Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
EMBO J ; 36(3): 260-273, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28007894

RESUMEN

The F-box protein FBXW7 is the substrate-recruiting subunit of an SCF ubiquitin ligase and a major tumor-suppressor protein that is altered in several human malignancies. Loss of function of FBXW7 results in the stabilization of numerous proteins that orchestrate cell proliferation and survival. Little is known about proteins that directly regulate the function of this protein. In the current work, we have mapped the interactome of the enigmatic pseudophosphatase STYX We reasoned that a catalytically inactive phosphatase might have adopted novel mechanisms of action. The STYX interactome contained several F-box proteins, including FBXW7. We show that STYX binds to the F-box domain of FBXW7 and disables its recruitment into the SCF complex. Therefore, STYX acts as a direct inhibitor of FBXW7, affecting the cellular levels of its substrates. Furthermore, we find that levels of STYX and FBXW7 are anti-correlated in breast cancer patients, which affects disease prognosis. We propose the STYX-FBXW7 interaction as a promising drug target for future investigations.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas F-Box/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Ligasas SKP Cullina F-box/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/patología , Proteína 7 que Contiene Repeticiones F-Box-WD , Humanos
5.
Haematologica ; 106(10): 2641-2653, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32703799

RESUMEN

The recruitment of neutrophils from the microvasculature to the site of injury or infection represents a key event in the inflammatory response. Vitronectin (VN) is a multifunctional macromolecule abundantly present in blood and extracellular matrix. The role of this glycoprotein in the extravasation process of circulating neutrophils remains elusive. Employing advanced in vivo/ex vivo imaging techniques in different mouse models as well as in vitro methods, we uncovered a previously unrecognized function of VN in the transition of dynamic to static intravascular interactions of neutrophils with microvascular endothelial cells. These distinct properties of VN require the heteromerization of this glycoprotein with plasminogen activator inhibitor-1 (PAI- 1) on the activated venular endothelium and subsequent interactions of this protein complex with the scavenger receptor low-density lipoprotein receptor-related protein-1 on intravascularly adhering neutrophils. This induces p38 mitogen-activated protein kinases-dependent intracellular signaling events which, in turn, regulates the proper clustering of the b2 integrin lymphocyte function associated antigen-1 on the surface of these immune cells. As a consequence of this molecular interplay, neutrophils become able to stabilize their adhesion to the microvascular endothelium and, subsequently, to extravasate to the perivascular tissue. Hence, endothelial-bound VN-PAI-1 heteromers stabilize intravascular adhesion of neutrophils by coordinating b2 integrin clustering on the surface of these immune cells, thereby effectively controlling neutrophil trafficking to inflamed tissue. Targeting this protein complex might be beneficial for the prevention and treatment of inflammatory pathologies.


Asunto(s)
Antígenos CD18 , Vitronectina , Animales , Adhesión Celular , Análisis por Conglomerados , Células Endoteliales , Ratones , Neutrófilos
6.
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
7.
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
8.
Arterioscler Thromb Vasc Biol ; 38(4): 829-842, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29371242

RESUMEN

OBJECTIVE: Ischemia-reperfusion (I/R) injury significantly contributes to organ dysfunction and failure after myocardial infarction, stroke, and transplantation. In addition to its established role in the fibrinolytic system, plasminogen activator inhibitor-1 has recently been implicated in the pathogenesis of I/R injury. The underlying mechanisms remain largely obscure. APPROACH AND RESULTS: Using different in vivo microscopy techniques as well as ex vivo analyses and in vitro assays, we identified that plasminogen activator inhibitor-1 rapidly accumulates on microvascular endothelial cells on I/R enabling this protease inhibitor to exhibit previously unrecognized functional properties by inducing an increase in the affinity of ß2 integrins in intravascularly rolling neutrophils. These events are mediated through low-density lipoprotein receptor-related protein-1 and mitogen-activated protein kinase-dependent signaling pathways that initiate intravascular adherence of these immune cells to the microvascular endothelium. Subsequent to this process, extravasating neutrophils disrupt endothelial junctions and promote the postischemic microvascular leakage. Conversely, deficiency of plasminogen activator inhibitor-1 effectively reversed leukocyte infiltration, microvascular dysfunction, and tissue injury on experimental I/R without exhibiting side effects on microvascular hemostasis. CONCLUSIONS: Our experimental data provide novel insights into the nonfibrinolytic properties of the fibrinolytic system and emphasize plasminogen activator inhibitor-1 as a promising target for the prevention and treatment of I/R injury.


