Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 507
Filtrar
1.
Prostaglandins Other Lipid Mediat ; 167: 106740, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37119935

RESUMEN

Epoxyeicosatrienoic acids (EETs), which are synthesized from arachidonic acid by cytochrome P450 epoxygenases, function primarily as autocrine and paracrine effectors in the cardiovascular system. So far, most research has focused on the vasodilatory, anti-inflammatory, anti-apoptotic and mitogenic properties of EETs in the systemic circulation. However, whether EETs could suppress tissue factor (TF) expression and prevent thrombus formation remains unknown. Here we utilized in vivo and in vitro models to investigate the effects and underlying mechanisms of exogenously EETs on LPS induced TF expression and inferior vein cava ligation induced thrombosis. We observed that the thrombus formation rate and the size of the thrombus were greatly reduced in 11,12-EET treated mice,accompanied by decreased TF and inflammatory cytokines expression. Further in vitro studies showed that by enhancing p38 MAPK activation and subsequent tristetraprolin (TTP) phosphorylation, LPS strengthened the stability of TF mRNA and induced increased TF expression. However, by strengthening PI3K-dependent Akt phosphorylation, which acted as a negative regulator of p38-TTP signaling pathway,11,12-EET reduced LPS-induced TF expression in monocytes. In addition, 11,12-EET inhibited LPS-induced NF-κB nuclear translocation by activating the PI3K/Akt pathway. Further study indicated that the inhibitory effect of 11,12-EET on TF expression was mediated by antagonizing LPS-induced activation of thromboxane prostanoid receptor. In conclusion, our study demonstrated that 11,12-EET prevented thrombosis by reducing TF expression and targeting the CYP2J2 epoxygenase pathway may represent a novel approach to mitigate thrombosis related diseases.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt , Trombosis , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Lipopolisacáridos/farmacología , Tromboplastina/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Transducción de Señal , Citocromo P-450 CYP2J2 , Ácido 8,11,14-Eicosatrienoico/metabolismo , Trombosis/tratamiento farmacológico , Estabilidad del ARN
2.
J Mater Chem B ; 11(12): 2778-2788, 2023 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-36891927

RESUMEN

The development of hemostatic materials suitable for diverse emergency scenarios is of paramount significance, and there is growing interest in wound-site delivery of hemostasis-enhancing agents that can leverage the body's inherent mechanisms. Herein we report the design and performance of a biomimetic nanoparticle system enclosing tissue factor (TF), the most potent known blood coagulation trigger, which was reconstituted into liposomes and shielded by the liposome-templated CaCO3 mineralization. The mineral coatings, which mainly comprised water-soluble amorphous and vateritic phases, synergized with the lipidated TF to improve blood coagulation in vitro. These coatings served as sacrificial masks capable of releasing Ca2+ coagulation factors or propelling the TF-liposomes via acid-aided generation of CO2 bubbles while endowing them with high thermostability under dry conditions. In comparison to commercially available hemostatic particles, CaCO3 mineralized TF-liposomes yielded significantly shorter hemostasis times and less blood loss in vivo. When mixed with organic acids, the CO2-generating formulation further improved hemostasis by delivering TF-liposomes deep into actively bleeding wounds with good biocompatibility, as observed in a rat hepatic injury model. Therefore, the designed composite mimicry of coagulatory components exhibited strong hemostatic efficacy, which in combination with the propulsion mechanism would serve as a versatile approach to treating a variety of severe hemorrhages.


Asunto(s)
Hemostáticos , Tromboplastina , Ratas , Animales , Tromboplastina/farmacología , Liposomas/farmacología , Dióxido de Carbono , Coagulación Sanguínea , Hemostáticos/farmacología , Hemorragia
3.
Diab Vasc Dis Res ; 19(5): 14791641221129877, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36250331

