Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(22)2022 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-36430180

RESUMEN

Factor-VII-activating protease (FSAP) is involved in the regulation of hemostasis and inflammation. Extracellular histones play a role in inflammation and the conversion of latent pro-FSAP into active FSAP. FSAP has been shown to regulate endothelial permeability, but the mechanisms are not clear. Here, we have investigated the effects of FSAP on endothelial permeability in vitro. A mixture of histones from calf thymus stimulated permeability, and the wild-type (WT) serine protease domain (SPD) of FSAP blocked this effect. WT-SPD-FSAP did not influence permeability on its own, nor that stimulated by thrombin or vascular endothelial growth factor (VEGF)-A165. Histones induced a large-scale rearrangement of the junction proteins VE-cadherin and zona occludens-1 from a clear junctional distribution to a diffuse pattern. The presence of WT-SPD-FSAP inhibited these changes. Permeability changes by histones were blocked by both TLR-2 and TLR4 blocking antibodies. Histones upregulated the expression of TLR-2, but not TLR-4, in HUVEC cells, and WT-SPD-FSAP abolished the upregulation of TLR-2 expression. An inactive variant, Marburg I (MI)-SPD-FSAP, did not have any of these effects. The inhibition of histone-mediated permeability may be an important function of FSAP with relevance to sepsis, trauma, and stroke and the need to be investigated further in in vivo experiments.


Asunto(s)
Histonas , Factor A de Crecimiento Endotelial Vascular , Humanos , Inflamación , Permeabilidad , Serina Endopeptidasas/metabolismo , Receptor Toll-Like 2/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
2.
Thromb Res ; 220: 24-34, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36265411

RESUMEN

Human genetic studies based on the Marburg I polymorphism in the factor VII activating protease (FSAP) encoding gene, analysis of FSAP activity in plasma and biochemical characterization of FSAP substrates indicate a possible causal link between FSAP activity and venous thrombosis. We hypothesized that a direct standardized assay to measure FSAP activity in plasma could provide convincing arguments for or against such a potential link. Using Ac-Pro-DTyr-Lys-Arg-AMC as a highly specific and sensitive substrate, histones as a trigger to activate pro-FSAP and plasma-purified active FSAP as a calibrator, we have developed a fluorogenic kinetic assay that reveals the FSAP generating potential in human plasma in real time. This assay is similar to the thrombin generation assay and allows analysis of lag phase, time to peak and velocity, as well as peak FSAP and the endogenous FSAP potential (EFP) of plasma samples. Carriers of the Marburg I polymorphism showed clearly delayed FSAP generation and lower peak FSAP and EFP level. There were no significant differences in all FSAP activity parameters between plasma from patients with a history of venous thrombosis and controls. When excluding Marburg I carriers, which were evenly distributed between groups, delayed FSAP generation significantly correlated with venous thrombosis in postmenopausal women. The novel FSAP activity assay is robust and easy to perform and will be a useful tool for analyzing plasma FSAP activity, also, in other pathophysiological conditions.


Asunto(s)
Factor VII , Trombosis de la Vena , Femenino , Humanos , Péptido Hidrolasas , Serina Endopeptidasas/genética , Trombina , Trombosis de la Vena/genética
3.
FASEB J ; 36(10): e22564, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36165219

RESUMEN

The outcome of ischemic stroke can be improved by further refinements of thrombolysis and reperfusion strategies. Factor VII activating protease (FSAP) is a circulating serine protease that could be important in this context. Its levels are raised in patients as well as mice after stroke and a single nucleotide polymorphism (SNP) in the coding sequence, which results in an inactive enzyme, is linked to an increased risk of stroke. In vitro, FSAP cleaves fibrinogen to promote fibrinolysis, activates protease-activated receptors, and decreases the cellular cytotoxicity of histones. Based on these facts, we hypothesized that FSAP can be used as a treatment for ischemic stroke. A combination of tissue plasminogen activator (tPA), a thrombolytic drug, and recombinant serine protease domain of FSAP (FSAP-SPD) improved regional cerebral perfusion and neurological outcome and reduced infarct size in a mouse model of thromboembolic stroke. FSAP-SPD also improved stroke outcomes and diminished the negative consequences of co-treatment with tPA in the transient middle cerebral artery occlusion model of stroke without altering cerebral perfusion. The inactive MI-isoform of FSAP had no impact in either model. FSAP enhanced the lysis of blood clots in vitro, but in the tail transection model of hemostasis, FSAP-SPD treatment provoked a faster clotting time indicating that it also has pro-coagulant actions. Thus, apart from enhancing thrombolysis, FSAP has multiple effects on stroke progression and represents a promising novel therapeutic strategy in the treatment of ischemic stroke.


Asunto(s)
Coagulantes , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Animales , Modelos Animales de Enfermedad , Factor VII , Fibrinógeno , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Histonas , Ratones , Péptido Hidrolasas , Receptores Proteinasa-Activados , Serina Endopeptidasas/genética , Accidente Cerebrovascular/tratamiento farmacológico , Activador de Tejido Plasminógeno/genética
4.
Thromb J ; 20(1): 13, 2022 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-35354492

RESUMEN

BACKGROUND: Coronavirus disease 2019 (COVID-19) infection is related to immune hyperactivity, the release of inflammatory cytokines, and immunothrombosis. Among the underlying mechanisms in COVID-19 thrombosis, neutrophil extracellular traps (NETs) formation, NETosis, may have a significant role. COVID-19 thrombi obtained from extracorporeal membrane oxygenation contained an accumulation of neutrophils and in a higher amount of NETs when compared with non-COVID-19 thrombi specimens. MAIN BODY: During sepsis and inflammatory status, NETs released from neutrophils and histones and nucleosomes extruded into the extracellular space and take part in the host innate immunity defense, inflammation, and thrombosis. Excessive NETosis is related to clinical progression and respiratory failure in infections and sepsis. NETosis act as a scaffold for thrombus formation, and new associative data support the relation between deregulated immune responses with thrombus formation and organ failure. NETosis is reported in COVID-19 patients. In COVID-19 infection, overproduction of tissue factor (TF) by neutrophils has a role in immunothrombosis. Additionally, NETs can trap TF pathway inhibitor (TFPI) as the only endogenous protein that effectively inhibits the activity of the significant proteases- complexes, TF-FVIIa and prothrombinase. CONCLUSION: Because of NETosis can induce intrinsic and extrinsic coagulation cascade activation through the production of TF, activation of FXII, and inhibition of TFPI and fibrinolysis and induce immunothrombosis, targeting NETosis may diminish thrombus formation related to NETs in COVID-19 patients.

5.
Mol Immunol ; 142: 95-104, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34973499

RESUMEN

BACKGROUND: Excessive bradykinin (BK) generation from high molecular weight kininogen (HK) by plasma kallikrein (PK) due to lack of protease inhibition is central to the pathophysiology of hereditary angioedema (HAE). Inadequate protease inhibition may contribute to HAE through a number of plasma proteases including factor VII activating protease (FSAP) that can also cleave HK. OBJECTIVE: To investigate the interaction between FSAP and C1 inhibitor (C1Inh) and evaluate the potential role of FSAP in HAE with C1Inh deficiency. MATERIALS AND METHODS: Plasma samples from 20 persons with HAE types 1 or 2 in remission were studied and compared to healthy controls. We measured and compared antigenic FSAP levels, spontaneous FSAP activity, FSAP generation potential, activation of plasma pre-kallikrein (PPK) by FSAP, and the formation of FSAP-C1Inh and FSAP-alpha2-antiplasmin (FSAP-α2AP) complexes. Furthermore, we measured HK cleavage and PK activation after activation of endogenous pro-FSAP and after addition of exogenous FSAP. RESULTS: In plasma from HAE patients, there is increased basal FSAP activity compared to healthy volunteers. HAE plasma exhibits decreased formation of FSAP-C1Inh complexes and increased formation of FSAP-α2AP complexes in histone-activated plasma. Although exogenous FSAP can cleave HK in plasma, this was not seen when endogenous plasma pro-FSAP was activated with histones in either group. PK was also not activated by FSAP in plasma. CONCLUSION: In this study, we established that FSAP activity is increased and the pattern of FSAP-inhibitor complexes is altered in HAE patients. However, we did not find evidence suggesting that FSAP contributes directly to HAE attacks.


Asunto(s)
Angioedemas Hereditarios/fisiopatología , Proteína Inhibidora del Complemento C1/genética , Quininógeno de Alto Peso Molecular/metabolismo , Serina Endopeptidasas/metabolismo , Angioedemas Hereditarios/sangre , Angioedemas Hereditarios/genética , Antifibrinolíticos/metabolismo , Bradiquinina/biosíntesis , Factor VII/metabolismo , Humanos , Calicreínas/sangre , Calicreínas/metabolismo , Serina Endopeptidasas/genética
6.
Pflugers Arch ; 474(2): 217-229, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34870751