Asunto(s)
Músculos Abdominales/irrigación sanguínea , Hígado/irrigación sanguínea , Microvasos/metabolismo , Infiltración Neutrófila , Neutrófilos/metabolismo , Inhibidor 1 de Activador Plasminogénico/metabolismo , Daño por Reperfusión/metabolismo , Músculos Abdominales/metabolismo , Músculos Abdominales/patología , Animales , Antígenos CD18/metabolismo , Permeabilidad Capilar , Línea Celular , Modelos Animales de Enfermedad , Humanos , Cinética , Rodamiento de Leucocito , Hígado/metabolismo , Hígado/patología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/patología , Activación Neutrófila , Neutrófilos/trasplante , Inhibidor 1 de Activador Plasminogénico/deficiencia , Inhibidor 1 de Activador Plasminogénico/genética , Conformación Proteica , Receptores de LDL/metabolismo , Daño por Reperfusión/patología , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo
9.
J Biol Chem ; 291(40): 21020-21028, 2016 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-27462075

RESUMEN

Deletion of the Habp2 gene encoding Factor VII-activating protease (FSAP) increases liver fibrosis in mice. A single nucleotide polymorphism (G534E) in HABP2 leads to lower enzymatic activity and is associated with enhanced liver fibrosis in humans. Liver fibrosis is associated with a decrease in FSAP expression but, to date, nothing is known about how this might be regulated. Primary mouse hepatocytes or the hepatocyte cell line, AML12, were treated with different factors, and expression of FSAP was determined. Of the various regulatory factors tested, only transforming growth factor-ß (TGF-ß) demonstrated a concentration- and time-dependent inhibition of FSAP expression at the mRNA and protein level. The TGF-ß-Type I receptor (ALK-5) antagonist SB431542 and Smad2 siRNA, but neither SIS3, which inhibits SMAD3, nor siRNA against Smad3 could block this effect. Various regions of the HABP2 promoter region were cloned into reporter constructs, and the promoter activity was determined. Accordingly, the promoter activity, which could phenocopy changes in Habp2 mRNA in response to TGF-ß, was found to be located in the 177-bp region upstream of the transcription start site, and this region did not contain any SMAD binding sites. Mutation analysis of the promoter and chromatin immunoprecipitation assays were performed to identify an important role for the ATF3 binding element. Thus, TGF-ß is the most likely mediator responsible for the decrease in FSAP expression in liver fibrosis.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Hepatocitos/metabolismo , Cirrosis Hepática/metabolismo , Serina Endopeptidasas/biosíntesis , Factor de Crecimiento Transformador beta/metabolismo , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Animales , Línea Celular , Hepatocitos/patología , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Ratones , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Elementos de Respuesta , Serina Endopeptidasas/genética , Proteínas Smad/genética , Proteínas Smad/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/genética
11.
Biochem Biophys Res Commun ; 491(2): 442-448, 2017 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-28712870

RESUMEN

Endoplasmic reticulum (ER) stress has been shown to play a key role during the initiation and clinical progression of the cardiovascular diseases, such as atherosclerosis. We have recently shown that expression of tissue factor pathway inhibitor (TFPI) in human monocyte-derived macrophages (MDMs) was induced by cholesterol crystals (CC). In the present study we aimed to determine the role of TFPI under ER stress conditions using human MDMs. qRT-PCR and immunohistochemistry analysis were performed to determine the presence of the ER stress marker CCAAT/enhancer binding protein homologous protein (CHOP) and TFPI in human carotid plaque material and also in human MDMs polarized into pro-inflammatory M1 or anti-inflammatory M2 populations. CHOP mRNA levels were upregulated in the plaques compared to healthy vessels, and CHOP protein was localized in the same area as TFPI in the plaques. Both CHOP and TFPI mRNA levels were upregulated after CC treatment, especially in the M2 phenotype, and the ER stress inhibitor 4-phenylbutyric acid (PBA) reversed this effect. Furthermore, CC treatment increased the levels of the pro-inflammatory cytokines TNF-α, IL-6, and IL-8, which for TNF-α and IL-8 was inhibited by PBA, and reduced the levels of the anti-inflammatory cytokine IL-10 in M2-polarized macrophages. Knockdown of TFPI prior to CC treatment exacerbated TNF-α and IL-6 levels, but reduced IL-8 and IL-10 levels. Our results show that CC induce TFPI and cytokine expression in M2-polarized macrophages through activation of the ER stress pathway and that TFPI has a protective effect against TNF-α and IL-6 mediated inflammation. These mechanisms may have implications for the pathogenesis of atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Colesterol/farmacología , Estrés del Retículo Endoplásmico/genética , Lipoproteínas/genética , Placa Aterosclerótica/genética , ARN Mensajero/genética , Aterosclerosis/inmunología , Aterosclerosis/patología , Aterosclerosis/cirugía , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/inmunología , Arterias Carótidas/patología , Arterias Carótidas/cirugía , Cristalización , Endarterectomía Carotidea , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Interleucina-10/genética , Interleucina-10/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-8/genética , Interleucina-8/inmunología , Lipoproteínas/antagonistas & inhibidores , Lipoproteínas/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Fenilbutiratos/farmacología , Placa Aterosclerótica/inmunología , Placa Aterosclerótica/patología , Placa Aterosclerótica/cirugía , Cultivo Primario de Células , ARN Mensajero/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
12.
Stroke ; 47(2): 307-16, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26732560