RESUMEN

PURPOSE: Combination of Rivaroxaban plus Aspirin improved cardiovascular outcome in patients with stable cardiovascular disease. The aim was to determine if Rivaroxaban and acetylsalicylic acid alone or in combination may protect mitochondrial mitophagy in human coronary artery endothelial cells (HCAEC) exposed to D-glucose. METHODS: HCAEC were incubated under different conditions: 5 mmol/L glucose D-glucose (control), 30 mmol/L D-Glucose with and without 50 nmol/L Rivaroxaban (Rivaroxaban), 0.33 mmol/L ASA (ASA) or Rivaroxaban (12.5 nmol/L)+ASA (0.33 mmol/L; (Riva+ASA). RESULTS: HCAEC incubated with D-glucose showed an increased Factor Xa expression. The mitochondrial content of Pink-1 and Parkin were significantly reduced in high glucose-incubated HCAEC compared to control. Rivaroxaban+ASA significantly increased the mitochondrial content of Pink-1 and Parkin, and the mitochondrial membrane potential compared to D-Glucose group. Both ASA alone and Riva+ASA reduced reactive oxygen species (ROS) and tissue factor production induced by high glucose exposure. CONCLUSION: Under high glucose condition combining Rivaroxaban+ASA increased the mitochondrial content of Pink-1 and Parkin, restored mitochondria membrane potential and reduced ROS and tissue factor expression in HCAEC. It suggests potential effects induced by dual use of Rivaroxaban and ASA on the coronary endothelium subjected to high glucose condition.


Asunto(s)
Aspirina , Rivaroxabán , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Endotelio , Factor Xa/metabolismo , Factor Xa/farmacología , Glucosa/metabolismo , Humanos , Mitocondrias , Mitofagia , Especies Reactivas de Oxígeno/metabolismo , Rivaroxabán/metabolismo , Rivaroxabán/farmacología , Tromboplastina/metabolismo , Tromboplastina/farmacología , Ubiquitina-Proteína Ligasas/metabolismo
4.
Bull Cancer ; 109(7-8): 795-804, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35680461

RESUMEN

INTRODUCTION: Tissue factor (TF) is an important predictor for poor prognosis of Hepatocellular carcinoma (HCC). TF can also upregulate the expression of BCL2, which is a key inhibitor of autophagy responses. This study aims to explore the role of BCL2-dependent autophagy in TF-regulated HCC carcinogenesis. METHODS: In this study, we explored the roles of TF in HCC using gene overexpression and silencing assays. Besides, we further identified the significance of BCL2-Beclin1-autophagy signaling on TF-regulated HCC tumorigenesis by combining TF silencing with pharmacological autophagy inhibitor (3-MA). RESULTS: The experimental data showed that the overexpressed TF promoted BCL2 protein expression and inhibited the autophagy activity (shown as LC3 conversion rate, p62 expression and autophagosomes) while maintaining the survival in HCC cells. In contrast, the silent TF showed the completely opposite results. Furthermore, TF knockdown promoted the dissociation of Beclin1 from BCL2-Beclin1 complex. In addition, the enhanced autophagy and inhibited survival by TF knockdown could be reversed by autophagy inhibition with 3-MA or spautin-1 (Beclin1 specific inhibitor) in HCC cells. Xenografts assays also showed that TF-silencing HCC cells had stronger tumorigenicity in vivo, which was recovered by spautin-1 administration. CONCLUSIONS: TF inhibits autophagy-related death by enhancing BCL2 expression, whereby promoting HCC tumorigenesis.


Asunto(s)
Autofagia , Carcinoma Hepatocelular , Neoplasias Hepáticas , Tromboplastina , Apoptosis , Autofagia/efectos de los fármacos , Autofagia/genética , Beclina-1/metabolismo , Carcinogénesis/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Humanos , Neoplasias Hepáticas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Tromboplastina/metabolismo , Tromboplastina/farmacología
5.
Curr Pharm Des ; 28(24): 2001-2009, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35619253

RESUMEN

BACKGROUND: Ruscogenin (RUS) has anti-inflammatory and antithrombotic effects, while its potential effects on deep venous thrombosis (DVT) and pulmonary embolism (PE) remain unclear. OBJECTIVE: We aimed to elucidate the effects of RUS on DVT and PE induced by the inferior vena cava stenosis (IVCS) model and investigate the underlying mechanism. METHODS: Male C57/BL6 mice were used to explore whether IVCS model could be complicated with deep venous thrombosis and pulmonary embolism. Then, effects of RUS on DVT and PE related inflammatory factors and coagulation were examined using H&E staining, ELISA, and real-time PCR. Western blot analysis was used to examine the effects of RUS on MEK/ERK/Egr-1/TF signaling pathway in PE. RESULTS: IVCS model induced DVT and complied with PE 48 h after surgery. Administration of RUS (0.01, 0.1, 1 mg/kg) inhibited DVT, decreased biomarker D-Dimer, cardiac troponin I, N-Terminal probrain natriuretic peptide in plasma to ameliorate PE induced by IVCS model. Meanwhile, RUS reduced tissue factor and fibrinogen content of lung tissue, inhibited P-selectin and C-reactive protein activity in plasma, and suppressed the expressions of interleukin-6 and interleukin-1ß in mice. Furthermore, RUS suppressed the phosphorylation of ERK1/2 and MEK1/2, decreasing the expressions of Egr-1 and TF in the lung. CONCLUSION: IVCS model contributed to the development of DVT and PE in mice and was associated with increased inflammation. RUS showed therapeutic effects by inhibiting inflammation as well as suppressing the activation of MEK/ERK/Egr-1/TF signaling pathway.


