Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Blood ; 130(24): 2678-2681, 2017 12 14.
Article in English | MEDLINE | ID: mdl-29021229

ABSTRACT

Sepsis concurrently activates both coagulation and complement systems. Although complement activation by bacteria is well documented, work in mice and in vitro suggests that coagulation proteases can directly cleave complement proteins. We aimed to determine whether generation of coagulation proteases in vivo can activate the complement cascade in 2 highly coagulopathic models. We compared temporal changes in activation biomarkers of coagulation (thrombin-antithrombin [TAT]), fibrinolysis (plasmin-antiplasmin [PAP]), and complement (C3b, C5a, C5b-9) in baboons infused with factor Xa (FXa) and phospholipids (FXa/phosphatidylcholine-phosphatidylserine [PCPS]) vs LD100 Escherichia coli We found that, albeit with different timing, both FXa/PCPS and E coli infusion led to robust thrombin and plasmin generation. Conversely, only E coli challenge activated the complement system, reaching a maximum at 2 hours postchallenge during the peaks of lipopolysaccharide and bacteremia but not of TAT and PAP. Despite inducing a strong burst of thrombin and plasmin, FXa/PCPS infusion did not produce measurable levels of complement activation in vivo. Similarly, ex vivo incubation of baboon serum with thrombin, plasmin, or FXa did not show noticeable complement cleavage unless supraphysiologic amounts of enzymes were used. Our results suggest that in vivo-generated thrombin and plasmin do not directly activate the complement in nonhuman primates.


Subject(s)
Complement Activation/immunology , Complement System Proteins/immunology , Fibrinolysin/immunology , Thrombin/immunology , Animals , Complement Activation/drug effects , Complement System Proteins/metabolism , Escherichia coli/immunology , Escherichia coli/metabolism , Factor Xa/immunology , Factor Xa/pharmacology , Fibrinolysin/metabolism , Humans , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Papio , Phosphatidylcholines/immunology , Phosphatidylcholines/pharmacology , Phosphatidylserines/immunology , Phosphatidylserines/pharmacology , Thrombin/metabolism
2.
Biochim Biophys Acta ; 1862(7): 1354-66, 2016 07.
Article in English | MEDLINE | ID: mdl-27094771

ABSTRACT

Ischaemic stroke, accompanied by neuroinflammation, impairs blood-brain barrier integrity through a complex mechanism involving both protein kinase C (PKC) and urokinase. Using an in vitro model of human blood-brain barrier (BBB) composed of brain microvascular endothelial cells (HBMEC) and astrocytes, this study assessed the putative roles of these elements in BBB damage evoked by enhanced availability of pro-inflammatory cytokine, TNF-α. Treatment of HBMEC with TNF-α significantly increased the mRNA and protein expressions of all plasminogen-plasmin system (PPS) components, namely tissue plasminogen activator, urokinase, urokinase plasminogen activator receptor and plasminogen activator inhibitor-1 and also the activities of urokinase, total PKC and extracellular MMP-2. Inhibition of urokinase by amiloride abated the effects of TNF-α on BBB integrity and MMP-2 activity without affecting that of total PKC. Conversely, pharmacological inhibition of conventional PKC isoforms dramatically suppressed TNF-α-induced overactivation of urokinase. Knockdown of PKC-α gene via specific siRNA in HBMEC suppressed the stimulatory effects of TNF-α on protein expression of all PPS components, MMP-2 activity, DNA fragmentation rates and pro-apoptotic caspase-3/7 activities. Establishment of co-cultures with BMEC transfected with PKC-α siRNA attenuated the disruptive effects of TNF-α on BBB integrity and function. This was partly due to elevations observed in expression of a tight junction protein, claudin-5 and partly to prevention of stress fibre formation. In conclusion, specific inhibition of PKC-α in cerebral conditions associated with exaggerated release of pro-inflammatory cytokines, notably TNF-α may be of considerable therapeutic value and help maintain endothelial cell viability, appropriate cytoskeletal structure and basement membrane.


Subject(s)
Blood-Brain Barrier/pathology , Fibrinolysin/immunology , Inflammation/pathology , Matrix Metalloproteinase 2/immunology , Plasminogen/immunology , Protein Kinase C-alpha/immunology , Tumor Necrosis Factor-alpha/immunology , Apoptosis , Blood-Brain Barrier/immunology , Cell Line , Gene Silencing , Humans , Inflammation/immunology , Protein Kinase C-alpha/genetics , Urokinase-Type Plasminogen Activator/immunology
3.
Biochim Biophys Acta ; 1862(3): 395-402, 2016 03.
Article in English | MEDLINE | ID: mdl-26493446

ABSTRACT

The plasminogen activation (PA) system consists in a group of proteases and protease inhibitors regulating the activation of the zymogen plasminogen into its proteolytically active form, plasmin. Here, we give an update of the current knowledge about the role of the PA system on different aspects of neuroinflammation. These include modification in blood-brain barrier integrity, leukocyte diapedesis, removal of fibrin deposits in nervous tissues, microglial activation and neutrophil functions. Furthermore, we focus on the molecular mechanisms (some of them independent of plasmin generation and even of proteolysis) and target receptors responsible for these effects. The description of these mechanisms of action may help designing new therapeutic strategies targeting the expression, activity and molecular mediators of the PA system in neurological disorders involving neuroinflammatory processes. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.


