Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 790
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Blood ; 143(10): 845-857, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38096370

RESUMEN

ABSTRACT: Protease activated receptors (PARs) are cleaved by coagulation proteases and thereby connect hemostasis with innate immune responses. Signaling of the tissue factor (TF) complex with factor VIIa (FVIIa) via PAR2 stimulates extracellular signal-regulated kinase (ERK) activation and cancer cell migration, but functions of cell autonomous TF-FVIIa signaling in immune cells are unknown. Here, we show that myeloid cell expression of FVII but not of FX is crucial for inflammatory cell recruitment to the alveolar space after challenge with the double-stranded viral RNA mimic polyinosinic:polycytidylic acid [Poly(I:C)]. In line with these data, genetically modified mice completely resistant to PAR2 cleavage but not FXa-resistant PAR2-mutant mice are protected from lung inflammation. Poly(I:C)-stimulated migration of monocytes/macrophages is dependent on ERK activation and mitochondrial antiviral signaling (MAVS) but independent of toll-like receptor 3 (TLR3). Monocyte/macrophage-synthesized FVIIa cleaving PAR2 is required for integrin αMß2-dependent migration on fibrinogen but not for integrin ß1-dependent migration on fibronectin. To further dissect the downstream signaling pathway, we generated PAR2S365/T368A-mutant mice deficient in ß-arrestin recruitment and ERK scaffolding. This mutation reduces cytosolic, but not nuclear ERK phosphorylation by Poly(I:C) stimulation, and prevents macrophage migration on fibrinogen but not fibronectin after stimulation with Poly(I:C) or CpG-B, a single-stranded DNA TLR9 agonist. In addition, PAR2S365/T368A-mutant mice display markedly reduced immune cell recruitment to the alveolar space after Poly(I:C) challenge. These results identify TF-FVIIa-PAR2-ß-arrestin-biased signaling as a driver for lung infiltration in response to viral nucleic acids and suggest potential therapeutic interventions specifically targeting TF-VIIa signaling in thrombo-inflammation.


Asunto(s)
Factor VIIa , Monocitos , Animales , Ratones , Factor VIIa/metabolismo , Monocitos/metabolismo , Tromboplastina/metabolismo , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Transducción de Señal/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrinógeno/metabolismo , beta-Arrestinas/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 44(4): 843-865, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38385286

RESUMEN

BACKGROUND: Accumulating evidence implicates the activation of G-protein-coupled PARs (protease-activated receptors) by coagulation proteases in the regulation of innate immune responses. METHODS: Using mouse models with genetic alterations of the PAR2 signaling platform, we have explored contributions of PAR2 signaling to infection with coxsackievirus B3, a single-stranded RNA virus provoking multiorgan tissue damage, including the heart. RESULTS: We show that PAR2 activation sustains correlates of severe morbidity-hemodynamic compromise, aggravated hypothermia, and hypoglycemia-despite intact control of the virus. Following acute viral liver injury, canonical PAR2 signaling impairs the restoration process associated with exaggerated type I IFN (interferon) signatures in response to viral RNA recognition. Metabolic profiling in combination with proteomics of liver tissue shows PAR2-dependent reprogramming of liver metabolism, increased lipid droplet storage, and gluconeogenesis. PAR2-sustained hypodynamic compromise, reprograming of liver metabolism, as well as imbalanced IFN responses are prevented in ß-arrestin coupling-deficient PAR2 C-terminal phosphorylation mutant mice. Thus, wiring between upstream proteases and immune-metabolic responses results from biased PAR2 signaling mediated by intracellular recruitment of ß-arrestin. Importantly, blockade of the TF (tissue factor)-FVIIa (coagulation factor VIIa) complex capable of PAR2 proteolysis with the NAPc2 (nematode anticoagulant protein c2) mitigated virus-triggered pathology, recapitulating effects seen in protease cleavage-resistant PAR2 mice. CONCLUSIONS: These data provide insights into a TF-FVIIa signaling axis through PAR2-ß-arrestin coupling that is a regulator of inflammation-triggered tissue repair and hemodynamic compromise in coxsackievirus B3 infection and can potentially be targeted with selective coagulation inhibitors.


Asunto(s)
Insuficiencia Multiorgánica , Tromboplastina , Animales , Ratones , Tromboplastina/metabolismo , beta-Arrestinas/metabolismo , Receptor PAR-2/genética , Factor VIIa/metabolismo , Endopeptidasas/metabolismo
3.
Blood ; 139(1): 118-133, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34469511