Asunto(s)
Embolia Pulmonar , Trombosis de la Vena , Animales , Constricción Patológica/complicaciones , Inflamación/complicaciones , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/uso terapéutico , Embolia Pulmonar/tratamiento farmacológico , Transducción de Señal , Espirostanos , Tromboplastina/metabolismo , Tromboplastina/farmacología , Tromboplastina/uso terapéutico , Vena Cava Inferior/metabolismo , Trombosis de la Vena/tratamiento farmacológico
6.
Nitric Oxide ; 122-123: 6-18, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35202833

RESUMEN

BACKGROUND: Extracellular vesicles (EVs) derived from endothelial cells are elevated in cardiovascular disease and promote inflammation and coagulation. Hypoxia is often a key feature and is itself a potent stimulator of increased EV production. Inorganic nitrite (NO2-) has beneficial and protective effects that are enhanced in hypoxia. OBJECTIVES: Investigate the impact of hypoxia on the functional capacity of EV derived from endothelial cells under hypoxia, and assess whether pre-treatment of endothelial cells with NO2- can alter EV function. METHODS: Differential ultracentrifugation was used to isolate EV from the cultured endothelial cell line HECV (CEV), and from primary human umbilical cord derived endothelial cells (PEV), with time-resolved fluorescence used to assess EV protein composition. Clot formation was induced by thrombin and calcium in two assays; using an Alexa Fluor 594 human fibrinogen conjugate assay and standard turbidometry. Platelet aggregation was determined using multiple electrode aggregometry. Scanning electron microscopy was used to visualise fibrin clots. RESULTS: Hypoxia exposure (1% O2) significantly increased CEV production in comparison to normoxia (21% O2) (1825 ± 72 EVs/cell vs 117 ± 9 EVs/cell, p < 0.001, respectively) but had no effect on CEV mean size (221 ± 6 nm vs 203 ± 4 nm, p > 0.05). Hypoxia-derived PEVs contained significantly more tissue factor than normoxia-derived EVs (Relative Fluorescence Units (RFU) = 7666 ± 1698 vs 5958 ± 1644, p < 0.001, respectively) and less tissue factor pathway inhibitor (RFU = 9799 ± 2353 vs 19723 ± 2698, p < 0.05). Hypoxia significantly increased CEV induced fibrin polymer formation compared to normoxia (% area = 46.98 ± 0.97 vs 36.36 ± 0.72, p < 0.05). Pre-treatment of endothelial cells with NO2- in hypoxia abrogated this effect (% area = 15.70 ± 1.99, p < 0.001). Hypoxia derived CEV non-significantly increased the maximum clot formed, shortened time to max clot, and increased time to clot lysis by turbidometry. ADP-mediated platelet aggregation was significantly elevated with PEV derived from hypoxia compared to normoxia (888.0 ± 32.2 AU*min vs 671.5.2 ± 28.3 AU*min, p < 0.01). This was abrogated by pre-treatment of hypoxic endothelial cells with NO2- (716.5 ± 744.3 AU*min, p < 0.001). CONCLUSIONS: Hypoxia-derived PEVs and CEVs exhibit increased procoagulant activity compared to normoxia-derived EVs, which we confirm to be mediated by an imbalance of TF/TFPI. Pre-treatment of endothelial cells with NO2- reduces the pro-coagulant activity of EVs via a mechanism that is Hypoxia-inducible factor 1 (HIF-1) dependent, but independent of TF/TFPI.