RESUMEN

BACKGROUND AND PURPOSE: Although a genetic contribution to ischemic stroke is well recognized, only a handful of stroke loci have been identified by large-scale genetic association studies to date. Hypothesizing that genetic effects might be stronger for early- versus late-onset stroke, we conducted a 2-stage meta-analysis of genome-wide association studies, focusing on stroke cases with an age of onset <60 years. METHODS: The discovery stage of our genome-wide association studies included 4505 cases and 21 968 controls of European, South-Asian, and African ancestry, drawn from 6 studies. In Stage 2, we selected the lead genetic variants at loci with association P<5×10(-6) and performed in silico association analyses in an independent sample of ≤1003 cases and 7745 controls. RESULTS: One stroke susceptibility locus at 10q25 reached genome-wide significance in the combined analysis of all samples from the discovery and follow-up stages (rs11196288; odds ratio =1.41; P=9.5×10(-9)). The associated locus is in an intergenic region between TCF7L2 and HABP2. In a further analysis in an independent sample, we found that 2 single nucleotide polymorphisms in high linkage disequilibrium with rs11196288 were significantly associated with total plasma factor VII-activating protease levels, a product of HABP2. CONCLUSIONS: HABP2, which encodes an extracellular serine protease involved in coagulation, fibrinolysis, and inflammatory pathways, may be a genetic susceptibility locus for early-onset stroke.


Asunto(s)
Isquemia Encefálica/genética , Serina Endopeptidasas/genética , Accidente Cerebrovascular/genética , Adulto , Edad de Inicio , Anciano , Pueblo Asiatico/genética , Población Negra/genética , Isquemia Encefálica/complicaciones , Cromosomas Humanos Par 10 , Simulación por Computador , ADN Intergénico/genética , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Polimorfismo de Nucleótido Simple , Serina Endopeptidasas/metabolismo , Accidente Cerebrovascular/etiología , Población Blanca/genética
13.
J Biol Chem ; 288(10): 7193-203, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23341458

RESUMEN

Factor VII-activating protease (FSAP) is a circulating protease involved in the pathogenesis of atherosclerosis, calcification, and fibrotic processes. To understand how FSAP controls the balance of local growth factors, we have investigated its effect on the regulation of bone morphogenetic proteins (BMPs). BMP-2 is produced as a large pro-form and secreted as a mature heparin-binding growth factor after intracellular processing by pro-protein convertases (PCs). In this study, we discovered that FSAP enhances the biological activity of mature BMP-2 as well as its pro-form, as shown by osteogenic differentiation of C2C12 myoblasts. These findings were complemented by knockdown of FSAP in hepatocytes, which revealed BMP-2 processing by endogenous FSAP. N-terminal sequencing indicated that pro-BMP-2 was cleaved by FSAP at the canonical PC cleavage site, giving rise to mature BMP-2 (Arg(282)↓Gln(283)), as well as in the N-terminal heparin binding region of mature BMP-2, generating a truncated mature BMP-2 peptide (Arg(289)↓Lys(290)). Similarly, mature BMP-2 was also cleaved to a truncated peptide within its N-terminal region (Arg(289)↓Lys(290)). Plasmin exhibited a similar activity, but it was weaker compared with FSAP. Thrombin, Factor VIIa, Factor Xa, and activated protein C were not effective. These results were further supported by the observation that the mutation of the heparin binding region of BMP-2 inhibited the processing by FSAP but not by PC. Thus, the proteolysis and activation of pro-BMP-2 and mature BMP-2 by FSAP can regulate cell differentiation and calcification in vasculature and may explain why polymorphisms in the gene encoding for FSAP are related to vascular diseases.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Precursores de Proteínas/metabolismo , Serina Endopeptidasas/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Proteína Morfogenética Ósea 2/genética , Línea Celular , Proliferación Celular , Células HEK293 , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Ratones , Mutación , Mioblastos/citología , Mioblastos/metabolismo , Fosforilación , Proproteína Convertasas/metabolismo , Precursores de Proteínas/genética , Proteolisis , Interferencia de ARN , Serina Endopeptidasas/genética , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta1/genética
14.
Blood ; 120(25): 5073-83, 2012 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-23065156