RESUMEN

Coagulation protease, factor VIIa (FVIIa), binds to endothelial cell protein C receptor (EPCR) and induces anti-inflammatory and endothelial barrier protective responses via protease-activated receptor-1 (PAR1)-mediated, biased signaling. Our recent studies had shown that the FVIIa-EPCR-PAR1 axis induces the release of extracellular vesicles (EVs) from endothelial cells. In the present study, we investigated the mechanism of FVIIa release of endothelial EVs (EEVs) and the contribution of FVIIa-released EEVs to anti-inflammatory and vascular barrier protective effects, in both in vitro and in vivo models. Multiple signaling pathways regulated FVIIa release of EVs from endothelial cells, but the ROCK-dependent pathway appeared to be a major mechanism. FVIIa-released EEVs were enriched with anti-inflammatory microRNAs (miRs), mostly miR10a. FVIIa-released EEVs were taken up readily by monocytes/macrophages and endothelial cells. The uptake of FVIIa-released EEVs by monocytes conferred anti-inflammatory phenotype to monocytes, whereas EEV uptake by endothelial cells resulted in barrier protection. In additional experiments, EEV-mediated delivery of miR10a to monocytes downregulated the expression of TAK1 and activation of the NF-κB-mediated inflammatory pathway. In in vivo experiments, administration of FVIIa-released EEVs to wild-type mice attenuated LPS-induced increased inflammatory cytokines in plasma and vascular leakage into vital tissues. The incorporation of anti-miR10a into FVIIa-released EEVs diminished the ability of FVIIa-released EEVs to confer cytoprotective effects. Administration of the ROCK inhibitor Y27632, which significantly inhibits FVIIa release of EEVs into the circulation, to mice attenuated the cytoprotective effects of FVIIa. Overall, our study revealed novel insights into how FVIIa induces cytoprotective effects and communicates with various cell types.


Asunto(s)
Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Factor VIIa/metabolismo , Inflamación/metabolismo , MicroARNs/metabolismo , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones Endogámicos C57BL , Monocitos/metabolismo , Células THP-1
4.
J Obstet Gynaecol Res ; 49(7): 1677-1683, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37186495

RESUMEN

AIM: Tissue factor (TF), the primary initiator of the extrinsic coagulation pathway, contributes to the generation of a hypercoagulable and prothrombotic state in cancer patients. TF pathway inhibitor (TFPI) is a major inhibitor of TF-mediated coagulation pathway. The two proteins, TFPI1 and TFPI2, are encoded by separate genes. Indeed, various cancer patients with venous thromboembolism (VTE) had significantly lower TFPI1 levels than those without VTE. In contrast, serum TFPI2 level was found to increase in ovarian cancer patients with VTE. It remains unclear why TFPI2, unlike TFPI1, is elevated in ovarian cancer patients with VTE. The aim of this review is to explore the pathophysiological role of TFPI2 on the coagulation and fibrinolysis system. METHODS: A literature search was performed from inception to April 30, 2022 in the PubMed and Google Scholar databases. RESULTS: TFPI1 and TFPI2 are homologs with different protease inhibitory activities in the coagulation and fibrinolysis system. TFPI1 inhibits TF/factor VIIa (FVIIa) catalyzed factor X (FX) activation. On the other hand, TFPI2 is unlikely to affect TF-initiated thrombin generation, but it has strong inhibitory activity against plasmin. Plasmin is involved in fibrin degradation, clot lysis, and inactivation of several coagulation factors (such as FV, FVIII, FIX, and FX). TFPI2 may increase the risk of VTE by inhibiting plasmin-dependent fibrinolysis. CONCLUSION: TFPI1 and TFPI2 may have different key functions in regulating the coagulation and fibrinolytic systems.


Asunto(s)
Fibrinólisis , Glicoproteínas , Tromboembolia Venosa , Humanos , Coagulación Sanguínea , Factor VIIa/metabolismo , Fibrinolisina , Tromboplastina/metabolismo , Glicoproteínas/metabolismo
5.
Blood ; 136(21): 2469-2472, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-32604409

RESUMEN

Recent reports indicate that suspended skeletal and cardiac myosin, such as might be released during injury, can act as procoagulants by providing membrane-like support for factors Xa and Va in the prothrombinase complex. Further, skeletal myosin provides membrane-like support for activated protein C. This raises the question of whether purified muscle myosins retain procoagulant phospholipid through purification. We found that lactadherin, a phosphatidyl-l-serine-binding protein, blocked >99% of prothrombinase activity supported by rabbit skeletal and by bovine cardiac myosin. Similarly, annexin A5 and phospholipase A2 blocked >95% of myosin-supported activity, confirming that contaminating phospholipid is required to support myosin-related prothrombinase activity. We asked whether contaminating phospholipid in myosin preparations may also contain tissue factor (TF). Skeletal myosin supported factor VIIa cleavage of factor X equivalent to contamination by ∼1:100 000 TF/myosin, whereas cardiac myosin had TF-like activity >10-fold higher. TF pathway inhibitor inhibited the TF-like activity similar to control TF. These results indicate that purified skeletal muscle and cardiac myosins support the prothrombinase complex indirectly through contaminating phospholipid and also support factor X activation through TF-like activity. Our findings suggest a previously unstudied affinity of skeletal and cardiac myosin for phospholipid membranes.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Factor V/efectos de los fármacos , Factor Xa/efectos de los fármacos , Músculo Esquelético/química , Miocardio/química , Miosinas/farmacología , Fosfolípidos/farmacología , Animales , Antígenos de Superficie/farmacología , Miosinas Cardíacas/aislamiento & purificación , Miosinas Cardíacas/metabolismo , Miosinas Cardíacas/farmacología , Bovinos , Contaminación de Medicamentos , Factor VIIa/metabolismo , Factor Xa/metabolismo , Humanos , Lipoproteínas/farmacología , Proteínas de la Leche/farmacología , Miosinas/aislamiento & purificación , Miosinas/metabolismo , Fosfolipasas A2/farmacología , Conejos , Tromboplastina/farmacología
6.
J Biol Chem ; 295(2): 517-528, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31801825