Asunto(s)
Vesículas Extracelulares , Trombosis , Células Endoteliales/metabolismo , Vesículas Extracelulares/metabolismo , Fibrina/metabolismo , Fibrina/farmacología , Humanos , Hipoxia/metabolismo , Nitritos/metabolismo , Dióxido de Nitrógeno , Tromboplastina/metabolismo , Tromboplastina/farmacología , Trombosis/metabolismo
7.
J Biomed Mater Res B Appl Biomater ; 109(8): 1116-1123, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33369080

RESUMEN

Uncontrolled hemorrhage accounts for a significant proportion of annual mortality worldwide. The development of bioinspired hemostatic composites can effectively reduce hemorrhage and related deaths. This work aims to develop an efficient hemostatic agent by incorporating tissue factor (TF) integrated liposomes and collagen, which are capable of augmenting different inherent hemostatic mechanisms, into hemostasis-stimulating alginate matrix. The composite of TF, collagen and alginate (TCA) was made into hydrogel beads with a diameter range of 2.5-3.5 mm, followed by electron microscopy, infrared spectroscopy, rheological, and swelling characterization to confirm its composition and hydrogel nature. When the TCA beads were introduced into simulated body fluid, a controlled release of the loaded TF-liposomes was observed, which also accelerated with the increase of temperature, obtaining intact free proteoliposomes as demonstrated by fluorescence measurement. It is further seen that TCA beads induced the coagulation of whole rabbit blood in about 4.5 min, as compared to ~14.4 min for the control with only recalcified blood. The lipidated TF, collagen and alginate in TCA beads showed a positive synergistic effect on coagulation, while among them a decreasing procoagulant effect was observed. Finally, we demonstrated by a live/dead cell assay that TCA particles had undetectable cytotoxicity. Thus, the TCA hydrogel macrobeads may offer a potential platform for the development of potent hemostatic agents.


Asunto(s)
Alginatos , Colágeno , Hemostáticos , Hidrogeles , Tromboplastina , Alginatos/química , Alginatos/farmacología , Animales , Colágeno/química , Colágeno/farmacología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacología , Hemostáticos/química , Hemostáticos/farmacología , Hidrogeles/química , Hidrogeles/farmacología , Ratones , Células 3T3 NIH , Conejos , Tromboplastina/química , Tromboplastina/farmacología
8.
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
9.
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
10.
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
11.
Int J Oncol ; 55(4): 823-832, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31432158

RESUMEN

The selective induction of tumor vascular thrombosis using truncated tissue factor (tTF) delivered via a target ligand is a promising novel antitumor strategy. In the present study, an anti­neuropilin­1 (NRP­1) monoclonal antibody (mAb)­streptavidin (SA):tTF­biotin (B) composite system was established. In this system, anti­NRP­1­mAb located tTF to the tumor vascular endothelial cell surface and induced vascular embolization. Due to their high binding affinity, SA and B were used to enhance thrombogenic activity. mAb was conjugated with SA using a coupling method with water­soluble 1­ethyl­3­(3­dimethylaminopropyl) carbodiimide and N­hydroxysulfosuccinimide. Biotinylated tTF (tTF­B) was prepared using a B­labeling kit subsequent to the generation and purification of fusion protein tTF. Confocal microscopy and flow cytometry indicated that the anti­NRP­1­mAb­SA conjugate retained mAb targeting activity. The preservation of B­conjugate binding capacity was confirmed using a competitive ELISA, and factor X­activation analysis revealed that tTF­B retained the procoagulant activity exhibited by tTF. Live imaging was performed to assess mAb­SA distribution and tumor­targeting capability, and this yielded promising results. The results of in vivo studies in mice with subcutaneous xenografts demonstrated that this composite system significantly induced tumor vascular thrombosis and inhibited tumor growth, whereas these histological changes were not observed in normal organs.


Asunto(s)
Antineoplásicos Inmunológicos/administración & dosificación , Neoplasias Hepáticas/tratamiento farmacológico , Neuropilina-1/inmunología , Tromboplastina/administración & dosificación , Trombosis/inducido químicamente , Animales , Antineoplásicos Inmunológicos/química , Antineoplásicos Inmunológicos/farmacología , Factor X/metabolismo , Femenino , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/metabolismo , Ratones , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/farmacología , Estreptavidina/química , Tromboplastina/química , Tromboplastina/farmacología , Trombosis/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Clin Chem Lab Med ; 57(9): 1349-1357, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-30763258

RESUMEN

Background In the Netherlands, each new lot of test strips for the CoaguChek XS is validated by a group of collaborating centers. The purpose of this study was to assess the accuracy of the international normalized ratio (INR) measured with consecutive test strip lots and the suitability of frozen plasma pools for accuracy evaluation. Methods Each year, a particular lot of CoaguChek XS test strips is used as reference lot. The reference lots have been validated with the International Standard for thromboplastin rTF/09, yielding a mathematical relationship (R1) between reference lot INR and International Standard INR. New lots are compared to the reference lot using patients' capillary blood samples, yielding a relationship (R2) between the new lot INR and the reference lot INR. INRs of the blood samples were within the 1.5-4.5 interval. In parallel, three frozen plasmas pools are analyzed with the test strips. The distance of each plasma point to the line of relationship R2 was assessed. Results Fifty-four test strip lots have been evaluated during 3 years (2014-2016). Mean INR differences between test strip lot and International Standard rTF/09 varied between -0.14 and +0.20 (-4% and +8%, respectively). A positive trend with strip lot sequence number was observed (p<0.001). In several cases, the distance of the frozen plasmas to the whole blood relationship (R2) was greater than the critical value for commutability. Conclusions Using whole blood, all evaluated test strip lots met the analytical bias criterion of ±10%. Frozen plasma pools behave differently compared to whole blood and are not suitable for assessing absolute accuracy of new CoaguChek XS test strips.


Asunto(s)
Tiempo de Protrombina/métodos , Tiras Reactivas/normas , Manejo de Especímenes/métodos , Anticoagulantes/farmacología , Coagulación Sanguínea/efectos de los fármacos , Recolección de Muestras de Sangre/métodos , Recolección de Datos , Monitoreo de Drogas/métodos , Femenino , Humanos , Relación Normalizada Internacional/normas , Masculino , Persona de Mediana Edad , Países Bajos , Plasma , Pruebas en el Punto de Atención/normas , Reproducibilidad de los Resultados , Tromboplastina/farmacología
13.
J Drug Target ; 27(8): 885-895, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30628471

RESUMEN

The cellular transmembrane receptor Neuropilin-1(NRP-1) is overexpressed in tumour tissue and endothelial cells of tumour vessels, whereas it has limited expression in normal tissues. This study aimed to design a novel recombinant protein tTF-EG3287, which consisting of the truncated tissue factor (tTF) and the NRP-1 targeting peptide EG3287. The procoagulant protein selectively activates blood coagulation in tumour vessels once bound to the cell surface of the tumour vasculature by a targeting peptide EG3287. In this study, procoagulant activity of the recombinant protein tTF-EG3287 was evaluated by Spectozyme FXa assay. NRP-1 targeting ability was analysed by fluorescence confocal microscopy and flow cytometry. The living imaging system was used to assess the tumour targeting ability of recombinant proteins tTF-EG3287 in vivo. Tumour growth inhibition showed effective antitumor activity in HepG2 tumour-bearing nude mice. Histological study showed obvious thrombosis and thromboembolism in tumour vessels and cell necrosis of tumour tissue, without any clear side effect such as thrombosis in other organs.


Asunto(s)
Neuropilina-1/metabolismo , Tromboplastina/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Fragmentos de Péptidos/farmacología , Péptidos/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
14.
Mol Carcinog ; 57(12): 1707-1722, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30129687

RESUMEN

Apart from blood coagulation, coagulation proteases are involved inextricably in cancer progression/propagation via intra/inter-cellular signaling, mediated predominantly by protease-activated receptors (PARs). Microvesicles (MVs), a plasma membrane shredded component, has recently been identified as an important contributor to human breast cancer metastasis. However, the role of PAR2 in promoting MVs generation from breast cancer cells remains largely unexplored. The objective of this study is to investigate whether coagulation protease-mediated human breast cancer propagation commences via MVs and also to decipher the underlying signaling mechanism. Here, we elicited that coagulation factor-FVIIa and Trypsin activates PAR2, which governs MVs shedding from MDAMB231 cells by altering actomyosin dynamics. Treatment of cells with PAR2 activators facilitate MVs generation by activating three independent (MAPK, P38, and Rho) signaling cascades. MAPK, signals through activating MLCK followed by MLC phosphorylation to alter myosin organization whereas, P38 reorganizes actin dynamics by the sequential activation of MK2 and HSP27. RhoA-dependent ROCK-II activation again contributes to remodeling myosin II activity. Further, both our in vitro and in vivo analyses showed that these MVs potentiate invasive and migratory property to the recipient cells. Breast cancer patients blood show an elevation of TF-bearing, pro-metastatic MVs than normal. These findings give an insight into the detailed signaling mechanism involved in the production of MVs with transforming ability from PAR2-activated human breast cancer cells. Understanding these mechanistic details will certainly help to identify crucial targets for therapeutic interventions in MVs-associated human breast cancer metastasis.


Asunto(s)
Actomiosina/metabolismo , Neoplasias de la Mama/metabolismo , Micropartículas Derivadas de Células/trasplante , Receptores Acoplados a Proteínas G/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Micropartículas Derivadas de Células/metabolismo , Factor VIIa/farmacología , Femenino , Humanos , Células MCF-7 , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Fosforilación , Receptor PAR-2 , Transducción de Señal , Tromboplastina/farmacología , Tripsina/farmacología
15.
Clin Exp Med ; 18(3): 325-336, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29417256

RESUMEN

The effect of direct oral anticoagulants (DOACs) on turbidimetric measurements of plasma clot formation and susceptibility to fibrinolysis may facilitate a comparison between different classes of anticoagulants in plasma samples. We obtained 424 citrate plasma samples from 226 atrial fibrillation patients on anticoagulation and 24 samples without anticoagulation serving as controls. As comparators, we measured the international normalized ratio (INR) for phenprocoumon samples (N = 166), anti-Xa for low molecular weight heparin (LMWH) samples (N = 42), and DOAC levels with mass spectrometry (dabigatran N = 40, rivaroxaban N = 110, apixaban N = 42). Plasma clot formation and lysis were recorded continuously on a photometer after addition of an activation mix (tissue factor 2 pmol/l and tissue plasminogen activator 333 ng/ml). We used linear regression and ANCOVA for correlation analysis. Clot formation lag phase was prolonged in the presence of anticoagulants in a concentration-dependent manner for DOACs (dabigatran Spearman r = 0.74; rivaroxaban r = 0.78; apixaban r = 0.72, all p < 0.0001), INR dependent for phenprocoumon (r = 0.59, p < 0.0001), anti-Xa level dependent in LMWH samples (r = 0.90, p < 0.0001). Maximum rate of clot formation and peak clot turbidity were reduced in the presence of anticoagulants, but correlated only moderately with the comparator measures of anticoagulation. The clot lysis time was inversely correlated with DOAC concentrations in the presence of recombinant thrombomodulin. A direct ex vivo comparison between the effects of different classes of anticoagulants is possible with turbidimetric measurement of plasma clot formation and lysis. Anticoagulation inhibited clot formation in a plasma concentration manner for DOACs, INR dependent for phenprocoumon, and anti-Xa dependent for LMWH. Susceptibility to fibrinolysis increased with increasing DOAC concentrations.


Asunto(s)
Anticoagulantes/farmacología , Fibrilación Atrial/tratamiento farmacológico , Coagulación Sanguínea/efectos de los fármacos , Anciano , Fibrilación Atrial/sangre , Fibrilación Atrial/fisiopatología , Estudios de Casos y Controles , Dabigatrán/farmacología , Inhibidores del Factor Xa/farmacología , Femenino , Tiempo de Lisis del Coágulo de Fibrina , Heparina de Bajo-Peso-Molecular/farmacología , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Fenprocumón/farmacología , Plasma/efectos de los fármacos , Plasma/metabolismo , Pirazoles/farmacología , Piridonas/farmacología , Rivaroxabán/farmacología , Tromboplastina/farmacología , Activador de Tejido Plasminógeno/farmacología
16.
Proc Natl Acad Sci U S A ; 114(47): 12454-12459, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109275

RESUMEN

Recombinant factor VIIa (FVIIa) variants with increased activity offer the promise to improve the treatment of bleeding episodes in patients with inhibitor-complicated hemophilia. Here, an approach was adopted to enhance the activity of FVIIa by selectively optimizing substrate turnover at the membrane surface. Under physiological conditions, endogenous FVIIa engages its cell-localized cofactor tissue factor (TF), which stimulates activity through membrane-dependent substrate recognition and allosteric effects. To exploit these properties of TF, a covalent complex between FVIIa and the soluble ectodomain of TF (sTF) was engineered by introduction of a nonperturbing cystine bridge (FVIIa Q64C-sTF G109C) in the interface. Upon coexpression, FVIIa Q64C and sTF G109C spontaneously assembled into a covalent complex with functional properties similar to the noncovalent wild-type complex. Additional introduction of a FVIIa-M306D mutation to uncouple the sTF-mediated allosteric stimulation of FVIIa provided a final complex with FVIIa-like activity in solution, while exhibiting a two to three orders-of-magnitude increase in activity relative to FVIIa upon exposure to a procoagulant membrane. In a mouse model of hemophilia A, the complex normalized hemostasis upon vascular injury at a dose of 0.3 nmol/kg compared with 300 nmol/kg for FVIIa.