Subject(s)
Blood-Brain Barrier/immunology , Central Nervous System Diseases/immunology , Inflammation/immunology , Microglia/immunology , Plasminogen/immunology , Animals , Blood-Brain Barrier/pathology , Central Nervous System Diseases/pathology , Fibrin/immunology , Fibrinolysin/immunology , Humans , Inflammation/pathology , Leukocytes/immunology , Leukocytes/pathology , Microglia/pathology , Tissue Plasminogen Activator/immunology
4.
Gastroenterology ; 148(3): 565-578.e4, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25490065

ABSTRACT

BACKGROUND & AIMS: Activated proteases such as plasmin and matrix metalloproteinases (MMPs) are activated in intestinal tissues of patients with active inflammatory bowel diseases. We investigated the effect of plasmin on the progression of acute colitis. METHODS: Colitis was induced in Mmp9(-/-), Plg(-/-), and C57BL/6 (control) mice by the administration of dextran sulfate sodium, trinitrobenzene sulfonic acid, or CD40 antibody. Plasmin was inhibited in control mice by intraperitoneal injection of YO-2, which blocks its active site. Mucosal and blood samples were collected and analyzed by reverse-transcription polymerase chain reaction and immunohistochemical analyses, as well as for mucosal inflammation and levels of cytokines and chemokines. RESULTS: Circulating levels of plasmin were increased in mice with colitis, compared with controls. Colitis did not develop in control mice injected with YO-2 or in Plg(-/-) mice. Colons from these mice had reduced infiltration of Gr1+ neutrophils and F4/80+ macrophages, and reduced levels of inflammatory cytokines and chemokines. Colonic inflammation and colitis induction required activation of endogenous MMP9. After colitis induction, mice given YO-2, Plg(-/-) mice, and Mmp9(-/-) mice had reduced serum levels of tumor necrosis factor and C-X-C motifĀ chemokine ligand 5, compared with control mice. CONCLUSIONS: In mice, plasmin induces a feedback mechanism in which activation of the fibrinolytic system promotes the development of colitis via activation of MMP9 or proteolytic enzymes. The proteolytic environment stimulates the influx of myeloid cells into the colonic epithelium and the production of tumor necrosis factor and C-X-C motif chemokine ligand 5. In turn, myeloid CD11b+ cells release the urokinase plasminogen activator, which accelerates plasmin production. Disruption of the plasmin-induced chronic inflammatory circuit therefore might be a strategy for colitis treatment.


Subject(s)
Colitis/metabolism , Fibrinolysin/antagonists & inhibitors , Matrix Metalloproteinase 9/metabolism , Myeloid Cells/metabolism , Animals , CD40 Antigens/antagonists & inhibitors , Chemokine CXCL5/immunology , Colitis/chemically induced , Colitis/immunology , Dextran Sulfate/toxicity , Dipeptides/pharmacology , Disease Models, Animal , Fibrinolysin/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Intestinal Mucosa/immunology , Macrophages/immunology , Matrix Metalloproteinase 9/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Neutrophils/immunology , Trinitrobenzenesulfonic Acid/toxicity , Tumor Necrosis Factor-alpha/immunology
5.
J Immunol ; 193(7): 3654-63, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25165151

ABSTRACT

The plasminogen (Plg)/plasmin (Pla) system is associated with a variety of biological activities beyond the classical dissolution of fibrin clots, including cell migration, tissue repair, and inflammation. Although the capacity of Plg/Pla to induce cell migration is well defined, the mechanism underlying this process in vivo is elusive. In this study, we show that Pla induces in vitro migration of murine fibroblasts and macrophages (RAW 264.7) dependent on the MEK/ERK pathway and by requiring its proteolytic activity and lysine binding sites. Plasmin injection into the pleural cavity of BALB/c mice induced a time-dependent influx of mononuclear cells that was associated with augmented ERK1/2 and IκB-α phosphorylation and increased levels of CCL2 and IL-6 in pleural exudates. The inhibition of protease activity by using a serine protease inhibitor leupeptin or two structurally different protease-activated receptor-1 antagonists (SCH79797 and RWJ56110) abolished Pla-induced mononuclear recruitment and ERK1/2 and IκB-α phosphorylation. Interestingly, inhibition of the MEK/ERK pathway abolished Pla-induced CCL2 upregulation and mononuclear cell influx. In agreement with a requirement for the CCL2/CCR2 axis to Pla-induced cell migration, the use of a CCR2 antagonist (RS504393) prevented the Plg/Pla-induced recruitment of mononuclear cells to the pleural cavity and migration of macrophages at transwell plates. Therefore, Pla-induced mononuclear cell recruitment in vivo was dependent on protease-activated receptor-1 activation of the MEK/ERK/NF-κB pathway, which led to the release of CCL2 and activation of CCR2.