RESUMEN

Two decades of research have uncovered the mechanism by which the complex of tissue factor (TF) and the plasma serine protease factor VIIa (FVIIa) mediates the initiation of blood coagulation. Membrane-anchored TF directly interacts with substrates and induces allosteric effects in the protease domain of FVIIa. These properties are also recapitulated by the soluble ectodomain of TF (sTF). At least two interdependent allosteric activation pathways originate at the FVIIa:sTF interface are proposed to enhance FVIIa activity upon sTF binding. Here, we sought to engineer an sTF-independent FVIIa variant by stabilizing both proposed pathways, with one pathway terminating at segment 215-217 in the activation domain and the other pathway terminating at the N terminus insertion site. To stabilize segment 215-217, we replaced the flexible 170 loop of FVIIa with the more rigid 170 loop from trypsin and combined it with an L163V substitution (FVIIa-VYT). The FVIIa-VYT variant exhibited 60-fold higher amidolytic activity than FVIIa, and displayed similar FX activation and antithrombin inhibition kinetics to the FVIIa.sTF complex. The sTF-independent activity of FVIIa-VYT was partly mediated by an increase in the N terminus insertion and, as shown by X-ray crystallography, partly by Tyr-172 inserting into a cavity in the activation domain stabilizing the S1 substrate-binding pocket. The combination with L163V likely drove additional changes in a delicate hydrogen-bonding network that further stabilized S1-S3 sites. In summary, we report the first FVIIa variant that is catalytically independent of sTF and provide evidence supporting the existence of two TF-mediated allosteric activation pathways.


Asunto(s)
Coagulación Sanguínea , Factor VIIa/metabolismo , Ingeniería de Proteínas , Tromboplastina/metabolismo , Regulación Alostérica , Secuencia de Aminoácidos , Cristalografía por Rayos X , Factor VIIa/química , Factor VIIa/genética , Humanos , Modelos Moleculares , Mutagénesis , Desplegamiento Proteico , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
7.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-35008781

RESUMEN

In hemostasis and thrombosis, the complex process of thrombus formation involves different molecular pathways of platelet and coagulation activation. These pathways are considered as operating together at the same time, but this has not been investigated. The objective of our study was to elucidate the time-dependency of key pathways of thrombus and clot formation, initiated by collagen and tissue factor surfaces, where coagulation is triggered via the extrinsic route. Therefore, we adapted a microfluidics whole-blood assay with the Maastricht flow chamber to acutely block molecular pathways by pharmacological intervention at desired time points. Application of the technique revealed crucial roles of glycoprotein VI (GPVI)-induced platelet signaling via Syk kinase as well as factor VIIa-induced thrombin generation, which were confined to the first minutes of thrombus buildup. A novel anti-GPVI Fab EMF-1 was used for this purpose. In addition, platelet activation with the protease-activating receptors 1/4 (PAR1/4) and integrin αIIbß3 appeared to be prolongedly active and extended to later stages of thrombus and clot formation. This work thereby revealed a more persistent contribution of thrombin receptor-induced platelet activation than of collagen receptor-induced platelet activation to the thrombotic process.


Asunto(s)
Coagulación Sanguínea , Plaquetas/metabolismo , Colágeno/farmacología , Tromboplastina/farmacología , Trombosis/patología , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Factor VIIa/metabolismo , Fibrina/metabolismo , Humanos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Receptores Proteinasa-Activados/metabolismo , Transducción de Señal , Quinasa Syk/metabolismo , Factores de Tiempo
8.
Apoptosis ; 25(7-8): 519-534, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32458278

RESUMEN

The tissue factor/coagulation factor VIIa (TF/FVIIa) complex induces transactivation of the IGF-1 receptor (IGF-1R) in a number of different cell types. The mechanism is largely unknown. The transactivation leads to protection from apoptosis and nuclear translocation of the IGF-1R. The aim of this study was to clarify the signaling pathway between TF and IGF-1R after FVIIa treatment with PC3 and DU145 prostate or MDA-MB-231 breast cancer cells as model systems. Protein interactions, levels, and phosphorylations were assessed by proximity ligation assay or flow cytometry in intact cells and by western blot on cell lysates. The transactivation of the IGF-1R was found dependent on TF/FVIIa-induced activation of ß1-integrins. A series of experiments led to the conclusion that the caveolae protein caveolin-1 prevented IGF-1R activation in resting cells via its scaffolding domain. TF/FVIIa/ß1-integrins terminated this inhibition by activation of Src family kinases and subsequent phosphorylation of caveolin-1 on tyrosine 14. This phosphorylation was not seen after treatment with PAR1 or PAR2 agonists. Consequently, the protective effect of FVIIa against apoptosis induced by the death receptor agonist TRAIL and the de novo synthesis of cyclin D1 induced by nuclear IGF-1R accumulation were both significantly reduced by down-regulation of ß1-integrins or overexpression of the caveolin-1 scaffolding domain. In conclusion, we present a plausible mechanism for the interplay between TF and IGF-1R involving FVIIa, ß1-integrins, Src family proteins, and caveolin-1. Our results increase the knowledge of diseases associated with TF and IGF-1R overexpression in general but specifically of TF-mediated signaling with focus on cell survival.