Asunto(s)
Terapia Biológica/métodos , Factor VIIa/química , Hemofilia A/terapia , Ingeniería de Proteínas/métodos , Tromboplastina/química , Regulación Alostérica , Animales , Coagulación Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Factor VIIa/genética , Factor VIIa/farmacología , Factor VIIa/uso terapéutico , Femenino , Hemofilia A/fisiopatología , Humanos , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simulación de Dinámica Molecular , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Tromboplastina/genética , Tromboplastina/farmacología , Tromboplastina/uso terapéutico
17.
Lung Cancer ; 113: 121-127, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-29110838

RESUMEN

OBJECTIVES: Zinc-binding protease aminopeptidase N (CD13) is expressed on tumor vascular cells and tumor cells. It represents a potential candidate for molecular targeted therapy, e.g. employing truncated tissue factor (tTF)-NGR, which can bind CD13 and thereby induce tumor vascular infarction. We performed a comprehensive analysis of CD13 expression in a clinically well characterized cohort of patients with small cell lung cancer (SCLC) to evaluate its potential use for targeted therapies in this disease. MATERIAL AND METHODS: CD13 expression was analyzed immunohistochemically in 27 SCLC patients and correlated with clinical course and outcome. In CD-1 nude mice bearing human HTB119 SCLC xenotransplants, the systemic effects of the CD13-targeting fusion protein tTF-NGR on tumor growth were tested. RESULTS AND CONCLUSION: In 52% of the investigated SCLC tissue samples, CD13 was expressed in tumor stroma cells, while the tumor cells were negative for CD13. No prognostic effect was found in the investigated SCLC study collective with regard to overall survival (p>0.05). In CD-1 nude mice, xenografts of CD13 negative HTB119 SCLC cells showed CD13 expression in the intratumoral vascular and perivascular cells, and the systemic application of CD13-targeted tissue factor tTF-NGR led to a significant reduction of tumor growth. We here present first data on the expression of CD13 in SCLC tumor samples. Our results strongly recommend the further investigation of tTF-NGR and other molecules targeted by NGR-peptides in SCLC patients. Considering the differential expression of CD13 in SCLC samples pre-therapeutic CD13 analysis is proposed for testing as investigational predictive biomarker for patient selection.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Antígenos CD13/biosíntesis , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Anciano , Animales , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/biosíntesis , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Antígenos CD13/antagonistas & inhibidores , Línea Celular Tumoral , Femenino , Humanos , Infarto/metabolismo , Estimación de Kaplan-Meier , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/metabolismo , Masculino , Ratones Desnudos , Persona de Mediana Edad , Terapia Molecular Dirigida , Péptidos/genética , Péptidos/metabolismo , Péptidos/farmacología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Carcinoma Pulmonar de Células Pequeñas/irrigación sanguínea , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo , Tromboplastina/farmacología
18.
Knee ; 24(6): 1454-1461, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28865948

RESUMEN

BACKGROUND: Total knee arthroplasty is associated with blood loss during the intervention and may require allogenic blood transfusion. Treatments such as tranexamic acid and fibrin sealants improved the bleeding control in several clinical trials, but the hemorrhage associated with the intervention is still significant. Thus far, very few studies have evaluated hemostatic treatments in animal models of total knee arthroplasty. This work describes a sheep model of bleeding associated with total knee arthroplasty and investigates a new class of hemostatic treatment based on recombinant tissue factor. METHODS: Sheep were treated with the anticoagulant heparin, and the joint was accessed by a paramedial incision. Ligaments and menisci were eliminated and femoral condyles and tibia plateau were sectioned exposing the trabecular bone. An intra-articular drain was used to recover and quantify the blood loss during the 90-min period after treatment. The efficacy of one milligram and three milligrams of TT-173 was evaluated and compared with tranexamic acid. The occurrence of analytical alterations and systemic absorption was also investigated. RESULTS: Treatment with TT-173 reduced the blood loss in comparison with control or tranexamic acid. No significant differences were observed between the two doses evaluated. Moreover, a dose of six milligrams of TT-173 did not induce any clinical or analytical alteration, and significant systemic absorption was not observed. CONCLUSION: Data obtained strongly suggest that TT-173 could be useful in reducing the blood loss associated with total knee arthroplasty and without safety concerns derived from the systemic absorption of the product.


Asunto(s)
Artroplastia de Reemplazo de Rodilla/efectos adversos , Pérdida de Sangre Quirúrgica/prevención & control , Hemostáticos/farmacología , Tromboplastina/farmacología , Animales , Anticoagulantes/farmacología , Antifibrinolíticos/farmacología , Drenaje , Femenino , Hemostáticos/efectos adversos , Heparina/farmacología , Masculino , Ovinos , Tromboplastina/efectos adversos , Ácido Tranexámico/farmacología
19.
Vet J ; 223: 5-11, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28671072

RESUMEN

Rivaroxaban is an oral, direct factor Xa inhibitor used in human thrombotic disorders. In view of the in vitro concentration dependent anticoagulant effects of rivaroxaban in dogs, the time course of its anticoagulant effects was characterized in healthy dogs. Twenty-four healthy Beagles were randomized into three groups (n = 8 per group) and received orally either a placebo or 20 mg rivaroxaban once or twice at an 8 h interval. Fifteen blood samples were collected over a 30 h period, and blindly assayed for prothrombin time (PT), activated partial thromboplastin time (aPTT), tissue factor induced thrombin generation (TG) and anti-factor Xa activity. Thromboelastography (TEG) was evaluated at 0, 1, 4, 8 and 24 h. Peak/baseline anticoagulant effect ratios were analyzed with generalized linear models using ß distributions and times to return to baseline with survival analyses (α = 0.05). Peak/baseline anticoagulant effect ratios of PT, aPTT, anti-factor Xa activity, TG and R (TEG) differed significantly between placebo and both rivaroxaban groups (P <0.0001). The peak anticoagulant effect of rivaroxaban occurred 1.5 to 2 h after dosing. The median return to baseline occurred significantly sooner (P <0.01) with 20 mg rivaroxaban administered once (7.9-18.7 h) versus twice (17.5-26.8 h). The inter-individual variability differed amongst assays, but overall was moderate to large. No adverse effects were recorded. Twice oral administration of 2 mg/kg rivaroxaban at an 8 h interval maintained 24 h anticoagulant activity, but larger studies are needed to establish guidelines for the use of rivaroxaban in dogs.


Asunto(s)
Anticoagulantes , Perros/sangre , Rivaroxabán/farmacología , Rivaroxabán/farmacocinética , Animales , Anticoagulantes/farmacocinética , Anticoagulantes/farmacología , Disponibilidad Biológica , Pruebas de Coagulación Sanguínea/veterinaria , Inhibidores del Factor Xa/sangre , Femenino , Tiempo de Tromboplastina Parcial , Placebos , Tiempo de Protrombina , Rivaroxabán/administración & dosificación , Trombina/biosíntesis , Tromboplastina/farmacología
20.
Blood Coagul Fibrinolysis ; 28(1): 112-116, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27755018

RESUMEN

The objective is to evaluate Grifols' DG-PT L Rec liquid reagent for prothrombin time (PT) determination in comparison to the laboratory's reference reagent (Siemens' Thromborel S). For linearity, the average master curve for PT and five nominal prothrombin concentrations was obtained from five calibration curves. Within-assay precision (repeatability) was calculated after measuring 20 successive tests of normal and pathological controls. For correlation, 581 routine clinical citrated plasma samples were assessed with both reagents. The BCS XP hemostasis analyzer was used. Linearity of the DG-PT L Rec was good (P < 0.001). The coefficient of variation met the desirable imprecision of less than 2% (normal controls: 1.7%; pathological controls: 0.9%). Correlation between DG-PT L Rec and Thromborel S was high (r = 0.9795; PT in %). In subgroups of anticoagulated, low fibrinogen, lipemic, jaundice, and hemolyzed samples the correlation was more than 0.95. Performance of DG-PT L Rec was high and comparable to the reference reagent.


Asunto(s)
Pruebas de Coagulación Sanguínea/métodos , Tiempo de Protrombina/métodos , Tromboplastina/uso terapéutico , Hemostasis , Humanos , Estudios Prospectivos , Tromboplastina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...