RESUMEN

Proteolytic activation of the epithelial sodium channel (ENaC) by aberrantly filtered serine proteases is thought to contribute to renal sodium retention in nephrotic syndrome. However, the identity of the responsible proteases remains elusive. This study evaluated factor VII activating protease (FSAP) as a candidate in this context. We analyzed FSAP in the urine of patients with nephrotic syndrome and nephrotic mice and investigated its ability to activate human ENaC expressed in Xenopus laevis oocytes. Moreover, we studied sodium retention in FSAP-deficient mice (Habp2-/-) with experimental nephrotic syndrome induced by doxorubicin. In urine samples from nephrotic humans, high concentrations of FSAP were detected both as zymogen and in its active state. Recombinant serine protease domain of FSAP stimulated ENaC-mediated whole-cell currents in a time- and concentration-dependent manner. Mutating the putative prostasin cleavage site in γ-ENaC (γRKRK178AAAA) prevented channel stimulation by the serine protease domain of FSAP. In a mouse model for nephrotic syndrome, active FSAP was present in nephrotic urine of Habp2+/+ but not of Habp2-/- mice. However, Habp2-/- mice were not protected from sodium retention compared to nephrotic Habp2+/+ mice. Western blot analysis revealed that in nephrotic Habp2-/- mice, proteolytic cleavage of α- and γ-ENaC was similar to that in nephrotic Habp2+/+ animals. In conclusion, active FSAP is excreted in the urine of nephrotic patients and mice and activates ENaC in vitro involving the putative prostasin cleavage site of γ-ENaC. However, endogenous FSAP is not essential for sodium retention in nephrotic mice.


Asunto(s)
Canales Epiteliales de Sodio/metabolismo , Factor VII/metabolismo , Riñón/metabolismo , Síndrome Nefrótico/metabolismo , Péptido Hidrolasas/metabolismo , Sodio/metabolismo , Animales , Doxorrubicina/metabolismo , Doxorrubicina/farmacología , Humanos , Transporte Iónico/efectos de los fármacos , Transporte Iónico/fisiología , Riñón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteolisis/efectos de los fármacos , Serina Endopeptidasas/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Xenopus laevis/metabolismo
7.
Thromb Res ; 196: 193-199, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32891905

RESUMEN

INTRODUCTION: Extracellular histones inhibit tissue plasminogen activator (t-PA)-mediated fibrinolysis by modifying fibrin structure and rheological properties. However, other plasminogen activators involved in intravascular and extravascular fibrinolysis have not been considered yet. OBJECTIVES: We investigated the effect of histones on fibrinolysis driven by different plasminogen activators. METHODS: Clot lysis induced by t-PA, urokinase (u-PA) and its single chain precursor (scu-PA) was evaluated by turbidimetry. Conversion of scu-PA to u-PA and activation of factor seven activating protease (FSAP) were assessed by fluorogenic and chromogenic assays, respectively. RESULTS: Histones delayed t-PA- and u-PA-mediated fibrinolysis but strongly accelerated scu-PA-driven clot lysis through the enhancement of scu-PA to u-PA conversion. This effect required a plasma factor identified as FSAP by the following findings: 1) histones enhanced neither scu-PA activation nor scu-PA-mediated clot lysis under purified conditions; 2) in plasma, the enhancement of fibrinolytic activity by histones was abolished by a neutralizing anti-FSAP antibody; and 3) histones promoted the activation of plasma FSAP. The effect of the natural mixture of histones on scu-PA-driven fibrinolysis was differentially recapitulated by the individual recombinant histones, H4 displaying the strongest activity. When complexed to DNA, histones still accelerated scu-PA-mediated fibrinolysis but with a lesser efficiency due to a reduced FSAP activation. Finally, preincubation of histones with heparin or activated protein C, two known inhibitors of histones, further amplified histone-mediated boost of scu-PA-driven fibrinolysis. CONCLUSIONS: Enhancement of FSAP-mediated scu-PA activity by histones may play yet unforeseen roles in intravascular fibrinolysis and contribute to extravascular proteolysis and tissue damage.


Asunto(s)
Histonas , Activador de Plasminógeno de Tipo Uroquinasa , Factor VII , Fibrinólisis , Humanos , Péptido Hidrolasas , Activador de Tejido Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
8.
J Am Heart Assoc ; 9(17): e016445, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32856552

RESUMEN

Background Factor VII activating protease (FSAP) is of interest as a marker for vascular inflammation and plaque destabilization. The aim of this study was to analyze the expression profile of FSAP in endarterectomy specimens that were taken from patients with asymptomatic and symptomatic carotid atherosclerotic plaques and to compare them with circulating FSAP levels. Methods and Results Plasma FSAP concentration, activity, and mRNA expression were measured in endarterectomy specimens and in monocytes and platelets. Plaque and plasma FSAP levels were higher in symptomatic patients (n=10) than in asymptomatic patients (n=14). Stronger FSAP immunostaining was observed in advanced symptomatic lesions, in intraplaque hemorrhage-related structures, and in lipid-rich areas within the necrotic core. FSAP was also colocalized with monocytes and macrophages (CD11b/CD68-positive cells) and platelets (CD41-positive cells) of the plaques. Moreover, human platelets expressed FSAP in vitro, at both the mRNA and protein levels. Expression is stimulated by thrombin receptor-activating peptide and ADP and reduced by acetylsalicylic acid. Conclusions Plasma FSAP levels were significantly increased in patients with symptomatic carotid stenosis and thus may be involved in plaque development This plaque-associated FSAP may be produced by platelets or macrophages or may be taken up from the circulation. To establish FSAP's utility as a circulating or plaque biomarker in patients with symptomatic carotid atherosclerotic plaques, further studies are needed.


Asunto(s)
Arterias Carótidas/patología , Estenosis Carotídea/patología , Factor VII/metabolismo , Placa Aterosclerótica/metabolismo , Anciano , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Plaquetas/metabolismo , Estenosis Carotídea/cirugía , Estudios de Casos y Controles , Endarterectomía Carotidea/métodos , Femenino , Humanos , Inflamación/metabolismo , Macrófagos/metabolismo , Masculino , Persona de Mediana Edad , Monocitos/metabolismo , Fragmentos de Péptidos/metabolismo , Péptido Hidrolasas/metabolismo , Placa Aterosclerótica/tratamiento farmacológico , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , ARN Mensajero/metabolismo
9.
Chembiochem ; 21(13): 1875-1884, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32180321

RESUMEN

We describe a novel, easy and efficient combinatorial phage display peptide substrate-mining method to map the substrate specificity of proteases. The peptide library is displayed on the pVII capsid of the M13 bacteriophage, which renders pIII necessary for infectivity and efficient retrieval, in an unmodified state. As capture module, the 3XFLAG was chosen due to its very high binding efficiency to anti-FLAG mAbs and its independency of any post-translational modification. This library was tested with Factor-VII activating protease (WT-FSAP) and its single-nucleotide polymorphism variant Marburg-I (MI)-FSAP. The WT-FSAP results confirmed the previously reported Arg/Lys centered FSAP cleavage site consensus as dominant, as well as reinforcing MI-FSAP as a loss-of-function mutant. Surprisingly, rare substrate clones devoid of basic amino acids were also identified. Indeed one of these peptides was cleaved as free peptide, thus suggesting a broader range of WT-FSAP substrates than previously anticipated.


Asunto(s)
Péptidos/metabolismo , Serina Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Cromatografía Líquida de Alta Presión , Humanos , Espectrometría de Masas , Biblioteca de Péptidos , Péptidos/análisis , Péptidos/química , Especificidad por Sustrato
10.
Thromb Res ; 188: 74-78, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32087413

RESUMEN

Factor VII activating protease (FSAP) is a circulating serine protease of broad specificity that is likely to be involved in many pathophysiological processes. The activation of the circulating zymogen form of FSAP by histones, released from damaged cells, underlines its roles in regulating host responses to tissue damage and inflammation. Some of the direct cellular effects of FSAP are mediated through protease-activated receptors (PARs). Knock-down of each one of the four PARs in endothelial cells indicated that PAR-1 and -3 are involved in regulating endothelial permeability in response to FSAP. Overexpression of PARs in cell lines led to the conclusion that PAR-2 and -1 were the main receptors for FSAP. Studies with synthetic peptides and receptor mutants demonstrate that FSAP cleaves PAR-1 and -2 at their canonical cleavage site. However, PAR-1 is not activated by FSAP in all cells, which may be related to other, as yet, undefined factors. Inhibition of apoptosis by FSAP is mediated through PAR-1 and was observed in neurons, astrocytes and A549 cells. FSAP also mediates cellular effects by modulating the activity of growth factors, generation of bradykinin, C5a and C3a generation or histone inactivation. These cellular effects need to be further investigated at the in vivo level.


Asunto(s)
Factor VII , Péptido Hidrolasas , Células Endoteliales , Receptor PAR-1/genética , Serina Endopeptidasas/genética
11.
FASEB J ; 34(1): 1079-1090, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914657

RESUMEN

Factor VII activating protease (FSAP) is a circulating serine protease implicated in thrombosis, atherosclerosis, stroke, and cancer. Using an overexpression strategy, we have systematically investigated the role of protease activated receptors (PAR)-1, -2, -3, and -4 on FSAP-mediated signaling in HEK293T and A549 cells. Cleavage of PAR-reporter constructs and MAPK phosphorylation was used to monitor receptor activation. FSAP cleaved PAR-2 and to a lesser degree PAR-1, but not PAR-3 or PAR-4 in both cell types. Robust MAPK activation in response to FSAP was observed after PAR-2, but not PAR-1 overexpression in HEK293T. Recombinant serine protease domain of wild type FSAP, but not the Marburg I isoform of FSAP, could reproduce the effects of plasma purified FSAP. Canonical cleavage of both PARs was suggested by mass spectrometric analysis of synthetic peptide substrates from the N-terminus of PARs and site directed mutagenesis studies. Surprisingly, knockdown of endogenous PAR-1, but not PAR-2, prevented the apoptosis-inhibitory effect of FSAP, suggesting that PAR1 is nevertheless a direct or indirect target in some cell types. This molecular characterization of PAR-1 and -2 as cellular receptors of FSAP will help to define the actions of FSAP in the context of cancer and vascular biology.


Asunto(s)
Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Serina Endopeptidasas/metabolismo , Apoptosis , Línea Celular Tumoral , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Mutagénesis Sitio-Dirigida , Péptidos/química , Fosforilación , Isoformas de Proteínas , Transducción de Señal , Trombosis
12.
Thromb Res ; 182: 124-132, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31479940

RESUMEN

The zymogen form of circulating Factor VII activating protease (FSAP) is activated by histones that are released as a consequence of tissue damage or excessive inflammation. This is likely to have consequences in a number of disease conditions such as stroke, atherosclerosis, liver fibrosis, thrombosis and cancer. To investigate the existence, as well as the concentration of active FSAP (FSAPa) in complex biological systems an active site probe is needed. We used Hybrid Combinatorial Substrate Library (HyCoSuL) to screen for natural and unnatural amino acids that specifically bind to P4-P2 pockets of FSAPa. This information was used to designing a fluorogenic substrate (Ac-Pro-DTyr-Lys-Arg-ACC) as well as an irreversible, fluorogenic activity-based probe Cy5-6-Ahx-Pro-DTyr-Lys-ArgP(OPh)2. In normal human plasma the probe showed very low non-specific reactivity with some plasma proteins but upon activation of pro-FSAP with histones, strong labelling of FSAPa was observed. This labelling could be inhibited by aprotinin and was not found in the plasma of a subject that was homozygous for a polymorphism, which leads to loss of activity, or in plasma that was depleted of FSAP by antibodies. This 2nd generation substrate exhibited 6-fold higher catalytic efficiency than the 1st generation substrate and a much higher selectivity for FSAPa over other plasma proteases. This substrate and probe can be useful to detect and localize FSAPa in normal and pathological tissue and plasma to gain more insight into its functions.


Asunto(s)
Carbocianinas/química , Colorantes Fluorescentes/química , Serina Endopeptidasas/sangre , Aminoácidos/química , Pruebas de Enzimas/métodos , Humanos , Oligopéptidos/química , Serina Endopeptidasas/análisis , Especificidad por Sustrato
13.
Biochim Biophys Acta Mol Basis Dis ; 1864(10): 3397-3406, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30076961

RESUMEN

Factor VII Activating Protease (FSAP) is a plasma protease affecting both coagulation and fibrinolysis. Although a role in hemostasis is still unclear, the identification of additional physiologic substrates will help to elucidate its role in this context. FSAP has been reported to cleave fibrinogen, but the functional consequences of this are not known. We have therefore undertaken this study to determine the implications of this cleavage for fibrin-clot formation and its lysis. Treatment of human fibrinogen with FSAP released an N-terminal peptide from the Bß chain (Bß1-53) and subsequently the fibrinopeptide B; within the Aα chain a partial truncation of the αC-region by multiple cleavages was seen. The truncated fibrinogen showed a delayed thrombin-catalyzed polymerization and formed fibrin clots of reduced turbidity, indicative of thinner fibrin fibers. Confocal laser scanning and scanning electron microscopy of these clots revealed a less coarse fibrin network with thinner fibers and a smaller pore size. A lower pore size was also seen in permeability studies. Unexpectedly, FSAP-treated fibrinogen or plasma exhibited a significantly faster tPA-driven lysis, which correlated exclusively with cleavage of fibrinogen and not with activation of plasminogen activators. Similar observations were also made in plasma after activation of endogenous zymogen FSAP, but not in plasma of carrier of the rare Marburg I single nucleotide polymorphism. In conclusion, altering fibrin clot properties by fibrinogenolysis is a novel function of FSAP in the vasculature, which facilitates clot lysis and may in vivo contribute to reduced fibrin deposition during thrombosis.


Asunto(s)
Fibrinógeno/química , Fibrinógeno/metabolismo , Serina Endopeptidasas/metabolismo , Coagulación Sanguínea , Fibrinólisis , Fibrinopéptido B/metabolismo , Humanos , Microscopía Confocal , Microscopía Electrónica de Rastreo
14.
Thromb Res ; 161: 36-42, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29178989

RESUMEN

The circulating zymogen form of Factor VII activating protease (FSAP) can be activated by histones and nucleosomes in vivo. These cell-death-associated nuclear factors are also actively extruded into the extracellular space by neutrophils through a process called neutrophil extracellular trap (NET) formation (NETosis). NETs are thought to be involved in host defense, inflammation as well as thrombosis. We have investigated the bidirectional interactions of FSAP and NETs. Phorbol ester-mediated NET formation was marginally stimulated by FSAP. Plasma-derived FSAP as well as exogenous FSAP bound to NETs. There was co-localization of FSAP and NETs in coronary thrombi from patients with acute myocardial infarction. Contrary to our expectations no activation of pro-FSAP by NETs was evident. However, after disintegration of NETs with DNase, a robust activation of pro-FSAP, due to release of histones from nucleosomes, was detected. The released histones were in turn degraded by FSAP. Histone cytotoxicity towards endothelial cells was neutralized by FSAP more potently than by activated protein C (APC). One more consequence of histone degradation was a decrease in nucleosome release from apoptotic neutrophils. Taken together, NETs bind to FSAP, but do not activate pro-FSAP unless histones are released from NETs by DNAse. This activation of FSAP is likely to be important in diminishing the cytotoxic effect of histones, thus limiting the damaging effect of NETosis.


Asunto(s)
Trampas Extracelulares/metabolismo , Neutrófilos/metabolismo , Serina Endopeptidasas/sangre , Trombosis/sangre , Trombosis Coronaria/sangre , Trombosis Coronaria/patología , Desoxirribonucleasas/metabolismo , Factor VII/metabolismo , Histonas/sangre , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/sangre , Neutrófilos/efectos de los fármacos , Neutrófilos/enzimología , Neutrófilos/patología , Nucleosomas/metabolismo , Acetato de Tetradecanoilforbol/farmacología
15.
Atherosclerosis ; 265: 133-139, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28881271

RESUMEN

BACKGROUND AND AIMS: The factor VII activating protease (FSAP) knockout mice have a bigger neointima after vascular injury and a larger infarct volume after stroke. The Marburg I (MI) single nucleotide polymorphism (SNP) in the FSAP-encoding gene is associated with an increased risk of stroke and carotid stenosis in humans. We hypothesize that the regulation of gene expression by FSAP in vascular cells accounts for its vasculo-regulatory properties. METHODS: Vascular smooth muscle cells (VSMC) and endothelial cells (EC) were stimulated with FSAP and a microarray-based expression analysis was performed. Selected genes were further investigated by qPCR. Receptor- and pathway-inhibitors were used to elucidate the mechanisms involved. RESULTS: Pathways significantly activated by FSAP include those related to inflammation, apoptosis and cell growth in VSMC and inflammation in EC. The key upregulated genes in VSMC were AREG, PTGS2 and IL6; and in EC these were SELE, VCAM1, and IL8. Secretion of IL6 in VSMC and IL8 in EC was also stimulated by FSAP. Recombinant wild type protease domain of FSAP, but not the MI-isoform, could recapitulate most of these effects. In VSMC, but not EC, gene expression by FSAP was impaired by PAR1 (protease-activated receptor1) receptor antagonists. In VSMC, FSAP-induced expression of AREG and IL6 was blocked by cAMP and MAPK pathway inhibitors indicating that multiple signalling pathways are likely to be involved. CONCLUSIONS: The stimulation of inflammation- and proliferative/apoptosis-related genes in VSMC and EC provides a comprehensive basis for understanding the role of FSAP in vascular diseases.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/enzimología , Mediadores de Inflamación/metabolismo , Inflamación/enzimología , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Serina Endopeptidasas/metabolismo , Anfirregulina/genética , Anfirregulina/metabolismo , Apoptosis/genética , Proliferación Celular/genética , Células Cultivadas , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/metabolismo , Selectina E/genética , Selectina E/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inflamación/genética , Inflamación/patología , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Músculo Liso Vascular/patología , Receptor PAR-1/genética , Receptor PAR-1/metabolismo , Serina Endopeptidasas/genética , Transducción de Señal , Factores de Tiempo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
J Biol Chem ; 292(15): 6381-6388, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28246168

RESUMEN

HABP2 (hyaluronan-binding protein 2) is a Ca2+-dependent serine protease with putative roles in blood coagulation and fibrinolysis. A G221E substitution, known as the Marburg I polymorphism, reportedly affects HABP2 function and has been associated with increased risk for cardiovascular disease. However, the importance of Gly-221 for HABP2 activity is unclear. Here, we used G221E, G221A, and G221S mutants to assess the role of Gly-221 in HABP2 catalysis. The G221E variant failed to activate the single-chain urokinase-type plasminogen activator, and the G221A and G221S variants displayed moderately reduced single-chain urokinase-type plasminogen activator activation. Activity toward the peptide substrate S-2288 was markedly decreased in all HABP2 variants, with G221E being the most defective and G221A being the least defective. In the absence of Ca2+, S-2288 cleavage by wild-type HABP2 was Na+-dependent, with Km decreasing from 3.0 to 0.6 mm upon titration from 0 to 0.3 m Na+ In the presence of 5 mm Ca2+, Km was further reduced to 0.05 mm, but without an appreciable contribution of Na+ At physiological concentrations of Na+ and Ca2+, the three HABP2 variants, and particularly G221E, displayed a major Km increase for S-2288. Chemical footprinting revealed that Ile-16 is significantly less protected from chemical modification in G221E than in wild-type HABP2, suggesting impaired insertion of the N terminus into the G221E protease domain, with a concomitant impact on catalytic activity. Homology modeling suggested that the Glu-221 side chain could sterically hinder insertion of the N terminus into the HABP2 protease domain, helping to explain the detrimental effects of Glu-221 substitution on HABP2 activity.


Asunto(s)
Serina Endopeptidasas/química , Sustitución de Aminoácidos , Calcio/química , Catálisis , Glicina/química , Glicina/genética , Mutación Missense , Dominios Proteicos , Serina Endopeptidasas/genética , Activador de Plasminógeno de Tipo Uroquinasa/química , Activador de Plasminógeno de Tipo Uroquinasa/genética
17.
Blood Rev ; 30(4): 257-61, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26776504

RESUMEN

Disseminated intravascular coagulation (DIC) is a heterogeneous group of disorders, which manifest as a spectrum of haemorrhage and thrombosis complicating many primary conditions including sepsis, trauma and malignancies. The pathophysiology of this condition is complex. In the recent years there is growing evidence that damage associated molecular patterns (DAMPs) play a crucial role in the pathogenesis of DIC. Upon cell-death and/or cell activation of hematopoietic and parenchymal cells extracellular cell-free DNA as well as DNA binding proteins (e.g. histones and high mobility group box 1 protein [HMGB1]) are released into circulation. This release is a highly regulated process mediated among others by serine proteases, such as factor VII-activating protease (FSAP) and DNase1. Circulating cell-free DNA has been demonstrated to influence primary and secondary hemostasis by inducing platelet aggregation, promoting coagulation activation, inhibition of fibrinolysis and directly interfering with clot stability. In this respect cell-free DNA in tissue as well as released into the circulation after neutrophil activation in the form of neutrophil extracellular traps (NETs) has been shown to be cytotoxic and highly procoagulant. DNA-binding proteins such as histones and HMGB1 are also strongly procoagulant and are involved in the pathogenesis of DIC. The present review gives an overview on how extracellular DNA is released into circulation and the structure of circulating DNA. In addition it summarizes the effect of extracellular DNA and DNA-binding proteins on platelet activation, plasmatic coagulation as well as fibrinolysis.


Asunto(s)
Coagulación Sanguínea/genética , Coagulación Intravascular Diseminada/genética , Activación Neutrófila/genética , Proteínas de Unión al ADN , Humanos
18.
J Thromb Haemost ; 13(1): 41-6, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25370187

RESUMEN

BACKGROUND: The plasma protease factor VII-activating protease (FSAP) can release nucleosomes from late apoptotic cells. Nucleosomes are markers of cell death, and extracellular cell-free DNA has been suggested to play an important role in inflammation and has been demonstrated to correlate with severity and outcome in sepsis patients. OBJECTIVE: To investigate FSAP activation in patients suffering from Burkholderia pseudomallei infection (melioidosis), an important cause of Gram-negative sepsis in Southeast Asia. As diabetes mellitus (DM) is the most important risk factor for both melioidosis and sepsis, we were also able to examine the role of DM in FSAP activation in this cohort of patients. METHODS: In a prospective observational study, complexes of FSAP with α2 -antiplasmin (AP) were assayed in 44 patients with melioidosis, 34 of whom were classified as diabetic. Eighty-two healthy subjects served as controls (52 with DM and 30 without). RESULTS: FSAP-AP complex levels were markedly elevated in patients as compared with controls. The FSAP level increased by 16.82 AU mL(-1) in patients with melioidosis after adjustment for the effect of DM in the regression model. As expected, FSAP activation was correlated with nucleosome release (slope = 0.74). No difference in FSAP activation on admission was seen between survivors and non-survivors, but the extent of FSAP activation correlated with stage of the disease; repeated testing during convalescence showed a return towards normal values (day 0 vs. day 28, 4.16 AU mL(-1) , 95% confidence interval [CI] 1.42-12.22). CONCLUSION: Patients with Gram-negative sepsis caused by B. pseudomallei have abundant FSAP activation, which significantly correlates with stage of disease. The presence of DM, however, does not influence the extent of FSAP activation.


Asunto(s)
Diabetes Mellitus/enzimología , Melioidosis/enzimología , Serina Endopeptidasas/sangre , Adolescente , Adulto , Anciano , Biomarcadores/sangre , Estudios de Casos y Controles , Diabetes Mellitus/sangre , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/epidemiología , Activación Enzimática , Femenino , Humanos , Masculino , Melioidosis/sangre , Melioidosis/diagnóstico , Melioidosis/epidemiología , Melioidosis/microbiología , Persona de Mediana Edad , Nucleosomas/metabolismo , Estudios Prospectivos , Unión Proteica , Tailandia/epidemiología , Adulto Joven , alfa 2-Antiplasmina/metabolismo
19.
Thromb Haemost ; 113(4): 870-80, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25427855

RESUMEN

Factor VII (FVII) activating protease (FSAP) is a circulating protease with a putative function in blood coagulation and fibrinolysis. Genetic epidemiological studies have implied a role for FSAP in carotid stenosis, stroke and thrombosis. To date, no in vivo evidence is available to support these claims. We have, for the first time, used FSAP-/- mice to define its role in thrombosis and haemostasis in vivo and to characterise the molecular mechanisms involved. FeCl3-induced arterial thrombosis in carotid and mesenteric artery revealed that the occlusion time was significantly increased in FSAP-/- mice (p< 0.01) and that some FSAP-/- mice did not occlude at all. FSAP-/- mice were protected from lethal pulmonary thromboembolism induced by collagen/ epinephrine infusion (p< 0.01). Although no spontaneous bleeding was evident, in the tail bleeding assay a re-bleeding pattern was observed in FSAP-/- mice. To explain these observations at a mechanistic level we then determined how haemostasis factors and putative FSAP substrates were altered in FSAP-/- mice. Tissue factor pathway inhibitor (TFPI) levels were higher in FSAP-/- mice compared to WT mice whereas FVIIa levels were unchanged. Other coagulation factors as well as markers of platelet activation and function revealed no significant differences between WT and FSAP-/- mice. This phenotype of FSAP-/- mice could be reversed by application of exogenous FSAP. In conclusion, a lack of endogenous FSAP impaired the formation of stable, occlusive thrombi in mice. The underlying in vivo effect of FSAP is more likely to be related to the modulation of TFPI rather than FVIIa.


Asunto(s)
Enfermedades de las Arterias Carótidas/prevención & control , Hemostasis , Oclusión Vascular Mesentérica/prevención & control , Serina Endopeptidasas/deficiencia , Trombosis/prevención & control , Tromboembolia Venosa/enzimología , Animales , Pruebas de Coagulación Sanguínea , Arterias Carótidas/enzimología , Enfermedades de las Arterias Carótidas/sangre , Enfermedades de las Arterias Carótidas/inducido químicamente , Enfermedades de las Arterias Carótidas/enzimología , Enfermedades de las Arterias Carótidas/genética , Cloruros , Colágeno , Modelos Animales de Enfermedad , Compuestos Férricos , Predisposición Genética a la Enfermedad , Hemostasis/genética , Venas Yugulares/enzimología , Lipoproteínas/sangre , Arterias Mesentéricas/enzimología , Oclusión Vascular Mesentérica/sangre , Oclusión Vascular Mesentérica/inducido químicamente , Oclusión Vascular Mesentérica/enzimología , Oclusión Vascular Mesentérica/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Norepinefrina , Fenotipo , Serina Endopeptidasas/administración & dosificación , Serina Endopeptidasas/genética , Trombosis/sangre , Trombosis/inducido químicamente , Trombosis/enzimología , Trombosis/genética , Tromboembolia Venosa/sangre , Tromboembolia Venosa/inducido químicamente , Tromboembolia Venosa/genética
20.
Proteomics Clin Appl ; 8(5-6): 438-46, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24497464

RESUMEN

Factor VII activating protease (FSAP) is a multifunctional serine protease that is mainly synthesized and secreted by hepatocytes. This enzyme is highly evolutionarily conserved and contains three epidermal growth factor like domains, a kringle domain and a trypsin-like serine protease signature at its C-terminus. Animal experimentation and clinical findings indicate that FSAP influences a range of inflammatory fibroproliferative diseases. In particular, recent work demonstrated that FSAP is anti-fibrotic and influences liver fibrosis progression. The relative high physiological concentration, occurrence of gene variants affecting the proteolytic activity of FSAP and eclectic substrate specificity should in principal presuppose this protease to be frequently found in studies in which quantitative proteomics is performed. However, presently there are only a few studies available that have identified FSAP in applications using 2D gels, MS or other proteomic-associated techniques. We summarize here the actual knowledge about FSAP functions in initiation and progression of hepatic fibrosis and comment on proteome studies in which altered expression or activity of FSAP was reported.


Asunto(s)
Cirrosis Hepática/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Progresión de la Enfermedad , Humanos , Cirrosis Hepática/tratamiento farmacológico , Proteómica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...