Asunto(s)
Caveolina 1/genética , Factor VIIa/farmacología , Regulación Neoplásica de la Expresión Génica , Integrina beta1/genética , Receptor IGF Tipo 1/genética , Tromboplastina/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Caveolas/efectos de los fármacos , Caveolas/metabolismo , Caveolina 1/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Factor VIIa/genética , Factor VIIa/metabolismo , Humanos , Integrina beta1/metabolismo , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo , Receptor PAR-2/genética , Receptor PAR-2/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Tromboplastina/genética , Tromboplastina/metabolismo , Activación Transcripcional/efectos de los fármacos , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
9.
Blood ; 131(6): 674-685, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29246902

RESUMEN

The tissue factor (TF) pathway serves both hemostasis and cell signaling, but how cells control these divergent functions of TF remains incompletely understood. TF is the receptor and scaffold of coagulation proteases cleaving protease-activated receptor 2 (PAR2) that plays pivotal roles in angiogenesis and tumor development. Here we demonstrate that coagulation factor VIIa (FVIIa) elicits TF cytoplasmic domain-dependent proangiogenic cell signaling independent of the alternative PAR2 activator matriptase. We identify a Lys-Gly-Glu (KGE) integrin-binding motif in the FVIIa protease domain that is required for association of the TF-FVIIa complex with the active conformer of integrin ß1. A point mutation in this motif markedly reduces TF-FVIIa association with integrins, attenuates integrin translocation into early endosomes, and reduces delayed mitogen-activated protein kinase phosphorylation required for the induction of proangiogenic cytokines. Pharmacologic or genetic blockade of the small GTPase ADP-ribosylation factor 6 (arf6) that regulates integrin trafficking increases availability of TF-FVIIa with procoagulant activity on the cell surface, while inhibiting TF-FVIIa signaling that leads to proangiogenic cytokine expression and tumor cell migration. These experiments delineate the structural basis for the crosstalk of the TF-FVIIa complex with integrin trafficking and suggest a crucial role for endosomal PAR2 signaling in pathways of tissue repair and tumor biology.


Asunto(s)
Factor VIIa/química , Factor VIIa/metabolismo , Integrina beta1/metabolismo , Dominios y Motivos de Interacción de Proteínas , Receptor PAR-2/metabolismo , Factor 6 de Ribosilación del ADP , Animales , Sitios de Unión/genética , Células Cultivadas , Factor VIIa/genética , Humanos , Integrina beta1/química , Ratones , Células 3T3 NIH , Neovascularización Fisiológica/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Mapas de Interacción de Proteínas , Receptor PAR-2/genética , Transducción de Señal/genética , Tromboplastina/química , Tromboplastina/metabolismo
10.
Blood ; 131(21): 2379-2392, 2018 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-29669778

RESUMEN

Recent studies show that endothelial cell protein C receptor (EPCR) interacts with diverse ligands, in addition to its known ligands protein C and activated protein C (APC). We showed in earlier studies that procoagulant clotting factor VIIa (FVIIa) binds EPCR and downregulates EPCR-mediated anticoagulation and induces an endothelial barrier protective effect. Here, we investigated the effect of FVIIa's interaction with EPCR on endothelial cell inflammation and lipopolysaccharide (LPS)-induced inflammatory responses in vivo. Treatment of endothelial cells with FVIIa suppressed tumor necrosis factor α (TNF-α)- and LPS-induced expression of cellular adhesion molecules and adherence of monocytes to endothelial cells. Inhibition of EPCR or protease-activated receptor 1 (PAR1) by either specific antibodies or small interfering RNA abolished the FVIIa-induced suppression of TNF-α- and LPS-induced expression of cellular adhesion molecules and interleukin-6. ß-Arrestin-1 silencing blocked the FVIIa-induced anti-inflammatory effect in endothelial cells. In vivo studies showed that FVIIa treatment markedly suppressed LPS-induced inflammatory cytokines and infiltration of innate immune cells into the lung in wild-type and EPCR-overexpressing mice, but not in EPCR-deficient mice. Mechanistic studies revealed that FVIIa treatment inhibited TNF-α-induced ERK1/2, p38 MAPK, JNK, NF-κB, and C-Jun activation indicating that FVIIa-mediated signaling blocks an upstream signaling event in TNFα-induced signaling cascade. FVIIa treatment impaired the recruitment of TNF-receptor-associated factor 2 into the TNF receptor 1 signaling complex. Overall, our present data provide convincing evidence that FVIIa binding to EPCR elicits anti-inflammatory signaling via a PAR1- and ß-arrestin-1 dependent pathway. The present study suggests new therapeutic potentials for FVIIa, which is currently in clinical use for treating bleeding disorders.


Asunto(s)
Receptor de Proteína C Endotelial/metabolismo , Factor VIIa/metabolismo , Inflamación/metabolismo , Receptor PAR-1/metabolismo , Transducción de Señal , Animales , Biomarcadores , Receptor de Proteína C Endotelial/genética , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/genética , Mediadores de Inflamación/metabolismo , Lipopolisacáridos/efectos adversos , Lipopolisacáridos/inmunología , Ratones , Receptor PAR-1/genética , Factor de Necrosis Tumoral alfa/metabolismo , beta-Arrestinas/metabolismo
11.
Arterioscler Thromb Vasc Biol ; 39(10): 2038-2048, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31412737

RESUMEN

OBJECTIVE: Regulation of TF (tissue factor):FVIIa (coagulation factor VIIa) complex procoagulant activity is especially critical in tissues where plasma can contact TF-expressing cells. One example is the liver, where hepatocytes are routinely exposed to plasma because of the fenestrated sinusoidal endothelium. Although liver-associated TF contributes to coagulation, the mechanisms controlling the TF:FVIIa complex activity in this tissue are not known. Approach and Results: Common bile duct ligation in mice triggered rapid hepatocyte TF-dependent intrahepatic coagulation coincident with increased plasma bile acids, which occurred at a time before observable liver damage. Similarly, plasma TAT (thrombin-antithrombin) levels increased in cholestatic patients without concurrent hepatocellular injury. Pathologically relevant concentrations of the bile acid glycochenodeoxycholic acid rapidly increased hepatocyte TF-dependent procoagulant activity in vitro, independent of de novo TF synthesis and necrotic or apoptotic cell death. Glycochenodeoxycholic acid increased hepatocyte TF activity even in the presence of the phosphatidylserine-blocking protein lactadherin. Interestingly, glycochenodeoxycholic acid and taurochenodeoxycholic acid increased the procoagulant activity of the TF:FVIIa complex relipidated in unilamellar phosphatidylcholine vesicles, which was linked to an apparent decrease in the Km for FX (coagulation factor X). Notably, the zwitterionic detergent 3-[(3-cholamidopropyl)dimethylammonio]-1-propanesulfonate, a bile acid structural analog, did not increase relipidated TF:FVIIa activity. Bile acids directly enhanced factor X activation by recombinant soluble TF:FVIIa complex but had no effect on FVIIa alone. CONCLUSIONS: The results indicate that bile acids directly accelerate TF:FVIIa-driven coagulation reactions, suggesting a novel mechanism whereby elevation in a physiological mediator can directly increase TF:FVIIa procoagulant activity.


Asunto(s)
Conductos Biliares/cirugía , Colestasis Intrahepática/metabolismo , Colestasis Intrahepática/fisiopatología , Factor VIIa/metabolismo , Factor X/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Coagulación Sanguínea/fisiología , Trastornos de la Coagulación Sanguínea/fisiopatología , Pruebas de Coagulación Sanguínea , Células Cultivadas , Modelos Animales de Enfermedad , Hepatocitos/metabolismo , Humanos , Cinética , Ligadura/métodos , Ratones , Ratones Endogámicos C57BL , Fosfatidilserinas/metabolismo , Distribución Aleatoria
12.
Biochem J ; 476(19): 2909-2926, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31537632

RESUMEN

The mechanism of generation of factor VIIa, considered the initiating protease in the tissue factor-initiated extrinsic limb of blood coagulation, is obscure. Decreased levels of plasma VIIa in individuals with congenital factor IX deficiency suggest that generation of VIIa is dependent on an activation product of factor IX. Factor VIIa activates IX to IXa by a two-step removal of the activation peptide with cleavages occurring after R191 and R226. Factor IXaα, however, is IX cleaved only after R226, and not after R191. We tested the hypothesis that IXaα activates VII with mutant IX that could be cleaved only at R226 and thus generate only IXaα upon activation. Factor IXaα demonstrated 1.6% the coagulant activity of IXa in a contact activation-based assay of the intrinsic activation limb and was less efficient than IXa at activating factor X in the presence of factor VIIIa. However, IXaα and IXa had indistinguishable amidolytic activity, and, strikingly, both catalyzed the cleavage required to convert VII to VIIa with indistinguishable kinetic parameters that were augmented by phospholipids, but not by factor VIIIa or tissue factor. We propose that IXa and IXaα participate in a pathway of reciprocal activation of VII and IX that does not require a protein cofactor. Since both VIIa and activated IX are equally plausible as the initiating protease for the extrinsic limb of blood coagulation, it might be appropriate to illustrate this key step of hemostasis as currently being unknown.


Asunto(s)
Coagulación Sanguínea/fisiología , Factor IX/metabolismo , Factor VII/metabolismo , Factor VIIa/metabolismo , Factor X/metabolismo , Células HEK293 , Humanos , Cinética , Proteínas Recombinantes/metabolismo
13.
Biophys J ; 116(10): 1823-1835, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31003762

RESUMEN

A critical step in injury-induced initiation of blood coagulation is the formation of the complex between the trypsin-like protease coagulation factor VIIa (FVIIa) and its cofactor tissue factor (TF), which converts FVIIa from an intrinsically poor enzyme to an active protease capable of activating zymogens of downstream coagulation proteases. Unlike its constitutively active ancestor trypsin, FVIIa is allosterically activated (by TF). Here, ensemble refinement of crystallographic structures, which uses multiple copies of the entire structure as a means of representing structural flexibility, is applied to explore the impacts of inhibitor binding to trypsin and FVIIa, as well as cofactor binding to FVIIa. To assess the conformational flexibility and its role in allosteric pathways in these proteases, main-chain hydrogen bond networks are analyzed by calculating the hydrogen-bond propensity. Mapping pairwise propensity differences between relevant structures shows that binding of the inhibitor benzamidine to trypsin has a minor influence on the protease flexibility. For FVIIa, in contrast, the protease domain is "locked" into the catalytically competent trypsin-like configuration upon benzamidine binding as indicated by the stabilization of key structural features: the nonprime binding cleft and the oxyanion hole are stabilized, and the effect propagates from the active site region to the calcium-binding site and to the vicinity of the disulphide bridge connecting with the light chain. TF binding to FVIIa furthermore results in stabilization of the 170 loop, which in turn propagates an allosteric signal from the TF-binding region to the active site. Analyses of disulphide bridge energy and flexibility reflect the striking stability difference between the unregulated enzyme and the allosterically activated form after inhibitor or cofactor binding. The ensemble refinement analyses show directly, for the first time to our knowledge, whole-domain structural footprints of TF-induced allosteric networks present in x-ray crystallographic structures of FVIIa, which previously only have been hypothesized or indirectly inferred.


Asunto(s)
Factor VIIa/química , Factor VIIa/metabolismo , Regulación Alostérica , Apoenzimas/química , Apoenzimas/metabolismo , Benzamidinas/farmacología , Cristalografía por Rayos X , Disulfuros/química , Activación Enzimática/efectos de los fármacos , Modelos Moleculares , Dominios Proteicos , Pliegue de Proteína , Tripsina/química , Tripsina/metabolismo , Tripsinógeno/metabolismo
14.
Blood ; 130(14): 1661-1670, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28729433

RESUMEN

Safe and effective antithrombotic therapy requires understanding of mechanisms that contribute to pathological thrombosis but have a lesser impact on hemostasis. We found that the extrinsic tissue factor (TF) coagulation initiation complex can selectively activate the antihemophilic cofactor, FVIII, triggering the hemostatic intrinsic coagulation pathway independently of thrombin feedback loops. In a mouse model with a relatively mild thrombogenic lesion, TF-dependent FVIII activation sets the threshold for thrombus formation through contact phase-generated FIXa. In vitro, FXa stably associated with TF-FVIIa activates FVIII, but not FV. Moreover, nascent FXa product of TF-FVIIa can transiently escape the slow kinetics of Kunitz-type inhibition by TF pathway inhibitor and preferentially activates FVIII over FV. Thus, TF synergistically primes FIXa-dependent thrombin generation independently of cofactor activation by thrombin. Accordingly, FVIIa mutants deficient in direct TF-dependent thrombin generation, but preserving FVIIIa generation by nascent FXa, can support intrinsic pathway coagulation. In ex vivo flowing blood, a TF-FVIIa mutant complex with impaired free FXa generation but activating both FVIII and FIX supports efficient FVIII-dependent thrombus formation. Thus, a previously unrecognized TF-initiated pathway directly yielding FVIIIa-FIXa intrinsic tenase complex may be prohemostatic before further coagulation amplification by thrombin-dependent feedback loops enhances the risk of thrombosis.


Asunto(s)
Coagulación Sanguínea , Factor VIII/metabolismo , Factor VIIa/metabolismo , Factor Xa/metabolismo , Tromboplastina/metabolismo , Factor VIIIa/metabolismo , Humanos , Trombina/metabolismo
15.
Oncology ; 96(4): 217-222, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30844808

RESUMEN

OBJECTIVES: Tissue factor overexpression is associated with tumor progression, venous thromboembolism, and worsened survival in patients with cancer. Tissue factor and activated factor VII (FVIIa) complex may contribute to tumor invasiveness by promoting cell migration and angiogenesis. The study objective was to evaluate safety, pharmacokinetics, and efficacy of PCI-27483, a selective FVIIa inhibitor. METHODS: This was an open-label, multicenter phase 2 trial of patients with advanced pancreatic cancer. Part A of the study was an intrapatient dose escalation lead-in portion in patients concurrently receiving gemcitabine, and in part B, patients were randomized 1: 1 to the recommended phase 2 dose combination PCI-27483-gemcitabine versus gemcitabine alone. RESULTS: Target international normalized ratio (between 2.0-3.0) was achieved following PCI-27483 treatment. Overall safety of PCI-27483-gemcitabine (n = 26) was similar to gemcitabine alone (n = 16), with a higher incidence of mostly low-grade bleeding events (65% vs. 19%). Progression-free survival (PFS) and overall survival (OS) were not significantly different between patients treated with PCI-27483-gemcitabine (PFS: 3.7 months, OS: 5.7 months) and those treated with gemcitabine alone (PFS: 1.9 months, OS: 5.6 months). CONCLUSIONS: Targeted inhibition of the coagulation cascade was achieved by administering PCI-27483. PCI-27483-gemcitabine was well tolerated, but superiority to single agent gemcitabine was not demonstrated.


Asunto(s)
Anticoagulantes/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Ácido Aspártico/análogos & derivados , Bencimidazoles/administración & dosificación , Coagulación Sanguínea/efectos de los fármacos , Carcinoma Ductal Pancreático/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Factor VIIa/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Anticoagulantes/efectos adversos , Anticoagulantes/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Ácido Aspártico/administración & dosificación , Ácido Aspártico/efectos adversos , Ácido Aspártico/farmacocinética , Bencimidazoles/efectos adversos , Bencimidazoles/farmacocinética , Carcinoma Ductal Pancreático/sangre , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/secundario , Desoxicitidina/administración & dosificación , Desoxicitidina/efectos adversos , Monitoreo de Drogas/métodos , Factor VIIa/metabolismo , Femenino , Hemorragia/inducido químicamente , Humanos , Relación Normalizada Internacional , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/sangre , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Supervivencia sin Progresión , Factores de Tiempo , Gemcitabina
16.
Arterioscler Thromb Vasc Biol ; 38(4): 709-725, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29437578

RESUMEN

Tissue factor (TF) is the high-affinity receptor and cofactor for factor (F)VII/VIIa. The TF-FVIIa complex is the primary initiator of blood coagulation and plays an essential role in hemostasis. TF is expressed on perivascular cells and epithelial cells at organ and body surfaces where it forms a hemostatic barrier. TF also provides additional hemostatic protection to vital organs, such as the brain, lung, and heart. Under pathological conditions, TF can trigger both arterial and venous thrombosis. For instance, atherosclerotic plaques contain high levels of TF on macrophage foam cells and microvesicles that drives thrombus formation after plaque rupture. In sepsis, inducible TF expression on monocytes leads to disseminated intravascular coagulation. In cancer patients, tumors release TF-positive microvesicles into the circulation that may contribute to venous thrombosis. TF also has nonhemostatic roles. For instance, TF-dependent activation of the coagulation cascade generates coagulation proteases, such as FVIIa, FXa, and thrombin, which induce signaling in a variety of cells by cleavage of protease-activated receptors. This review will focus on the roles of TF in protective hemostasis and pathological thrombosis.


Asunto(s)
Hemostasis , Tromboplastina/metabolismo , Trombosis/sangre , Animales , Aterosclerosis/sangre , Aterosclerosis/complicaciones , Coagulación Sanguínea , Factor IX/metabolismo , Factor VIIa/metabolismo , Factor X/metabolismo , Fibrinolíticos/uso terapéutico , Regulación de la Expresión Génica , Hemostasis/efectos de los fármacos , Humanos , Neoplasias/sangre , Neoplasias/complicaciones , Factores de Riesgo , Sepsis/sangre , Sepsis/complicaciones , Transducción de Señal , Tromboplastina/antagonistas & inhibidores , Tromboplastina/genética , Trombosis/tratamiento farmacológico , Trombosis/etiología , Trombosis/genética
17.
Bull Exp Biol Med ; 166(4): 503-506, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30783838

RESUMEN

Immunohistochemistry of tissue factor, vimentin, and cytokeratin-8 was studied in the medical abortion material at gestation week 5-10 in 20 healthy women. No expression of tissue factor was found in vimentin-positive decidual cells of the parietal endometrium. By contrast, intensive immunostaining of the TF/FVIIα complex was detected in the marginal layer of Rohr fibrinoid and less intense staining was found in vimentin-positive decidual cells adjacent to the walls of modified spiral arteries in the utero-placental region under conditions of invasion of the interstitial and intravascular cytotrophoblast (marker: cytokeratin-8). In the villi, tissue factor was expressed only in the fibrinoid lumps at sites of the syncytiotrophoblast defects. We demonstrated the formation of local hemostasis system in the uteroplacental region at weeks 5-8. This system creates optimal conditions for external thrombogenesis aimed at realization of the cytotrophoblast invasion and subsequent flow of arterial blood between the villi at the end of the first trimester.


Asunto(s)
Hemostasis/fisiología , Tromboplastina/metabolismo , Adulto , Decidua/citología , Decidua/metabolismo , Factor VIIa/metabolismo , Femenino , Hemostasis/genética , Humanos , Inmunohistoquímica , Embarazo , Primer Trimestre del Embarazo
18.
Curr Opin Hematol ; 25(3): 219-226, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29461258

RESUMEN

PURPOSE OF REVIEW: Endothelial cell protein C receptor (EPCR), a transmembrane glycoprotein present on the surface of endothelial cells and other cell types, is an essential component of the protein C (PC) anticoagulant system. EPCR is also shown to play a critical role in mediating activated protein C (APC)-induced cytoprotective signaling. The purpose of this review is to outline the mechanisms of EPCR-dependent cell signaling and discuss recent findings made in this area. RECENT FINDINGS: Recent studies showed that the cleavage of protease-activated receptor (PAR)1 at a noncanonical site by APC-EPCR or the canonical site by thrombin when PC occupies EPCR induces ß-arrestin-2-mediated biased cytoprotective signaling. Factor VIIa binding to EPCR is also shown to induce the cytoprotective signaling. EPCR is found to be a reliable surface marker for identifying human hematopoietic stem cells in culture. EPCR, binding to diverse ligands, is thought to play a role in the pathogenesis of severe malaria, immune functions, and cancer by either blocking the APC-mediated signaling or by mechanisms that are yet to be elucidated. SUMMARY: Recent studies provide a mechanistic basis to how EPCR contributes to PAR1-mediated biased signaling. EPCR may play a role in influencing a wide array of biological functions by binding to diverse ligands.


Asunto(s)
Células Endoteliales/metabolismo , Receptor de Proteína C Endotelial/metabolismo , Factor VIIa/metabolismo , Proteína C/metabolismo , Receptor PAR-1/metabolismo , Transducción de Señal , Animales , Células Endoteliales/patología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Malaria/metabolismo , Malaria/patología , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Arrestina beta 2/metabolismo
19.
J Biol Chem ; 292(39): 16249-16256, 2017 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-28801460

RESUMEN

Interactions of soluble proteins with the cell membrane are critical within the blood coagulation cascade. Of particular interest are the interactions of γ-carboxyglutamic acid-rich domain-containing clotting proteins with lipids. Variability among conventional analytical methods presents challenges for comparing clotting protein-lipid interactions. Most previous studies have investigated only a single clotting protein and lipid composition and have yielded widely different binding constants. Herein, we demonstrate that a combination of lipid bilayer nanodiscs and a multiplexed silicon photonic analysis technology enables high-throughput probing of many protein-lipid interactions among blood-clotting proteins. This approach allowed direct comparison of the binding constants of prothrombin, factor X, activated factor VII, and activated protein C to seven different binary lipid compositions. In a single experiment, the binding constants of one protein interacting with all lipid compositions were simultaneously determined. A simple surface regeneration then facilitated similar binding measurements for three other coagulation proteins. As expected, our results indicated that all proteins exhibit tighter binding (lower Kd ) as the proportion of anionic lipid increases. Interestingly, at high proportions of phosphatidylserine, the Kd values of all four proteins began to converge. We also found that although koff values for all four proteins followed trends similar to those observed for the Kd values, the variation among the proteins was much lower, indicating that much of the variation came from the kinetic binding (kon) of the proteins. These findings indicate that the combination of silicon photonic microring resonator arrays and nanodiscs enables rapid interrogation of biomolecular binding interactions at model cell membrane interfaces.


Asunto(s)
Factor VIIa/metabolismo , Factor X/metabolismo , Ácidos Fosfatidicos/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidilserinas/metabolismo , Proteína C/metabolismo , Protrombina/metabolismo , Factor VIIa/química , Factor VIIa/genética , Factor X/química , Ensayos Analíticos de Alto Rendimiento , Humanos , Cinética , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Nanoestructuras/química , Fenómenos Ópticos , Ácidos Fosfatidicos/química , Fosfatidilcolinas/química , Fosfatidilserinas/química , Análisis por Matrices de Proteínas , Proteína C/química , Protrombina/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Silicio/química
20.
Br J Haematol ; 181(4): 528-536, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29611182

RESUMEN

Bypassing therapy is essential for the haemostatic management of patients with haemophilia A with inhibitor (PWHA-inh), but the therapeutic effects are inconsistent. We previously reported that activated prothrombin complex concentrates (aPCC) activated factor (F)VIIIin vitro, and was mediated mainly by the activated FVII (FVIIa) contained in aPCC. We have extended those studies to assess global coagulation in whole blood from 18 PWHA-inh in the co-presence of aPCC and FVIII using Ca2+ -triggered rotational thromboelastometry. The clot times (CTs) in the presence of both aPCC (0·05 iu/ml) and recombinant (r)FVIII (1 iu/ml) ex vivo were shortened compared to the aPCC alone (P < 0·01). These enhancing effects of rFVIII were observed, irrespective of recognizing inhibitor epitopes; however, the clot formation time and 'α'-angle were not significantly different. In samples from 7 PWHA-inh post-infusion of aPCC (70-80 iu/kg), only the CTs were shortened in the presence of rFVIIIex vivo compared to its absence (P < 0·05), indicating that the enhanced activity centred on the initiation phase of coagulation. Furthermore, experiments in the co-presence of rFVIIa and rFVIII demonstrated that FVIII accelerated only the CTs. We concluded that FVIII/FVIIa-related coagulation mechanism enhanced global haemostatic function by the co-presence of bypassing agents and FVIII in PWHA-inh.


Asunto(s)
Inhibidores de Factor de Coagulación Sanguínea/sangre , Factores de Coagulación Sanguínea/administración & dosificación , Coagulación Sanguínea/efectos de los fármacos , Factor VIII , Factor VIIa , Hemofilia A , Factores de Coagulación Sanguínea/farmacocinética , Factor VIII/administración & dosificación , Factor VIII/antagonistas & inhibidores , Factor VIII/metabolismo , Factor VIIa/antagonistas & inhibidores , Factor VIIa/metabolismo , Femenino , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Humanos , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA