chemistry , Protein C Inhibitor/chemistry , Protein C/antagonists & inhibitors , Blood Coagulation , Cardiolipins/chemistry , Cell Line, Tumor , Cell Membrane/metabolism , Diglycerides/chemistry , Dose-Response Relationship, Drug , Fatty Acids/chemistry , HEK293 Cells , Heparin/chemistry , Humans , Phosphatidic Acids/chemistry , Phosphatidylethanolamines/chemistry , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphatidylserines/chemistry , Phospholipids/chemistry , Protein Binding , Recombinant Proteins/chemistry , Serpins/chemistry , Signal Transduction
14.
BMC Biochem ; 14: 31, 2013 Nov 11.
Article in English | MEDLINE | ID: mdl-24215622

ABSTRACT

BACKGROUND: Alpha-1 proteinase inhibitor (API) is a plasma serpin superfamily member that inhibits neutrophil elastase; variant API M358R inhibits thrombin and activated protein C (APC). Fusing residues 1-75 of another serpin, heparin cofactor II (HCII), to API M358R (in HAPI M358R) was previously shown to accelerate thrombin inhibition over API M358R by conferring thrombin exosite 1 binding properties. We hypothesized that replacing HCII 1-75 region with the 13 C-terminal residues (triskaidecapeptide) of hirudin variant 3 (HV354-66) would further enhance the inhibitory potency of API M358R fusion proteins. We therefore expressed HV3API M358R (HV354-66 fused to API M358R) and HV3API RCL5 (HV354-66 fused to API F352A/L353V/E354V/A355I/I356A/I460L/M358R) API M358R) as N-terminally hexahistidine-tagged polypeptides in E. coli. RESULTS: HV3API M358R inhibited thrombin 3.3-fold more rapidly than API M358R; for HV3API RCL5 the rate enhancement was 1.9-fold versus API RCL5; neither protein inhibited thrombin as rapidly as HAPI M358R. While the thrombin/Activated Protein C rate constant ratio was 77-fold higher for HV3API RCL5 than for HV3API M358R, most of the increased specificity derived from the API F352A/L353V/E354V/A355I/I356A/I460L API RCL 5 mutations, since API RCL5 remained 3-fold more specific than HV3API RCL5. An HV3 54-66 peptide doubled the Thrombin Clotting Time (TCT) and halved the binding of thrombin to immobilized HCII 1-75 at lower concentrations than free HCII 1-75. HV3API RCL5 bound active site-inhibited FPR-chloromethyl ketone-thrombin more effectively than HAPI RCL5. Transferring the position of the fused HV3 triskaidecapeptide to the C-terminus of API M358R decreased the rate of thrombin inhibition relative to that mediated by HV3API M358R by 11-to 14-fold. CONCLUSIONS: Fusing the C-terminal triskaidecapeptide of HV3 to API M358R-containing serpins significantly increased their effectiveness as thrombin inhibitors, but the enhancement was less than that seen in HCII 1-75-API M358R fusion proteins. HCII 1-75 was a superior fusion partner, in spite of the greater affinity of the HV3 triskaidecapeptide, manifested both in isolated and API-fused form, for thrombin exosite 1. Our results suggest that HCII 1-75 binds thrombin exosite 1 and orients the attached serpin scaffold for more efficient interaction with the active site of thrombin than the HV3 triskaidecapeptide.


Subject(s)
Hirudins/metabolism , Serpins/metabolism , Thrombin/metabolism , alpha 1-Antitrypsin/metabolism , Amino Acid Sequence , Catalytic Domain , Hirudins/chemistry , Hirudins/genetics , Histidine/genetics , Histidine/metabolism , Humans , Kinetics , Mutation , Oligopeptides/genetics , Oligopeptides/metabolism , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Binding , Protein C/antagonists & inhibitors , Protein C/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Thrombin/antagonists & inhibitors , alpha 1-Antitrypsin/genetics
15.
J Food Sci ; 78(10): H1621-H1628, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24024564

ABSTRACT

This study investigated the combined effects of trans fat diet (TFD) and doxorubicin upon cardiac oxidative, inflammatory, and coagulatory stress. TFD increased trans fatty acid deposit in heart (P < 0.05), and decreased protein C and antithrombin-III activities in circulation (P < 0.05). TFD plus doxorubicin treatment elevated activities of plasminogen activator inhibitor-1, lactate dehydrogenase, and creatine phosphokinase (P < 0.05). This combination also raised xanthine oxidase activity, and enhanced cardiac levels of reactive oxygen species, interleukin (IL)-6, IL-10, tumor necrosis factor-alpha, and monocyte chemoattractant protein-1 than TFD or doxorubicin treatment alone (P < 0.05). TFD alone increased cardiac nuclear factor kappa B (NF-κB) activity (P < 0.05), but failed to affect expression of NF-κB and mitogen-activated protein kinase (MAPK) (P > 0.05). Doxorubicin treatment alone augmented cardiac activity, mRNA expression, and protein production of NF-κB and MAPK (P < 0.05). TFD plus doxorubicin treatment further upregulated cardiac expression of NF-κB p65, p-p38, and p-ERK1/2 (P < 0.05). These findings suggest that TFD exacerbates doxorubicin-induced cardiotoxicity.