RESUMEN

Protein S is a vitamin K-dependent glycoprotein, which, besides its anticoagulant function, acts as an agonist for the tyrosine kinase receptors Tyro3, Axl, and Mer. The endothelium expresses Tyro3, Axl, and Mer and produces protein S. The interaction of protein S with endothelial cells and particularly its effects on angiogenesis have not yet been analyzed. Here we show that human protein S, at circulating concentrations, inhibited vascular endothelial growth factor (VEGF) receptor 2-dependent vascularization of Matrigel plugs in vivo and the capacity of endothelial cells to form capillary-like networks in vitro as well as VEGF-A-induced endothelial migration and proliferation. Furthermore, protein S inhibited VEGF-A-induced endothelial VEGFR2 phosphorylation and activation of mitogen-activated kinase-Erk1/2 and Akt. Protein S activated the tyrosine phosphatase SHP2, and the SHP2 inhibitor NSC 87877 reversed the observed inhibition of VEGF-A-induced endothelial proliferation. Using siRNA directed against Tyro3, Axl, and Mer, we demonstrate that protein S-mediated SHP2 activation and inhibition of VEGF-A-stimulated proliferation were mediated by Mer. Our report provides the first evidence for the existence of a protein S/Mer/SHP2 axis, which inhibits VEGFR2 signaling, regulates endothelial function, and points to a role for protein S as an endogenous angiogenesis inhibitor.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Neovascularización Fisiológica , Proteína S/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Proliferación Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Activación Enzimática , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteína S/administración & dosificación , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa c-Mer
15.
J Immunol ; 188(6): 2858-65, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22308306

RESUMEN

Severe tissue injury results in early activation of serine protease systems including the coagulation and complement cascade. In this context, little is known about factor VII-activating protease (FSAP), which is activated by substances released from damaged cells such as histones and nucleosomes. Therefore, we have measured FSAP activation in trauma patients and have identified novel FSAP substrates in human plasma. Mass spectrometry-based methods were used to identify FSAP binding proteins in plasma. Anaphylatoxin generation was measured by ELISA, Western blotting, protein sequencing, and chemotaxis assays. Plasma samples from trauma patients were analyzed for FSAP Ag and activity, nucleosomes, C5a, and C3a. Among others, we found complement components C3 and C5 in FSAP coimmunoprecipitates. C3 and C5 were cleaved by FSAP in a dose- and time-dependent manner generating functional C3a and C5a anaphylatoxins. Activation of endogenous FSAP in plasma led to increased C5a generation, but this was not the case in plasma of a homozygous carrier of Marburg I single nucleotide polymorphism with lower FSAP activity. In multiple trauma patients there was a large increase in circulating FSAP activity and nucleosomes immediately after the injury. A high correlation between FSAP activity and C5a was found. These data suggest that activation of FSAP by tissue injury triggers anaphylatoxin generation and thereby modulates the posttraumatic inflammatory response in vivo. A strong link between C5a, nucleosomes, and FSAP activity indicates that this new principle might be important in the regulation of inflammation.


