Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
1.
J Thromb Haemost ; 17(9): 1461-1469, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31136076

RESUMEN

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 loop 5 of the protease domain, and did not bind to other serine proteases or nine other non-primate plasminogens. Pi was ~7 logs more potent in neutralizing plasmin cleavage of small-molecule substrates and >3 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. In 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 in vivo.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antifibrinolíticos/uso terapéutico , Fibrinolisina/antagonistas & inhibidores , Hemorragia/tratamiento farmacológico , Ácido Aminocaproico/farmacología , Ácido Aminocaproico/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Monoclonales Humanizados/farmacología , Afinidad de Anticuerpos , Unión Competitiva , Dominio Catalítico/inmunología , Reacciones Cruzadas , Evaluación Preclínica de Medicamentos , Femenino , Fibrinolisina/química , Fibrinolisina/inmunología , Fibrinólisis/efectos de los fármacos , Hemorragia/sangre , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Conformación Proteica , Dominios Proteicos , Distribución Aleatoria , Proteínas Recombinantes de Fusión/inmunología , Especificidad de la Especie , Especificidad por Sustrato
2.
Front Immunol ; 10: 591, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30972077

RESUMEN

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.


Asunto(s)
Fibrinolisina/inmunología , Accidente Cerebrovascular/patología , Terapia Trombolítica/métodos , Activador de Tejido Plasminógeno/inmunología , Activador de Tejido Plasminógeno/uso terapéutico , Animales , Citocinas/sangre , Modelos Animales de Enfermedad , Inmunomodulación/inmunología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Arteria Cerebral Media/patología , Plasminógeno/genética
3.
Int J Parasitol ; 49(5): 337-346, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30796952

RESUMEN

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.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Enfermedades de los Peces/inmunología , Proteínas del Helminto/inmunología , Hemostasis , Trematodos/inmunología , Infecciones por Trematodos/veterinaria , Secuencia de Aminoácidos , Animales , Anticoagulantes/química , Anticoagulantes/inmunología , Antifibrinolíticos/química , Antifibrinolíticos/inmunología , Carpas/sangre , Carpas/inmunología , Carpas/parasitología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/inmunología , Factor Xa/inmunología , Inhibidores del Factor Xa/química , Inhibidores del Factor Xa/inmunología , Fibrinolisina/inmunología , Enfermedades de los Peces/sangre , Enfermedades de los Peces/parasitología , Proteínas del Helminto/química , Proteínas del Helminto/genética , Interacciones Huésped-Parásitos , Calicreína Plasmática/antagonistas & inhibidores , Calicreína Plasmática/inmunología , Alineación de Secuencia , Trematodos/química , Trematodos/genética , Infecciones por Trematodos/sangre , Infecciones por Trematodos/inmunología , Infecciones por Trematodos/parasitología
4.
Blood ; 130(24): 2678-2681, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29021229

RESUMEN

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.


Asunto(s)
Activación de Complemento/inmunología , Proteínas del Sistema Complemento/inmunología , Fibrinolisina/inmunología , Trombina/inmunología , Animales , Activación de Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/metabolismo , Escherichia coli/inmunología , Escherichia coli/metabolismo , Factor Xa/inmunología , Factor Xa/farmacología , Fibrinolisina/metabolismo , Humanos , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Papio , Fosfatidilcolinas/inmunología , Fosfatidilcolinas/farmacología , Fosfatidilserinas/inmunología , Fosfatidilserinas/farmacología , Trombina/metabolismo
5.
EBioMedicine ; 5: 175-82, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27077125

RESUMEN

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.


Asunto(s)
Arterias/inmunología , Complemento C5a/inmunología , Fibrinolisina/inmunología , Trombosis de la Vena/inmunología , Animales , Antitrombina III/efectos de los fármacos , Antitrombina III/inmunología , Arterias/efectos de los fármacos , Arterias/patología , Activación de Complemento/efectos de los fármacos , Activación de Complemento/inmunología , Complemento C3a/biosíntesis , Complemento C3a/inmunología , Complemento C5a/biosíntesis , Complejo de Ataque a Membrana del Sistema Complemento/efectos de los fármacos , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Factor Xa/inmunología , Factor Xa/metabolismo , Fibrinolisina/metabolismo , Humanos , Ratones , Péptido Hidrolasas/efectos de los fármacos , Péptido Hidrolasas/inmunología , Activadores Plasminogénicos/administración & dosificación , Trombina/inmunología , Trombina/metabolismo , Trombosis de la Vena/tratamiento farmacológico , Trombosis de la Vena/patología
6.
Biochim Biophys Acta ; 1862(7): 1354-66, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27094771

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica/patología , Fibrinolisina/inmunología , Inflamación/patología , Metaloproteinasa 2 de la Matriz/inmunología , Plasminógeno/inmunología , Proteína Quinasa C-alfa/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Apoptosis , Barrera Hematoencefálica/inmunología , Línea Celular , Silenciador del Gen , Humanos , Inflamación/inmunología , Proteína Quinasa C-alfa/genética , Activador de Plasminógeno de Tipo Uroquinasa/inmunología
7.
Biochim Biophys Acta ; 1862(3): 395-402, 2016 03.
Artículo en Inglés | MEDLINE | ID: mdl-26493446

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica/inmunología , Enfermedades del Sistema Nervioso Central/inmunología , Inflamación/inmunología , Microglía/inmunología , Plasminógeno/inmunología , Animales , Barrera Hematoencefálica/patología , Enfermedades del Sistema Nervioso Central/patología , Fibrina/inmunología , Fibrinolisina/inmunología , Humanos , Inflamación/patología , Leucocitos/inmunología , Leucocitos/patología , Microglía/patología , Activador de Tejido Plasminógeno/inmunología
8.
J Thromb Haemost ; 13(4): 610-8, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25556624

RESUMEN

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.


Asunto(s)
Activación de Complemento , C3 Convertasa de la Vía Alternativa del Complemento , Complemento C3b/metabolismo , Fibrinolisina/metabolismo , Fibrinólisis , Inmunidad Innata , Macrófagos/enzimología , Fagocitosis , Animales , Línea Celular , Activación de Complemento/efectos de los fármacos , C3 Convertasa de la Vía Alternativa del Complemento/efectos de los fármacos , Complemento C3b/inmunología , Fibrinolisina/inmunología , Fibrinolisina/farmacología , Fibrinólisis/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Interleucina-12/inmunología , Interleucina-12/metabolismo , Macrófagos/inmunología , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Fagocitosis/efectos de los fármacos , Proteolisis , Conejos , Transducción de Señal , Factores de Tiempo
9.
Gastroenterology ; 148(3): 565-578.e4, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25490065

RESUMEN

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 motif 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.


Asunto(s)
Colitis/metabolismo , Fibrinolisina/antagonistas & inhibidores , Metaloproteinasa 9 de la Matriz/metabolismo , Células Mieloides/metabolismo , Animales , Antígenos CD40/antagonistas & inhibidores , Quimiocina CXCL5/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Sulfato de Dextran/toxicidad , Dipéptidos/farmacología , Modelos Animales de Enfermedad , Fibrinolisina/inmunología , Inflamación/inmunología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Noqueados , Células Mieloides/inmunología , Neutrófilos/inmunología , Ácido Trinitrobencenosulfónico/toxicidad , Factor de Necrosis Tumoral alfa/inmunología
10.
J Immunol ; 193(7): 3654-63, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25165151

RESUMEN

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.


Asunto(s)
Movimiento Celular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Fibrinolisina/inmunología , Quinasas Quinasa Quinasa PAM/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Monocitos/inmunología , Receptor PAR-1/inmunología , Receptores CCR2/inmunología , Animales , Benzoxazinas/farmacología , Movimiento Celular/efectos de los fármacos , Quimiocina CCL2/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , FN-kappa B/inmunología , Cavidad Pleural/inmunología , Receptores CCR2/antagonistas & inhibidores , Compuestos de Espiro/farmacología , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
11.
J Innate Immun ; 6(2): 240-50, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-23969887

RESUMEN

The globally significant human pathogen group A Streptococcus (GAS) sequesters the host protease plasmin to the cell surface during invasive disease initiation. Recent evidence has shown that localized plasmin activity prevents opsonization of several bacterial species by key components of the innate immune system in vitro. Here we demonstrate that plasmin at the GAS cell surface resulted in degradation of complement factor C3b, and that plasminogen acquisition is associated with a decrease in C3b opsonization and neutrophil-mediated killing in vitro. Furthermore, the ability to acquire cell surface plasmin(ogen) correlates directly with a decrease in C3b opsonization, neutrophil phagocytosis, and increased bacterial survival in a humanized plasminogen mouse model of infection. These findings demonstrate that localized plasmin(ogen) plays an important role in facilitating GAS escape from the host innate immune response and increases bacterial virulence in the early stages of infection.


Asunto(s)
Complemento C3b/inmunología , Neutrófilos/inmunología , Fagocitosis/inmunología , Plasminógeno/inmunología , Streptococcus pyogenes/inmunología , Animales , Western Blotting , Complemento C3b/metabolismo , Femenino , Fibrinolisina/inmunología , Fibrinolisina/metabolismo , Citometría de Flujo , Interacciones Huésped-Patógeno/inmunología , Humanos , Evasión Inmune/inmunología , Masculino , Ratones Transgénicos , Neutrófilos/metabolismo , Neutrófilos/microbiología , Plasminógeno/genética , Plasminógeno/metabolismo , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Streptococcus pyogenes/fisiología , Estreptoquinasa/inmunología , Estreptoquinasa/metabolismo
12.
J Biol Chem ; 288(35): 25229-25243, 2013 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-23861404

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/metabolismo , Borrelia burgdorferi/metabolismo , Plasminógeno/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Borrelia burgdorferi/química , Borrelia burgdorferi/genética , Borrelia burgdorferi/inmunología , Complemento C3b/química , Complemento C3b/genética , Complemento C3b/inmunología , Complemento C3b/metabolismo , Complemento C5/química , Complemento C5/genética , Complemento C5/inmunología , Complemento C5/metabolismo , Fibrinolisina/química , Fibrinolisina/genética , Fibrinolisina/inmunología , Fibrinolisina/metabolismo , Humanos , Inmunidad Innata , Enfermedad de Lyme/genética , Enfermedad de Lyme/inmunología , Enfermedad de Lyme/metabolismo , Plasminógeno/química , Plasminógeno/genética , Plasminógeno/inmunología , Unión Proteica , Estructura Terciaria de Proteína , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/inmunología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
14.
Rheumatology (Oxford) ; 52(8): 1448-53, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23598443

RESUMEN

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 ß2-glycoprotein I (ß2GPI) 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 ± 0.1 vs 1.9 ± 0.05 ng/ml (P < 0.0001), 6.6 ± 0.3 vs 2.1 ± 0.2 ng/ml (P < 0.0001) and 435 ± 34 vs 220 ± 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 ± 0.1 vs 1.9 ± 0.05 ng/ml; P = 0.0086, 6.2 ± 1.4 vs 2.2 ± 0.7 ng/ml; P = 0.147 and 412 ± 61 vs 260 ± 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.


Asunto(s)
Sangre Fetal/inmunología , Fibrinolisina/inmunología , Cardiopatías/inmunología , Lupus Eritematoso Sistémico/congénito , Receptores del Activador de Plasminógeno Tipo Uroquinasa/inmunología , Ribonucleoproteínas/inmunología , Anticuerpos Antinucleares/inmunología , Anticuerpos Antinucleares/metabolismo , Biomarcadores , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrinolisina/metabolismo , Cardiopatías/mortalidad , Cardiopatías/patología , Humanos , Inmunohistoquímica , Recién Nacido , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/mortalidad , Lupus Eritematoso Sistémico/patología , Macrófagos/inmunología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/metabolismo , Embarazo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/sangre , Valores de Referencia , Ribonucleoproteínas/metabolismo , Tasa de Supervivencia , Activador de Plasminógeno de Tipo Uroquinasa/sangre , Activador de Plasminógeno de Tipo Uroquinasa/inmunología
15.
J Biol Chem ; 288(10): 6849-63, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23341464

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/inmunología , Endopeptidasas/inmunología , Fibronectinas/inmunología , Evasión Inmune/inmunología , Inmunidad Innata/inmunología , Plasminógeno/inmunología , Adhesión Bacteriana/genética , Adhesión Bacteriana/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Western Blotting , Línea Celular Tumoral , Células Cultivadas , Complemento C3b/inmunología , Complemento C3b/metabolismo , Endopeptidasas/genética , Endopeptidasas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Fibrinolisina/inmunología , Fibrinolisina/metabolismo , Fibronectinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/inmunología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/microbiología , Humanos , Microscopía Confocal , Mutación , Plasminógeno/metabolismo , Unión Proteica , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/inmunología , Streptococcus pneumoniae/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/inmunología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
16.
J Leukoc Biol ; 92(3): 509-19, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22561604

RESUMEN

The serine protease plasmin generated from its zymogen plasminogen is best known for its function as a key enzyme of the fibrinolytic cascade. However, beyond fibrinolysis, plasmin has a number of crucial functions in a variety of processes, including inflammation. Various cells can bind plasminogen and plasmin via plasminogen-binding sites exposing a C-terminal lysine. Plasmin, generated as a result of plasminogen activation at the cell surface, is protected from its physiological inhibitors. Apart from its ability to facilitate cell migration in tissues, plasmin is capable of triggering signaling, which depends on cellular binding via its lysine-binding sites and its proteolytic activity. Plasmin-induced signaling affects various functions of monocytes, macrophages, DCs, and others, with the list of affected cells still growing. In vitro and in vivo studies have demonstrated the ability of plasmin to stimulate the production of cytokines, ROS, and other mediators, thereby contributing to inflammation. Plasmin-induced chemotaxis of monocytes and DCs indicates that it is also a potent chemoattractant for immune cells. Therefore, excessive activation of plasmin in chronic inflammatory or autoimmune diseases might exacerbate the activation of inflammatory cells and the pathogenesis of the disease. This review focuses on the available evidence for physiological and pathophysiological roles the serine protease plasmin in inflammatory processes.


Asunto(s)
Fibrinolisina/inmunología , Inflamación/inmunología , Transducción de Señal/inmunología , Animales , Humanos
17.
Exp Mol Pathol ; 92(1): 175-84, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22044461

RESUMEN

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.


Asunto(s)
Anexina A2/inmunología , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/patología , Mama/irrigación sanguínea , Neovascularización Patológica/prevención & control , Animales , Anexina A2/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/inmunología , Modelos Animales de Enfermedad , Femenino , Fibrinolisina/inmunología , Fibrinolisina/metabolismo , Humanos , Inmunohistoquímica , Ratones , Ratones Desnudos , Neovascularización Patológica/inmunología , Neovascularización Patológica/patología , Fenotipo , Proteínas Recombinantes , Activador de Tejido Plasminógeno/genética , Activador de Tejido Plasminógeno/metabolismo , Trasplante Heterólogo
18.
Microb Pathog ; 51(5): 360-5, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21802507

RESUMEN

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.


Asunto(s)
Fibrinolisina/inmunología , Evasión Inmune , Leptospira interrogans/patogenicidad , Leptospirosis/enzimología , Leptospirosis/inmunología , Fibrinolisina/metabolismo , Interacciones Huésped-Patógeno , Humanos , Leptospira interrogans/inmunología , Leptospira interrogans/fisiología , Leptospirosis/metabolismo , Leptospirosis/microbiología , Plasminógeno/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional
19.
J Biol Chem ; 286(26): 23559-69, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21566135

RESUMEN

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.


Asunto(s)
Artritis/metabolismo , Moléculas de Adhesión Celular/metabolismo , Heparina/metabolismo , Triptasas/metabolismo , alfa-Globulinas/inmunología , alfa-Globulinas/metabolismo , Animales , Artritis/inmunología , Biomarcadores/metabolismo , Células CHO , Moléculas de Adhesión Celular/inmunología , Cricetinae , Cricetulus , Fibrinolisina/inmunología , Fibrinolisina/metabolismo , Heparina/inmunología , Humanos , Articulaciones/inmunología , Articulaciones/metabolismo , Ratones , Triptasas/inmunología
20.
Thromb Haemost ; 104(1): 105-17, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20390229

RESUMEN

Plasmin, an active form of plasminogen, activates and inactivates factor VIII (FVIII) by limited proteolysis. We have previously identified lysine-binding site-independent plasmin-interactive sites on the FVIII A2 domain responsible for cleavages at Arg336 and Arg372, together with lysine-binding site-dependent plasmin sites on the light chain responsible for cleavage at Lys36. We have now characterised FVIII-interactive regions on plasmin. SDS-PAGE analysis demonstrated that a monoclonal antibody (mAb) against kringle (K)5-catalytic domain (K5-CD) of plasmin significantly blocked plasmin-catalysed cleavages at Arg336 and Arg372. K5-CD fragment and this mAb blocked plasmin-catalysed activation and inactivation of FVIII(a). Anti-K1-2-3 and anti-K4 mAbs blocked plasmin-catalysed cleavages at Lys36, and K1-2-3 and K4 fragments inhibited plasmin-catalysed inactivation of A11-336FVIIIa. The K5-CD preferentially bound to the A2 domain (Kdapp; 52 nM), whilst the K1-2-3 and K4 bound to the light chain (Kdapp; 75 and 106 nM, respectively) in ELISA. Binding was attributed to the A2 484-509 region and A3 1690-1705/1804-1818 region, respectively. 6-aminohexanoic acid, a lysine analogue, significantly inhibited the light chain/K1-2-3 (and K4) binding by approximately 90%, whilst A2/K5-CD binding was moderated by only approximately 35%. Furthermore, an anti-CD antibody blocked plasmin-catalysed cleavage by inhibiting the A2/K5-CD interaction. These data demonstrate that the K5-CD of plasmin (and plasminogen) interacts with the A2 domain independent of lysine-binding site, whilst interactions of K1-2-3 and K4 with the light chain are lysine-binding site-dependent. Interactions between the K5-CD and A2 likely constitute the major regulatory mechanism for activation and inactivation of FVIII(a) mediated by cleavage at Arg372 and Arg336.


Asunto(s)
Factor XIII/metabolismo , Fibrinolisina/metabolismo , Anticuerpos Bloqueadores/metabolismo , Biocatálisis , Coagulación Sanguínea , Activación Enzimática , Represión Enzimática , Fibrinolisina/inmunología , Humanos , Hidrólisis , Técnicas In Vitro , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...