Subject(s)
Cardiotoxins/toxicity , Dietary Fats/adverse effects , Doxorubicin/toxicity , Trans Fatty Acids/adverse effects , Animals , Antithrombin III/antagonists & inhibitors , Antithrombin III/metabolism , C-Reactive Protein/metabolism , Chemokine CCL2/metabolism , Creatine Kinase/blood , Fibrinogen/metabolism , Heart Diseases/chemically induced , Heart Diseases/pathology , Interleukin-10/metabolism , Interleukin-6/metabolism , L-Lactate Dehydrogenase/blood , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Plasminogen Activator Inhibitor 1/agonists , Plasminogen Activator Inhibitor 1/blood , Protein C/antagonists & inhibitors , Protein C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/blood , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Triglycerides/blood , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
16.
J Immunol ; 191(7): 3764-77, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23997223

ABSTRACT

Activated protein C (PC) is an anticoagulant involved in the interactions between the coagulation and immune systems. Activated PC has broad anti-inflammatory effects that are mediated through its ability to modulate leukocyte function and confer vascular barrier protection. We investigated the influence of activated PC on the pathogenesis of experimental autoimmune encephalomyelitis (EAE), the animal model for multiple sclerosis. We modulated activated PC levels in the circulation during EAE induction through systemic administration of a mAb against PC/activated PC (anti-PC). We initially hypothesized that inhibition of activated PC may result in a heightened inflammatory environment, leading to increased EAE pathogenesis. Contrary to this hypothesis, mice treated with anti-PC Ab (anti-PC mice) exhibited attenuated EAE. Interestingly, despite reduced disease severity and minimal pathogenic conditions in the CNS, anti-PC mice exhibited considerable leukocyte infiltration in the brain, comparable to control mice with severe EAE. Furthermore, CD4(+) T cells were diminished in the periphery of anti-PC mice, whereas various CD11b(+) populations were increased, notably the myeloid-derived suppressor cells (MDSCs), a CD11b(+) subset characterized as potent T cell suppressors. MDSCs from anti-PC mice exhibited increased expression of T cell suppressive factors and effectively inhibited T cell proliferation. Overall, our findings show that activated PC inhibition affected EAE pathogenesis at multiple fronts, specifically increasing vascular barrier permeability, as evidenced by considerable leukocyte infiltration in the brain. Additionally, inhibition of activated PC modulated the functional responses of CD11b(+) cells, leading to the expansion and increased activation of MDSCs, which are suppressive to the CD4(+) T cells required for EAE progression, thereby resulting in attenuated EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Protein C/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Arginase/metabolism , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Immunophenotyping , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Nitric Oxide Synthase Type II/metabolism , Protein C/antagonists & inhibitors , Protein C/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
17.
Blood Cells Mol Dis ; 50(4): 252-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23385154

ABSTRACT

Multiple protective effects of pharmacological activated protein C (APC) are reported in several organ pathologies. To help evaluate the endogenous murine PC system, we characterized a rat monoclonal anti-mouse PC antibody, SPC-54, which inhibited the amidolytic and anticoagulant activities of murine APC by>95%. SPC-54 blocked active site titration of purified APC using the active site titrant, biotinylated FPR-chloromethylketone, showing that SPC-54 blocks access to APC's active site to inhibit all enzymatic activity. A single injection of SPC-54 (10mg/kg) neutralized circulating PC in mice for at least 7days, and immunoblotting and immuno-precipitation with protein G-agarose confirmed that SPC-54 in vivo was bound to PC in plasma. Pre-infusion of SPC-54 in tissue factor-induced murine acute thromboembolism experiments caused a major decrease in mean survival time compared to controls (7min vs. 42.5min, P=0.0016). SPC-54 decreased lung perfusion in this model by 54% when monitored by vascular perfusion methodologies using infrared fluorescence of Evans blue dye. In LD50 endotoxemia murine models, SPC-54 infused at 7hr after endotoxin administration increased mortality from 42% to 100% (P<0.001). In summary, monoclonal antibody SPC-54 ablates in vitro and in vivo APC protective functions and enzymatic activity. The ability of SPC-54 to block the endogenous PC/APC system provides a powerful tool to understand better the role of the endogenous PC system in murine injury models and in cell bioassays and also to neutralize the enzymatic activities of murine APC in any assay system.


Subject(s)
Antibodies, Monoclonal/pharmacology , Protein C/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Neutralizing/administration & dosage , Antibodies, Neutralizing/adverse effects , Antibodies, Neutralizing/pharmacology , Disease Models, Animal , Endotoxemia/etiology , Endotoxemia/mortality , Enzyme Activation/drug effects , Mice , Protein Binding , Protein C/immunology , Protein C/metabolism , Rats , Thrombosis/etiology , Thrombosis/mortality
18.
J Thromb Haemost ; 11(2): 282-92, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23216621

ABSTRACT

BACKGROUND: Activated protein C (APC) exerts anticoagulant effects via inactivation of factors Va and VIIIa and cytoprotective effects via protease activated receptor (PAR)1. Inhibition of endogenous APC in endotoxemia and sepsis results in exacerbation of coagulation and inflammation, with consequent enhanced lethality. OBJECTIVES: We here sought to dissect the distinct roles of the anticoagulant and cytoprotective functions of endogenous APC in severe Gram-negative pneumonia-derived sepsis (melioidosis). METHODS: We infected wild-type (WT) mice with Burkholderia pseudomallei, a common sepsis pathogen in southeast Asia, and treated them with antibodies inhibiting both the anticoagulant and cytoprotective functions of APC (MPC1609) or the anticoagulant functions of APC (MAPC1591) only. Additionally, we administered SEW2871 (stimulating the S1P1-pathway downstream from PAR1) to control and MPC1609-treated mice. RESULTS: MPC1609, but not MAPC1591, significantly worsened survival, increased coagulation activation, facilitated bacterial growth and dissemination and enhanced the inflammatory response. The effects of MPC1609 could not be reversed by SEW2871, suggesting that S1P1 does not play a major role in this model. CONCLUSIONS: These results suggest that the mere inhibition of the anticoagulant function of APC does not interfere with its protective role during Gram-negative pneumosepsis, suggesting a more prominent role for cytoprotective effects of APC .


Subject(s)
Blood Coagulation , Burkholderia pseudomallei/pathogenicity , Lung/metabolism , Melioidosis/prevention & control , Protein C/metabolism , Sepsis/prevention & control , Animals , Antibodies, Monoclonal/administration & dosage , Bacterial Load , Blood Coagulation/drug effects , Burkholderia pseudomallei/growth & development , Cytokines/blood , Cytoprotection , Disease Models, Animal , Female , Inflammation/blood , Inflammation/immunology , Inflammation/microbiology , Inflammation Mediators/blood , Liver/microbiology , Lung/immunology , Lung/microbiology , Lysophospholipids/metabolism , Melioidosis/blood , Melioidosis/immunology , Melioidosis/microbiology , Mice , Mice, Inbred C57BL , Oxadiazoles/administration & dosage , Protein C/antagonists & inhibitors , Protein C/immunology , Receptor, PAR-1/metabolism , Sepsis/blood , Sepsis/immunology , Sepsis/microbiology , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Thiophenes/administration & dosage , Time Factors
20.
Nat Med ; 18(7): 1123-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22729286

ABSTRACT

Tissue damage induced by ionizing radiation in the hematopoietic and gastrointestinal systems is the major cause of lethality in radiological emergency scenarios and underlies some deleterious side effects in patients undergoing radiation therapy. The identification of target-specific interventions that confer radiomitigating activity is an unmet challenge. Here we identify the thrombomodulin (Thbd)-activated protein C (aPC) pathway as a new mechanism for the mitigation of total body irradiation (TBI)-induced mortality. Although the effects of the endogenous Thbd-aPC pathway were largely confined to the local microenvironment of Thbd-expressing cells, systemic administration of soluble Thbd or aPC could reproduce and augment the radioprotective effect of the endogenous Thbd-aPC pathway. Therapeutic administration of recombinant, soluble Thbd or aPC to lethally irradiated wild-type mice resulted in an accelerated recovery of hematopoietic progenitor activity in bone marrow and a mitigation of lethal TBI. Starting infusion of aPC as late as 24 h after exposure to radiation was sufficient to mitigate radiation-induced mortality in these mice. These findings suggest that pharmacologic augmentation of the activity of the Thbd-aPC pathway by recombinant Thbd or aPC might offer a rational approach to the mitigation of tissue injury and lethality caused by ionizing radiation.


Subject(s)
Protein C/antagonists & inhibitors , Radiation Injuries/prevention & control , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Thrombomodulin/antagonists & inhibitors , Animals , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/metabolism , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Inbred C57BL , Protein C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radiation Injuries/genetics , Radiation Injuries/pathology , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Receptors, Thrombin , Survival Analysis , Thrombomodulin/genetics , Thrombomodulin/metabolism , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL
...