Asunto(s)
Complemento C5a/inmunología , Traumatismo Múltiple/inmunología , Serina Endopeptidasas/inmunología , Adulto , Anciano , Western Blotting , Complemento C5a/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunoprecipitación , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Traumatismo Múltiple/sangre , Serina Endopeptidasas/sangre , Adulto Joven
16.
J Thromb Thrombolysis ; 37(3): 317-25, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23575879

RESUMEN

Factor VII Activating Protease (FSAP) activates factor VII (FVII) as well as pro-urokinase (uPA). Our goal was to evaluate the relation between plasma levels of FSAP and clinical instability in atrial fibrillation (AF) and possible effects of oral omega-3 fatty acids (FA) supplements. 101 patients with persistent AF were analyzed in the OMEGA-AF Study. Plasma FSAP levels were measured at baseline and after 12 weeks of treatment with omega-3 FA. The median FSAP antigen concentration, in contrast to FSAP activity, was higher in patients with persistent AF. The maintenance of SR after successful cardioversion (CV) did not lead to a normalization of FSAP concentration. Supplementation with omega-3 FA but not placebo significantly reduced elevated FSAP concentration. Furthermore, elevated FSAP levels did not indicate a significantly increased risk of recurrence of AF after electrical CV or cardiovascular clinical events during 1 year of follow-up. Plasma FSAP concentration was increased in patients with AF and may be involved in the pathogenesis of this condition. The possible effects of omega-3 FA on clinical AF potential could be linked with modulation of circulating FSAP levels.


Asunto(s)
Fibrilación Atrial/sangre , Fibrilación Atrial/dietoterapia , Suplementos Dietéticos , Ácidos Grasos Omega-3/administración & dosificación , Serina Endopeptidasas/sangre , Adulto , Anciano , Anciano de 80 o más Años , Método Doble Ciego , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad
17.
J Hepatol ; 58(1): 104-11, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22989567

RESUMEN

BACKGROUND & AIMS: Factor VII activating protease (FSAP) is a circulating serine protease produced in the liver. A single nucleotide polymorphism (G534E, Marburg I, MI-SNP) in the gene encoding FSAP (HABP2) leads to lower enzymatic activity and is associated with enhanced liver fibrosis in humans. FSAP is activated by damaged cells and its substrates include growth factors and hemostasis proteins. METHODS: We have investigated the progression of liver fibrosis in FSAP deficient mice and FSAP expression in human liver fibrosis. RESULTS: Serum FSAP concentrations declined in patients with end-stage liver disease, and hepatic FSAP expression was decreased in patients with advanced liver fibrosis and liver inflammation. Moreover, there was an inverse correlation between hepatic FSAP expression and inflammatory chemokines, chemokine receptors as well as pro-fibrotic mediators. Upon experimental bile duct ligation, FSAP(-/-) mice showed enhanced liver fibrosis in comparison to wild type mice, alongside increased expression of α-smooth muscle actin, collagen type I and fibronectin that are markers of stellate cell activation. Microarray analyses indicated that FSAP modulates inflammatory pathways. CONCLUSIONS: Lower FSAP expression is associated with enhanced liver fibrosis and inflammation in patients with chronic hepatic disorders and murine experimental liver injury. This strengthens the concept that FSAP is a "protective factor" in liver fibrosis and explains why carriers of the Marburg I SNP have more pronounced liver fibrosis.


Asunto(s)
Hepatitis/inmunología , Cirrosis Hepática/inmunología , Hígado/enzimología , Hígado/inmunología , Serina Endopeptidasas/inmunología , Adolescente , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Activación Enzimática/genética , Femenino , Hepatitis/genética , Hepatitis/metabolismo , Humanos , Cirrosis Hepática/genética , Cirrosis Hepática/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , ARN Mensajero/metabolismo , Serina Endopeptidasas/sangre , Serina Endopeptidasas/genética , Transcriptoma , Adulto Joven
18.
Arterioscler Thromb Vasc Biol ; 32(2): 427-33, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22116096

RESUMEN

OBJECTIVE: Factor VII-activating protease (FSAP) activates both factor VII and pro-urokinase and inhibits platelet-derived growth factor-BB, thus regulating hemostasis- and remodeling-associated processes in the vasculature. A genetic variant of FSAP (Marburg I polymorphism) results in low enzymatic activity and is associated with an enhanced risk of carotid stenosis and stroke. We postulate that there are additional substrates for FSAP that will help to explain its role in vascular biology and have searched for such a substrate. METHODS AND RESULTS: Using screening procedures to determine the influence of FSAP on various hemostasis-related processes on endothelial cells, we discovered that FSAP inhibited tissue factor pathway inhibitor (TFPI), a major anticoagulant secreted by these cells. Proteolytic degradation of TFPI by FSAP could also be demonstrated by Western blotting, and the exact cleavage sites were determined by N-terminal sequencing. The Marburg I variant of FSAP had a diminished ability to inhibit TFPI. A monoclonal antibody to FSAP that specifically inhibited FSAP binding to TFPI reversed the inhibitory effect of FSAP on TFPI. CONCLUSIONS: The identification of TFPI as a sensitive substrate for FSAP increases our understanding of its role in regulating hemostasis and proliferative remodeling events in the vasculature.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Lipoproteínas/antagonistas & inhibidores , Proteolisis/efectos de los fármacos , Serina Endopeptidasas/farmacología , Anticuerpos Monoclonales/farmacología , Proliferación Celular , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Hemostasis/fisiología , Humanos , Lipoproteínas/efectos de los fármacos , Serina Endopeptidasas/efectos de los fármacos , Serina Endopeptidasas/inmunología
19.
Circulation ; 124(17): 1848-59, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-21969013