Subject(s)
Cell Movement/immunology , Extracellular Signal-Regulated MAP Kinases/immunology , Fibrinolysin/immunology , MAP Kinase Kinase Kinases/immunology , MAP Kinase Signaling System/immunology , Monocytes/immunology , Receptor, PAR-1/immunology , Receptors, CCR2/immunology , Animals , Benzoxazines/pharmacology , Cell Movement/drug effects , Chemokine CCL2/immunology , MAP Kinase Signaling System/drug effects , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred BALB C , NF-kappa B/immunology , Pleural Cavity/immunology , Receptors, CCR2/antagonists & inhibitors , Spiro Compounds/pharmacology , Up-Regulation/drug effects , Up-Regulation/immunology
6.
J Biol Chem ; 288(35): 25229-25243, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23861404

ABSTRACT

The Lyme disease spirochete Borrelia burgdorferi lacks endogenous, surface-exposed proteases. In order to efficiently disseminate throughout the host and penetrate tissue barriers, borreliae rely on recruitment of host proteases, such as plasmin(ogen). Here we report the identification of a novel plasminogen-binding protein, BBA70. Binding of plasminogen is dose-dependent and is affected by ionic strength. The BBA70-plasminogen interaction is mediated by lysine residues, primarily located in a putative C-terminal α-helix of BBA70. These lysine residues appear to interact with the lysine-binding sites in plasminogen kringle domain 4 because a deletion mutant of plasminogen lacking that domain was unable to bind to BBA70. Bound to BBA70, plasminogen activated by urokinase-type plasminogen activator was able to degrade both a synthetic chromogenic substrate and the natural substrate fibrinogen. Furthermore, BBA70-bound plasmin was able to degrade the central complement proteins C3b and C5 and inhibited the bacteriolytic effects of complement. Consistent with these functional activities, BBA70 is located on the borrelial outer surface. Additionally, serological evidence demonstrated that BBA70 is produced during mammalian infection. Taken together, recruitment and activation of plasminogen could play a beneficial role in dissemination of B. burgdorferi in the human host and may possibly aid the spirochete in escaping the defense mechanisms of innate immunity.


Subject(s)
Bacterial Proteins/metabolism , Borrelia burgdorferi/metabolism , Plasminogen/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Borrelia burgdorferi/chemistry , Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Complement C3b/chemistry , Complement C3b/genetics , Complement C3b/immunology , Complement C3b/metabolism , Complement C5/chemistry , Complement C5/genetics , Complement C5/immunology , Complement C5/metabolism , Fibrinolysin/chemistry , Fibrinolysin/genetics , Fibrinolysin/immunology , Fibrinolysin/metabolism , Humans , Immunity, Innate , Lyme Disease/genetics , Lyme Disease/immunology , Lyme Disease/metabolism , Plasminogen/chemistry , Plasminogen/genetics , Plasminogen/immunology , Protein Binding , Protein Structure, Tertiary , Urokinase-Type Plasminogen Activator/chemistry , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/immunology , Urokinase-Type Plasminogen Activator/metabolism
7.
J Biol Chem ; 288(10): 6849-63, 2013 Mar 08.
Article in English | MEDLINE | ID: mdl-23341464

ABSTRACT

Streptococcus pneumoniae infections remain a major cause of morbidity and mortality worldwide. Therefore a detailed understanding and characterization of the mechanism of host cell colonization and dissemination is critical to gain control over this versatile pathogen. Here we identified a novel 72-kDa pneumococcal protein endopeptidase O (PepO), as a plasminogen- and fibronectin-binding protein. Using a collection of clinical isolates, representing different serotypes, we found PepO to be ubiquitously present both at the gene and protein level. In addition, PepO protein was secreted in a growth phase-dependent manner to the culture supernatants of the pneumococcal isolates. Recombinant PepO bound human plasminogen and fibronectin in a dose-dependent manner and plasminogen did not compete with fibronectin for binding PepO. PepO bound plasminogen via lysine residues and the interaction was influenced by ionic strength. Moreover, upon activation of PepO-bound plasminogen by urokinase-type plasminogen activator, generated plasmin cleaved complement protein C3b thus assisting in complement control. Furthermore, direct binding assays demonstrated the interaction of PepO with epithelial and endothelial cells that in turn blocked pneumococcal adherence. Moreover, a pepO-mutant strain showed impaired adherence to and invasion of host cells compared with their isogenic wild-type strains. Taken together, the results demonstrated that PepO is a ubiquitously expressed plasminogen- and fibronectin-binding protein, which plays role in pneumococcal invasion of host cells and aids in immune evasion.


Subject(s)
Bacterial Proteins/immunology , Endopeptidases/immunology , Fibronectins/immunology , Immune Evasion/immunology , Immunity, Innate/immunology , Plasminogen/immunology , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Blotting, Western , Cell Line, Tumor , Cells, Cultured , Complement C3b/immunology , Complement C3b/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Fibrinolysin/immunology , Fibrinolysin/metabolism , Fibronectins/metabolism , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/microbiology , Humans , Microscopy, Confocal , Mutation , Plasminogen/metabolism , Protein Binding , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/immunology , Streptococcus pneumoniae/metabolism , Urokinase-Type Plasminogen Activator/immunology , Urokinase-Type Plasminogen Activator/metabolism
8.
Rheumatology (Oxford) ; 52(8): 1448-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23598443

ABSTRACT

OBJECTIVE: Cardiac neonatal lupus (cardiac-NL), initiated by surface binding of anti-Ro60 autoantibodies to apoptotic cardiocytes during development, activates the urokinase plasminogen activator/urokinase plasminogen activator receptor (uPA/uPAR) system. Subsequent accumulation of apoptotic cells and plasmin generation facilitates increased binding of anti-Ro60 by disrupting and cleaving circulating Ɵ2-glycoprotein I (Ɵ2GPI) thereby eliminating its protective effect. The association of soluble levels of components of the uPA/uPAR system with cardiac-NL was examined. METHODS: Levels of the uPA/uPAR system were assessed by ELISA in cord blood and immunohistological evaluation of autopsies. RESULTS: uPA, uPAR and plasminogen levels were each significantly higher in cord blood from cardiac-NL (n = 35) compared with non-cardiac-NL (n = 26) anti-Ro-exposed neonates: 3.3 Ā± 0.1 vs 1.9 Ā± 0.05 ng/ml (P < 0.0001), 6.6 Ā± 0.3 vs 2.1 Ā± 0.2 ng/ml (P < 0.0001) and 435 Ā± 34 vs 220 Ā± 19 ng/ml (P < 0.0001), respectively. In three twin pairs discordant for cardiac-NL, the twin with cardiac-NL had higher levels of uPA, uPAR and plasminogen than the unaffected twin (3.1 Ā± 0.1 vs 1.9 Ā± 0.05 ng/ml; P = 0.0086, 6.2 Ā± 1.4 vs 2.2 Ā± 0.7 ng/ml; P = 0.147 and 412 Ā± 61 vs 260 Ā± 27 ng/ml; P = 0.152, respectively). Immunohistological evaluation of three hearts from fetuses dying with cardiac-NL revealed macrophages and giant cells expressing uPA and plasminogen in the septal region. CONCLUSION: Increased soluble uPA, uPAR and plasminogen in cord blood and expression in affected tissue of fetuses with cardiac-NL supports the hypothesis that fetal cardiac injury is in part mediated by plasmin generation initiated by anti-Ro binding to the apoptotic cardiocyte.


Subject(s)
Fetal Blood/immunology , Fibrinolysin/immunology , Heart Diseases/immunology , Lupus Erythematosus, Systemic/congenital , Receptors, Urokinase Plasminogen Activator/immunology , Ribonucleoproteins/immunology , Antibodies, Antinuclear/immunology , Antibodies, Antinuclear/metabolism , Biomarkers , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Female , Fibrinolysin/metabolism , Heart Diseases/mortality , Heart Diseases/pathology , Humans , Immunohistochemistry , Infant, Newborn , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/mortality , Lupus Erythematosus, Systemic/pathology , Macrophages/immunology , Myocytes, Cardiac/immunology , Myocytes, Cardiac/metabolism , Pregnancy , Receptors, Urokinase Plasminogen Activator/blood , Reference Values , Ribonucleoproteins/metabolism , Survival Rate , Urokinase-Type Plasminogen Activator/blood , Urokinase-Type Plasminogen Activator/immunology
9.
J Biol Chem ; 286(26): 23559-69, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21566135

ABSTRACT

TSG-6 (TNF-α-stimulated gene/protein 6), a hyaluronan (HA)-binding protein, has been implicated in the negative regulation of inflammatory tissue destruction. However, little is known about the tissue/cell-specific expression of TSG-6 in inflammatory processes, due to the lack of appropriate reagents for the detection of this protein in vivo. Here, we report on the development of a highly sensitive detection system and its use in cartilage proteoglycan (aggrecan)-induced arthritis, an autoimmune murine model of rheumatoid arthritis. We found significant correlation between serum concentrations of TSG-6 and arthritis severity throughout the disease process, making TSG-6 a better biomarker of inflammation than any of the other arthritis-related cytokines measured in this study. TSG-6 was present in arthritic joint tissue extracts together with the heavy chains of inter-α-inhibitor (IαI). Whereas TSG-6 was broadly detectable in arthritic synovial tissue, the highest level of TSG-6 was co-localized with tryptases in the heparin-containing secretory granules of mast cells. In vitro, TSG-6 formed complexes with the tryptases murine mast cell protease-6 and -7 via either heparin or HA. In vivo TSG-6-tryptase association could also be detected in arthritic joint extracts by co-immunoprecipitation. TSG-6 has been reported to suppress inflammatory tissue destruction by enhancing the serine protease-inhibitory activity of IαI against plasmin. TSG-6 achieves this by transferring heavy chains from IαI to HA, thus liberating the active bikunin subunit of IαI. Because bikunin is also present in mast cell granules, we propose that TSG-6 can promote inhibition of tryptase activity via a mechanism similar to inhibition of plasmin.


Subject(s)
Arthritis/metabolism , Cell Adhesion Molecules/metabolism , Heparin/metabolism , Tryptases/metabolism , Alpha-Globulins/immunology , Alpha-Globulins/metabolism , Animals , Arthritis/immunology , Biomarkers/metabolism , CHO Cells , Cell Adhesion Molecules/immunology , Cricetinae , Cricetulus , Fibrinolysin/immunology , Fibrinolysin/metabolism , Heparin/immunology , Humans , Joints/immunology , Joints/metabolism , Mice , Tryptases/immunology
10.
Exp Mol Pathol ; 92(1): 175-84, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22044461

ABSTRACT

Activation of the fibrinolytic pathway has long been associated with human breast cancer. Plasmin is the major end product of the fibrinolytic pathway and is critical for normal physiological functions. The mechanism by which plasmin is generated in breast cancer is not yet fully described. We previously identified annexin II (ANX II), a fibrinolytic receptor, in human breast tumor tissue samples and observed a strong positive correlation with advanced stage cancer (Sharma et al., 2006a). We further demonstrated that tissue plasminogen activator (tPA) binds to ANX II in invasive breast cancer MDA-MB231cells, which leads to plasmin generation (Sharma et al., 2010). We hypothesize that ANX II-dependent plasmin generation in breast tumor is necessary to trigger the switch to neoangiogenesis, thereby stimulating a more aggressive cancer phenotype. Our immunohistochemical studies of human breast tumor tissues provide compelling evidence of a strong positive correlation between ANX II expression and neoangiogenesis, and suggest that ANX II is a potential target to slow or inhibit breast tumor growth by inhibiting neoangiogenesis. We now report that administration of anti-ANX II antibody potently inhibits the growth of human breast tumor in a xenograft model. Inhibition of tumor growth is at least partly due to attenuation of neoangiogenic activity within the tumor. In vitro studies demonstrate that anti-ANX II antibody inhibits angiogenesis on three dimensional matrigel cultures by eliciting endothelial cell (EC) death likely due to apoptosis. Taken together, these data suggest that selective disruption of the fibrinolytic activity of ANX II may provide a novel strategy for specific inhibition of neoangiogenesis in human breast cancer.


Subject(s)
Annexin A2/immunology , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Breast/blood supply , Neovascularization, Pathologic/prevention & control , Animals , Annexin A2/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Breast/metabolism , Breast/pathology , Breast Neoplasms/immunology , Disease Models, Animal , Female , Fibrinolysin/immunology , Fibrinolysin/metabolism , Humans , Immunohistochemistry , Mice , Mice, Nude , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Phenotype , Recombinant Proteins , Tissue Plasminogen Activator/genetics , Tissue Plasminogen Activator/metabolism , Transplantation, Heterologous
11.
Microb Pathog ; 51(5): 360-5, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21802507

ABSTRACT

Leptospirosis is a widespread re-emerging zoonosis of human and veterinary concern. It has been shown that virulent leptospires protect themselves against the host's innate immune system, a strategy that allows the bacteria to reach immunologically safe environments. Although extensive studies on host-pathogen interactions have been performed, little is known on how leptospires deal with host immune attack. In a previous work, we demonstrated the ability of leptospires to bind human plasminogen (PLG), that after treatment with activators, conferred plasmin (PLA) activity on the bacteria surface. In this study, we show that the PLA activity associated to the outer surface of Leptospira could interfere with the host immune attack by conferring some evasion advantage during infection. We demonstrate that PLA-coated leptospires interfere with complement C3b and IgG depositions on the bacterial surface, probably through the degradation of these components, thus diminishing opsonization process. Similar decrease on the deposition was observed when normal and immune sera from patients diagnosed with leptospirosis were employed as a source of IgG. We believe that decreasing opsonization by PLA generation might be an important aspect of the leptospiral immune escape strategy and survival. To our knowledge, this is the first proteolytic activity of plasmin associated-Leptospira related to anti-opsonic properties reported to date.


Subject(s)
Fibrinolysin/immunology , Immune Evasion , Leptospira interrogans/pathogenicity , Leptospirosis/enzymology , Leptospirosis/immunology , Fibrinolysin/metabolism , Host-Pathogen Interactions , Humans , Leptospira interrogans/immunology , Leptospira interrogans/physiology , Leptospirosis/metabolism , Leptospirosis/microbiology , Plasminogen/metabolism , Protein Binding , Protein Processing, Post-Translational
12.
Front Immunol ; 10: 591, 2019.
Article in English | MEDLINE | ID: mdl-30972077

ABSTRACT

Introduction: Acute ischemic stroke (AIS) is a potent trigger of immunosuppression, resulting in increased infection risk. While thrombolytic therapy with tissue-type plasminogen activator (t-PA) is still the only pharmacological treatment for AIS, plasmin, the effector protease, has been reported to suppress dendritic cells (DCs), known for their potent antigen-presenting capacity. Accordingly, in the major group of thrombolyzed AIS patients who fail to reanalyze (>60%), t-PA might trigger unintended and potentially harmful immunosuppressive consequences instead of beneficial reperfusion. To test this hypothesis, we performed an exploratory study to investigate the immunomodulatory properties of t-PA treatment in a mouse model of ischemic stroke. Methods: C57Bl/6J wild-type mice and plasminogen-deficient (plg-/-) mice were subjected to middle cerebral artery occlusion (MCAo) for 60 min followed by mouse t-PA treatment (0.9 mg/kg) at reperfusion. Behavioral testing was performed 23 h after occlusion, pursued by determination of blood counts and plasma cytokines at 24 h. Spleens and cervical lymph nodes (cLN) were also harvested and characterized by flow cytometry. Results: MCAo resulted in profound attenuation of immune activation, as anticipated. t-PA treatment not only worsened neurological deficit, but further reduced lymphocyte and monocyte counts in blood, enhanced plasma levels of both IL-10 and TNFα and decreased various conventional DC subsets in the spleen and cLN, consistent with enhanced immunosuppression and systemic inflammation after stroke. Many of these effects were abolished in plg-/- mice, suggesting plasmin as a key mediator of t-PA-induced immunosuppression. Conclusion: t-PA, via plasmin generation, may weaken the immune response post-stroke, potentially enhancing infection risk and impairing neurological recovery. Due to the large number of comparisons performed in this study, additional pre-clinical work is required to confirm these significant possibilities. Future studies will also need to ascertain the functional implications of t-PA-mediated immunosuppression for thrombolyzed AIS patients, particularly for those with failed recanalization.


Subject(s)
Fibrinolysin/immunology , Stroke/pathology , Thrombolytic Therapy/methods , Tissue Plasminogen Activator/immunology , Tissue Plasminogen Activator/therapeutic use , Animals , Cytokines/blood , Disease Models, Animal , Immunomodulation/immunology , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Cerebral Artery/pathology , Plasminogen/genetics
13.
J Thromb Haemost ; 17(9): 1461-1469, 2019 09.
Article in English | MEDLINE | ID: mdl-31136076

ABSTRACT

BACKGROUND: Excessive, plasmin-mediated fibrinolysis augments bleeding and contributes to death in some patients. Current therapies for fibrinolytic bleeding are limited by modest efficacy, low potency, and off-target effects. OBJECTIVES: To determine whether an antibody directed against unique loop structures of the plasmin protease domain may have enhanced specificity and potency for blocking plasmin activity, fibrinolysis, and experimental hemorrhage. METHODS: The binding specificity, affinity, protease cross-reactivity and antifibrinolytic properties of a monoclonal plasmin inhibitor antibody (Pi) were examined and compared with those of epsilon aminocaproic acid (EACA), which is a clinically used fibrinolysis inhibitor. RESULTS: Pi specifically recognized loopĀ 5 of the protease domain, and did not bind to other serine proteases or nine other non-primate plasminogens. Pi was ~7Ā logs more potent in neutralizing plasmin cleavage of small-molecule substrates and >3Ā logs more potent in quenching fibrinolysis than EACA. Pi was similarly effective in blocking catalysis of a small-molecule substrate as α2 -antiplasmin, which is the most potent covalent inhibitor of plasmin, and was a more potent fibrinolysis inhibitor. Fab or chimerized Fab fragments of Pi were equivalently effective. InĀ vivo, in a humanized model of fibrinolytic surgical bleeding, Pi significantly reduced bleeding to a greater extent than a clinical dose of EACA. CONCLUSIONS: A mAb directed against unique loop sequences in the protease domain is a highly specific, potent, competitive plasmin inhibitor that significantly reduces experimental surgical bleeding inĀ vivo.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antifibrinolytic Agents/therapeutic use , Fibrinolysin/antagonists & inhibitors , Hemorrhage/drug therapy , Aminocaproic Acid/pharmacology , Aminocaproic Acid/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Affinity , Binding, Competitive , Catalytic Domain/immunology , Cross Reactions , Drug Evaluation, Preclinical , Female , Fibrinolysin/chemistry , Fibrinolysin/immunology , Fibrinolysis/drug effects , Hemorrhage/blood , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Conformation , Protein Domains , Random Allocation , Recombinant Fusion Proteins/immunology , Species Specificity , Substrate Specificity
14.
Int J Parasitol ; 49(5): 337-346, 2019 04.
Article in English | MEDLINE | ID: mdl-30796952

ABSTRACT

Serine peptidases are involved in many physiological processes including digestion, haemostasis and complement cascade. Parasites regulate activities of host serine peptidases to their own benefit, employing various inhibitors, many of which belong to the Kunitz-type protein family. In this study, we confirmed the presence of potential anticoagulants in protein extracts of the haematophagous monogenean Eudiplozoon nipponicum which parasitizes the common carp. We then focused on a Kunitz protein (EnKT1) discovered in the E. nipponicum transcriptome, which structurally resembles textilinin-1, an antihemorrhagic snake venom factor from Pseudonaja textilis. The protein was recombinantly expressed, purified and biochemically characterised. The recombinant EnKT1 did inhibit in vitro activity of Factor Xa of the coagulation cascade, but exhibited a higher activity against plasmin and plasma kallikrein, which participate in fibrinolysis, production of kinins, and complement activation. Anti-coagulation properties of EnKT1 based on the inhibition of Factor Xa were confirmed by thromboelastography, but no effect on fibrinolysis was observed. Moreover, we discovered that EnKT1 significantly impairs the function of fish complement, possibly by inhibiting plasmin or Factor Xa which can act as a C3 and C5 convertase. We localised Enkt1 transcripts and protein within haematin digestive cells of the parasite by RNA in situ hybridisation and immunohistochemistry, respectively. Based on these results, we suggest that the secretory Kunitz protein of E. nipponicum has a dual function. In particular, it impairs both haemostasis and complement activation in vitro, and thus might facilitate digestion of a host's blood and protect a parasite's gastrodermis from damage by the complement. This study presents, to our knowledge, the first characterisation of a Kunitz protein from monogeneans and the first example of a parasite Kunitz inhibitor that impairs the function of the complement.


Subject(s)
Complement System Proteins/immunology , Fish Diseases/immunology , Helminth Proteins/immunology , Hemostasis , Trematoda/immunology , Trematode Infections/veterinary , Amino Acid Sequence , Animals , Anticoagulants/chemistry , Anticoagulants/immunology , Antifibrinolytic Agents/chemistry , Antifibrinolytic Agents/immunology , Carps/blood , Carps/immunology , Carps/parasitology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/immunology , Factor Xa/immunology , Factor Xa Inhibitors/chemistry , Factor Xa Inhibitors/immunology , Fibrinolysin/immunology , Fish Diseases/blood , Fish Diseases/parasitology , Helminth Proteins/chemistry , Helminth Proteins/genetics , Host-Parasite Interactions , Plasma Kallikrein/antagonists & inhibitors , Plasma Kallikrein/immunology , Sequence Alignment , Trematoda/chemistry , Trematoda/genetics , Trematode Infections/blood , Trematode Infections/immunology , Trematode Infections/parasitology
15.
Adv Exp Med Biol ; 632: 71-9, 2008.
Article in English | MEDLINE | ID: mdl-19025115

ABSTRACT

The complement system as a main column of innate immunity and the coagulation system as a main column in hemostasis undergo massive activation early after injury. Interactions between the two cascades have often been proposed but the precise molecular pathways of this interplay are still in the dark. To elucidate the mechanisms involved, the effects of various coagulation factors on complement activation and generation of anaphylatoxins were investigated and summarized in the light of the latest literature. Own in vitro findings suggest, that the coagulation factors FXa, FXIa and plasmin may cleave both C5 and C3, and robustly generate C5a and C3a (as detected by immunoblotting and ELISA). The produced anaphylatoxins were found to be biologically active as shown by a dose-dependent chemotactic response of neutrophils and HMC-1 cells, respectively. Thrombin did not only cleave C5 (Huber-Lang et al. 2006) but also in vitro-generated C3a when incubated with native C3. The plasmin-induced cleavage activity could be dose-dependently blocked by the serine protease inhibitor aprotinin and leupeptine. These findings suggest that various serine proteases belonging to the coagulation system are able to activate the complement cascade independently of the established pathways. Moreover, functional C5a and C3a are generated, both of which are known to be crucially involved in the inflammatory response.


Subject(s)
Blood Coagulation/immunology , Complement System Proteins/immunology , Anaphylatoxins/immunology , Animals , Complement Activation/immunology , Complement C1q/immunology , Complement C2/immunology , Complement C3/immunology , Complement C3a/immunology , Complement C4/immunology , Complement C4a/immunology , Complement C5/immunology , Complement C5a/immunology , Complement Factor B/immunology , Drug Interactions , Factor XIa/immunology , Factor Xa/immunology , Fibrinolysin/immunology , Hemostasis/immunology , Humans , Models, Biological , Serine Endopeptidases/immunology , Thrombin/immunology , Wound Healing/immunology
16.
EBioMedicine ; 5: 175-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27077125

ABSTRACT

Thrombus formation leading to vaso-occlusive events is a major cause of death, and involves complex interactions between coagulation, fibrinolytic and innate immune systems. Leukocyte recruitment is a key step, mediated partly by chemotactic complement activation factors C3a and C5a. However, mechanisms mediating C3a/C5a generation during thrombosis have not been studied. In a murine venous thrombosis model, levels of thrombin-antithrombin complexes poorly correlated with C3a and C5a, excluding a central role for thrombin in C3a/C5a production. However, clot weight strongly correlated with C5a, suggesting processes triggered during thrombosis promote C5a generation. Since thrombosis elicits fibrinolysis, we hypothesized that plasmin activates C5 during thrombosis. In vitro, the catalytic efficiency of plasmin-mediated C5a generation greatly exceeded that of thrombin or factor Xa, but was similar to the recognized complement C5 convertases. Plasmin-activated C5 yielded a functional membrane attack complex (MAC). In an arterial thrombosis model, plasminogen activator administration increased C5a levels. Overall, these findings suggest plasmin bridges thrombosis and the immune response by liberating C5a and inducing MAC assembly. These new insights may lead to the development of strategies to limit thrombus formation and/or enhance resolution.


Subject(s)
Arteries/immunology , Complement C5a/immunology , Fibrinolysin/immunology , Venous Thrombosis/immunology , Animals , Antithrombin III/drug effects , Antithrombin III/immunology , Arteries/drug effects , Arteries/pathology , Complement Activation/drug effects , Complement Activation/immunology , Complement C3a/biosynthesis , Complement C3a/immunology , Complement C5a/biosynthesis , Complement Membrane Attack Complex/drug effects , Complement Membrane Attack Complex/immunology , Factor Xa/immunology , Factor Xa/metabolism , Fibrinolysin/metabolism , Humans , Mice , Peptide Hydrolases/drug effects , Peptide Hydrolases/immunology , Plasminogen Activators/administration & dosage , Thrombin/immunology , Thrombin/metabolism , Venous Thrombosis/drug therapy , Venous Thrombosis/pathology
17.
Biochim Biophys Acta ; 525(1): 287-90, 1978 Jul 07.
Article in English | MEDLINE | ID: mdl-150860

ABSTRACT

Human thrombin-antithrombin III and plasmin-antiplasmin, two enzyme-inhibitor complexes composed of four different molecules, contain antigenic structures not present in the parent molecules, which can be directly quantitated in plasma with the use of non-cross-reacting antisera. It is anticipated that this neonatigenic expression is a more general phenomenon, which could provide a simple means of measuring activation of enzyme systems in biological fluids.


Subject(s)
Antigens , Fibrinolysin/immunology , Thrombin/immunology , Antigen-Antibody Reactions , Fibrinolysin/antagonists & inhibitors , Humans , Immunochemistry , Thrombin/antagonists & inhibitors , Urokinase-Type Plasminogen Activator
18.
Klin Oczna ; 107(7-9): 551-4, 2005.
Article in Polish | MEDLINE | ID: mdl-16417021

ABSTRACT

The eye is linked to the common mucosal immune system. This system play the part in preservation of the ocular surface. Tear fluid contains pro and anty-inflammatory factors such as lactoferrin, plasmin, immunoglobulins, and a lot of cytokines (interleukins, GM-CSF, TGF alfa and beta). The cornea is immunologicaly preferred, as a result of lack of resident lymphoreticular cells.


Subject(s)
Cytokines/immunology , Immunoglobulin A/immunology , Tears/immunology , Fibrinolysin/immunology , Humans , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/immunology
19.
J Thromb Haemost ; 13(4): 610-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25556624

ABSTRACT

BACKGROUND: The plasmin(ogen) and complement systems are simultaneously activated at sites of tissue injury, participating in hemostasis, wound healing, inflammation and immune surveillance. In particular, the C3 proteolytic fragment, iC3b, and its degradation product C3dg, which is generated by cleavage by factor I (FI) and the cofactor complement receptor CR1, are important in bridging innate and adaptive immunity. Via a thioester (TE) bond, iC3b and C3dg covalently tag pathogens, modulating phagocytosis and adaptive immune responses. OBJECTIVE: To examine plasmin-mediated proteolysis of iC3b, and to evaluate the functional consequences, comparing the effects with products generated by FI/CR1 cleavage of iC3b. METHODS: Dose-dependent and time-dependent plasmin-mediated cleavage of iC3b were characterized by analytical gel electrophoresis. The properties of the resultant TE bond-containing fragments on phagocytosis and induction of pro-inflammatory cytokines were measured in cell culture systems. RESULTS: At low concentrations, plasmin effectively cleaves iC3b, but at numerous previously undescribed sites, giving rise to novel C3c-like and C3dg-like moieties, the latter of which retain the TE bond. When attached to zymosan or erythrocytes and exposed to THP-1 macrophages, the C3dg-like proteins behave almost identically to the bona fide C3dg, yielding less phagocytosis as compared with the opsonin iC3b, and more macrophage secretion of the pro-inflammatory cytokine, IL-12. CONCLUSION: Plasmin cleavage of iC3b provides a complement regulatory pathway that is as efficient as FI/CR1 but does not require a cellular cofactor.


Subject(s)
Complement Activation , Complement C3 Convertase, Alternative Pathway , Complement C3b/metabolism , Fibrinolysin/metabolism , Fibrinolysis , Immunity, Innate , Macrophages/enzymology , Phagocytosis , Animals , Cell Line , Complement Activation/drug effects , Complement C3 Convertase, Alternative Pathway/drug effects , Complement C3b/immunology , Fibrinolysin/immunology , Fibrinolysin/pharmacology , Fibrinolysis/drug effects , Humans , Immunity, Innate/drug effects , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Macrophages/immunology , Peptide Fragments/immunology , Peptide Fragments/metabolism , Phagocytosis/drug effects , Proteolysis , Rabbits , Signal Transduction , Time Factors
20.
Thromb Haemost ; 39(1): 61-8, 1978 Feb 28.
Article in English | MEDLINE | ID: mdl-148118

ABSTRACT

An intermittent dosage scheme of streptokinase (standard initial dose 600,000 units SK infused over 30 min and repeated injections of 250,000 units SK at 24 hr intervals) was applied during 4 days in 9 patients with chronic obliterative arterial disease and in 8 patients with venous occlusion. Each dose of streptokinase produced an immediate fall in plasminogen to 17% (SEM 5.1) of the initial value, the level then rose to 50% (SEM 5.4) within 24 hr. Lowered levels of antiplasmin and fibrinogen (both less than 40% of the initial values) were maintained. This safe level of fibrinogen was maintained despite brief but high peaks of plasmin activity after each injection of SK. A parallel increase of the thrombin time and the fibrin (ogen) degradation products was obtained following each infusion. No bleeding was observed. The relative therapeutic effect of intermittent infusions of streptokinase has still to be compared with the continuous administration of streptokinase in controlled clinical trials.


Subject(s)
Arterial Occlusive Diseases/drug therapy , Streptokinase/therapeutic use , Thrombophlebitis/drug therapy , Antibodies , Antigen-Antibody Complex , Fibrin Fibrinogen Degradation Products/analysis , Fibrinogen/analysis , Fibrinolysin/immunology , Humans , Plasminogen/analysis
SELECTION OF CITATIONS
SEARCH DETAIL