RESUMEN

BACKGROUND: Urokinase-type plasminogen activator (uPA) has recently been implicated in the pathogenesis of ischemia-reperfusion (I/R) injury. The underlying mechanisms remain largely unclear. METHODS AND RESULTS: Using in vivo microscopy on the mouse cremaster muscle, I/R-elicited firm adherence and transmigration of neutrophils were found to be significantly diminished in uPA-deficient mice and in mice treated with the uPA inhibitor WX-340, but not in uPA receptor (uPAR)-deficient mice. Interestingly, postischemic leukocyte responses were significantly reduced on blockade of the integrin CD11b/Mac-1, which also serves as uPAR receptor. Using a cell transfer technique, postischemic adherence and transmigration of wild-type leukocytes were significantly decreased in uPA-deficient animals, whereas uPA-deficient leukocytes exhibited a selectively reduced transmigration in wild-type animals. On I/R or stimulation with recombinant uPA, >90% of firmly adherent leukocytes colocalized with CD31-immunoreactive endothelial junctions as detected by in vivo fluorescence microscopy. In a model of hepatic I/R, treatment with WX-340 significantly attenuated postischemic neutrophil infiltration and tissue injury. CONCLUSIONS: Our data suggest that endothelial uPA promotes intravascular adherence, whereas leukocyte uPA facilitates the subsequent paracellular transmigration of neutrophils during I/R. This process is regulated via CD11b/Mac-1, and does not require uPAR. Pharmacological blockade of uPA interferes with these events and effectively attenuates postischemic tissue injury.


Asunto(s)
Antígeno de Macrófago-1/fisiología , Neutrófilos/citología , Neutrófilos/fisiología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/fisiología , Migración Transendotelial y Transepitelial/fisiología , Activador de Plasminógeno de Tipo Uroquinasa/fisiología , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/fisiología , Distribución Aleatoria
20.
J Exp Med ; 203(13): 2801-7, 2006 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-17145954

RESUMEN

The G534E polymorphism (Marburg I [MI]) of factor VII-activating protease (FSAP) is associated with carotid stenosis and cardiovascular disease. We have previously demonstrated that FSAP is present in atherosclerotic plaques and it is a potent inhibitor of vascular smooth muscle proliferation and migration in vitro. The effect of wild-type (WT)- and MI-FSAP on neointima formation in the mouse femoral artery after wire-induced injury was investigated. Local application of WT-FSAP led to a 70% reduction in the neointima formation, and this effect was dependent on the protease activity of FSAP. MI-FSAP did not inhibit neointima formation in vivo. This is due to a reduced proteolytic activity of MI-FSAP, compared to WT-FSAP, toward platelet-derived growth factor BB, a key mediator of neointima development. The inability of MI-FSAP to inhibit vascular smooth muscle accumulation explains the observed linkage between the MI-polymorphism and increased cardiovascular risk. Hence, FSAP has a protective function in the vasculature, and analysis of MI polymorphism is likely to be clinically relevant in restenosis.


Asunto(s)
Reestenosis Coronaria/genética , Polimorfismo Genético , Serina Endopeptidasas/genética , Túnica Íntima/metabolismo , Actinas/metabolismo , Clorometilcetonas de Aminoácidos/farmacología , Animales , Becaplermina , Catálisis , Proliferación Celular/efectos de los fármacos , Reestenosis Coronaria/prevención & control , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Expresión Génica , Heparina/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Fosforilación , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Unión Proteica , Proteínas Proto-Oncogénicas c-sis , Serina Endopeptidasas/metabolismo , Serina Endopeptidasas/farmacología , Inhibidores de Serina Proteinasa/farmacología , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Factor de von Willebrand/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA