Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Z Naturforsch C J Biosci ; 76(1-2): 11-19, 2021 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-32614784

RESUMEN

Tissue factor (TF) which plays a key role in hemostasis and thrombosis appears to be an attractive target and medicinal plants having alkaloids inhibition TF activity benefit to cardiovascular disease (CVD). The aim of study is to explore further knowledge about alkaloids and TF. TF procoagulant activities were determined by the simplified chromogenic assay and their mRNA expression were then examined by reverse transcription and polymerase chain reaction. Besides, the potential of TF/FVIIa binding with four representative alkaloids were analyzed by molecular docking. The results indicated that these isoquinoline alkaloids with various structures had a different effect on suppression of TF activity. Molecular docking showed four alkaloids including l-corydalmine, berberine, jatrorrhizine, and tetrahydropalmatine were stably posed in the active binding pocket of TF/FVIIa. The SARs analysis showed the structural difference including planar, quaternary nitrogen, and the peripheral functional groups at C-8, C-9, C-10, have strong effect on inhibition of TF activity, which provided effective methods to modify isoquinoline alkaloids for inhibiting TF activity. This study provides a further evidence for the cardiovascular protection of isoquinoline alkaloids, and has physiological significance in the clinical challenge to use isoquinoline alkaloids or their potential analogs in the treatment of CVD.


Asunto(s)
Alcaloides/química , Isoquinolinas/química , Simulación del Acoplamiento Molecular , Tromboplastina/metabolismo , Alcaloides/farmacología , Sitios de Unión , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Isoquinolinas/farmacología , Unión Proteica , Relación Estructura-Actividad , Tromboplastina/química
2.
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
3.
Steroids ; 89: 1-10, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25042471

RESUMEN

Six new spirostane glycosides (1-6), named polygodosides A-F, one new furostanol glycoside, polygodoside G (7), one new cholestane glycoside, polygodoside H (8), and one new steroidal sapogenin, polygodosin A (9), together with thirteen known compounds (10-22) were isolated from a 90% MeOH extract of the fibrous roots of Polygonatum odoratum (Mill.) Druce. The structures of new compounds were elucidated by extensive 1D and 2D NMR spectroscopic analyses and mass spectrometry. The effects on TF procoagulant activity in THP-1 cells were tested for most of the compounds.


Asunto(s)
Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Colestanos/química , Glicósidos/química , Sapogeninas/química , Esteroles/química , Tromboplastina/metabolismo , Trastornos de la Coagulación Sanguínea/metabolismo , Colestanos/aislamiento & purificación , Medicamentos Herbarios Chinos/química , Glicósidos/aislamiento & purificación , Humanos , Estructura Molecular , Raíces de Plantas/química , Polygonatum/química , Sapogeninas/aislamiento & purificación , Esteroides/química , Esteroides/aislamiento & purificación , Esteroles/aislamiento & purificación , Tromboplastina/antagonistas & inhibidores , Tromboplastina/química
4.
J Thromb Haemost ; 12(7): 1054-65, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24766850

RESUMEN

BACKGROUND: Increased hypercoagulability has been reported with low doses of direct thrombin inhibitors but not with direct factor Xa inhibitors. OBJECTIVES: To compare the effects of rivaroxaban with those of melagatran and dabigatran on thrombin generation (TG) and tissue factor-induced hypercoagulability and to explore the possible involvement of the thrombin-thrombomodulin/activated protein C system. METHODS: In normal human plasma and in protein C-deficient plasma, TG was investigated in vitro in the presence and absence of recombinant human soluble thrombomodulin (rhs-TM). TG was determined by calibrated automated thrombography and an ELISA for prothrombin fragments 1+2 (F1+2 ). In an in vivo rat model, hypercoagulability was induced by tissue factor; levels of thrombin-antithrombin (TAT) and fibrinogen and the platelet count were determined. RESULTS: Rivaroxaban inhibited TG in a concentration-dependent manner. In the absence of rhs-TM, melagatran and dabigatran also inhibited TG concentration dependently. However, in the presence of rhs-TM, lower concentrations of melagatran (119-474 nmol L(-1) ) and dabigatran (68-545 nmol L(-1) ) enhanced endogenous thrombin potential, peak TG, and F1+2 formation in normal plasma but not in protein C-deficient plasma. In vivo, rivaroxaban dose-dependently inhibited TAT generation, whereas melagatran showed a paradoxical effect, with an increase in TAT and a small decrease in fibrinogen and platelet count at lower doses. CONCLUSION: Low concentrations of the direct thrombin inhibitors melagatran and dabigatran enhanced TG and hypercoagulability, possibly via inhibition of the protein C system. In contrast, rivaroxaban reduced TG and hypercoagulability under all conditions studied, suggesting that it does not suppress this negative-feedback system.


Asunto(s)
Antitrombinas/uso terapéutico , Inhibidores del Factor Xa/uso terapéutico , Morfolinas/uso terapéutico , Tiofenos/uso terapéutico , Trombofilia/tratamiento farmacológico , Tromboplastina/química , Animales , Azetidinas/uso terapéutico , Bencilaminas/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/citología , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Plasma/metabolismo , Recuento de Plaquetas , Proteína C/metabolismo , Protrombina/metabolismo , Ratas , Ratas Wistar , Rivaroxabán , Tromboelastografía , Trombina/metabolismo , Trombomodulina/metabolismo
5.
J Thromb Haemost ; 11(6): 1111-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23578206

RESUMEN

BACKGROUND: Rivaroxaban has been approved as an antithrombotic agent for prevention of venous thromboembolism with specific indications. At present no antidote is appointed and no guidelines have been formulated for the measurement of Rivaroxaban reversal. OBJECTIVES: In the present study, we have evaluated the influence of prothrombin complex concentrate (PCC) on the anticoagulant effects of Rivaroxaban as measured by prothrombin time (PT) and thrombin generation tests (TGTs). METHODS: Plasma and whole blood samples from healthy volunteers were spiked with Rivaroxaban (up to 800 µg L(-1) ) and PCC was added to these samples in concentration ranges as used clinically to reverse the effects of vitamin K antagonists. PT, endogenous thrombin potential (ETP) and calibrated automated thrombography (CAT) assays were performed with varying tissue factor (TF) concentrations. RESULTS: Addition of PCC to Rivaroxaban-spiked samples did not result in normalization of PT and TGT lag time/T-Lag in ETP and CAT, respectively. In contrast, normalization of ETP and CAT area under the curve did occur. However, the response to PCC addition was strongly TF concentration dependent and in whole blood less PCC was required for Rivaroxaban reversal as compared with plasma. CONCLUSIONS: Prothrombin complex concentrate does not neutralize the lengthening effect on PT and TGT lag time/T-Lag of Rivaroxaban anticoagulated blood in vitro; however, total thrombin potential could be normalized. Response of the different TGTs in this respect is assay condition dependent. Therefore, prospective studies are needed to clarify which assay condition and parameter describes in vivo hemostasis best in patients on Rivaroxaban who are treated with PCC.


Asunto(s)
Anticoagulantes/antagonistas & inhibidores , Factores de Coagulación Sanguínea/uso terapéutico , Morfolinas/antagonistas & inhibidores , Morfolinas/química , Tiofenos/antagonistas & inhibidores , Tiofenos/química , Trombina/química , Anticoagulantes/química , Área Bajo la Curva , Coagulación Sanguínea/efectos de los fármacos , Pruebas de Coagulación Sanguínea , Calibración , Fibrinolíticos/química , Humanos , Plasma/efectos de los fármacos , Protrombina/química , Tiempo de Protrombina , Rivaroxabán , Tromboplastina/química , Factores de Tiempo , Vitamina K/antagonistas & inhibidores
6.
Thromb Res ; 131(3): e100-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23313382

RESUMEN

INTRODUCTION: Defective thrombolysis, a thrombotic risk factor, can be attributed to the formation of a compact clot poorly accessible to fibrinolytic enzymes. Venous thrombi, rich in red blood cells (RBCs), and arterial thrombi containing various amounts of RBCS, plasma and whole blood (WB) clot permeability and degradability were compared. The effect of rivaroxaban, a potent direct factor Xa inhibitor, was also evaluated. MATERIALS AND METHODS: Fibrin permeability was determined by flow measurement through the clot. Clot degradability was evaluated by the amount of D-dimer generated by clot perfusion with plasminogen and tissue plasminogen activator. Fibrin clot structure was assessed by confocal microscopy. RESULTS: WB clot permeability (KS) and degradability were 6.7- and 38-fold lower, respectively, compared with plasma clots. This is attributed to 1) occlusion of fibrin pores by RBCs and 2) a consistent increase in thrombin generation due to platelets and RBCs inducing formation of a tighter clot. Rivaroxaban added to plasma or WB before clotting, in reducing thrombin generation, led to the formation of a looser clot that is more degradable by fibrinolytic enzymes. Permeability and degradability of whole blood clots formed in the presence of rivaroxaban were very similar to those of plasma clots. CONCLUSION: The resistance to fibrinolysis of WB clots was reduced considerably when clots were formed with rivaroxaban. These results may have implications for the development of antithrombotic agents.


Asunto(s)
Anticoagulantes/uso terapéutico , Sangre/efectos de los fármacos , Fibrina/química , Morfolinas/uso terapéutico , Plasma/efectos de los fármacos , Tiofenos/uso terapéutico , Terapia Trombolítica/métodos , Trombosis/tratamiento farmacológico , Coagulación Sanguínea , Plaquetas/citología , Eritrocitos/citología , Factor XIII/química , Productos de Degradación de Fibrina-Fibrinógeno/química , Fibrinólisis/efectos de los fármacos , Humanos , Permeabilidad , Factores de Riesgo , Rivaroxabán , Trombina/metabolismo , Tromboplastina/química , Trombosis/metabolismo , Factores de Tiempo
7.
J Biol Chem ; 288(5): 3346-58, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23223577

RESUMEN

Abnormally enhanced tissue factor (TF) activity is related to increased thrombosis risk in which oxidative stress plays a critical role. Human cytosolic thioredoxin (hTrx1) and thioredoxin reductase (TrxR), also secreted into circulation, have the power to protect against oxidative stress. However, the relationship between hTrx1/TrxR and TF remains unknown. Here we show reversible association of hTrx1 with TF in human serum and plasma samples. The association is dependent on hTrx1-Cys-73 that bridges TF-Cys-209 via a disulfide bond. hTrx1-Cys-73 is absolutely required for hTrx1 to interfere with FVIIa binding to purified and cell-surface TF, consequently suppressing TF-dependent procoagulant activity and proteinase-activated receptor-2 activation. Moreover, hTrx1/TrxR plays an important role in sensing the alterations of NADPH/NADP(+) states and transducing this redox-sensitive signal into changes in TF activity. With NADPH, hTrx1/TrxR readily facilitates the reduction of TF, causing a decrease in TF activity, whereas with NADP(+), hTrx1/TrxR promotes the oxidation of TF, leading to an increase in TF activity. By comparison, TF is more likely to favor the reduction by hTrx1-TrxR-NADPH. This reversible reduction-oxidation reaction occurs in the TF extracellular domain that contains partially opened Cys-49/-57 and Cys-186/-209 disulfide bonds. The cell-surface TF procoagulant activity is significantly increased after hTrx1-knockdown. The response of cell-surface TF procoagulant activity to H(2)O(2) is efficiently suppressed through elevating cellular TrxR activity via selenium supplementation. Our data provide a novel mechanism for redox regulation of TF activity. By modifying Cys residues or regulating Cys redox states in TF extracellular domain, hTrx1/TrxR function as a safeguard against inappropriate TF activity.


Asunto(s)
Compuestos de Sulfhidrilo/metabolismo , Reductasa de Tiorredoxina-Disulfuro/metabolismo , Tiorredoxinas/metabolismo , Tromboplastina/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Membrana Celular/metabolismo , Cisteína/metabolismo , Disulfuros/metabolismo , Espacio Extracelular/metabolismo , Factor VIIa/metabolismo , Humanos , Datos de Secuencia Molecular , Oxidación-Reducción , Unión Proteica , Receptor PAR-2/metabolismo , Tromboplastina/antagonistas & inhibidores , Tromboplastina/química , Tromboplastina/aislamiento & purificación
8.
Int J Mol Sci ; 13(1): 409-26, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22312261

RESUMEN

Xylan is one of most abundant polymer after cellulose. However, its potential has yet to be completely recognized. Corn cobs contain a considerable reservoir of xylan. The aim of this work was to study some of the biological activities of xylan obtained from corn cobs after alkaline extraction enhanced by ultrasonication. Physical chemistry and infrared analyses showed 130 kDa heteroxylan containing mainly xylose:arabinose: galactose:glucose (5.0:1.5:2.0:1.2). Xylan obtained exhibited total antioxidant activity corresponding to 48.5 mg of ascorbic acid equivalent/g of xylan. Furthermore, xylan displayed high ferric chelating activity (70%) at 2 mg/mL. Xylan also showed anticoagulant activity in aPTT test. In antimicrobial assay, the polysaccharide significantly inhibited bacterial growth of Klebsiella pneumoniae. In a test with normal and tumor human cells, after 72 h, only HeLa tumor cell proliferation was inhibited (p < 0.05) in a dose-dependent manner by xylan, reaching saturation at around 2 mg/mL, whereas 3T3 normal cell proliferation was not affected. The results suggest that it has potential clinical applications as antioxidant, anticoagulant, antimicrobial and antiproliferative compounds.


Asunto(s)
Antiinfecciosos/química , Anticoagulantes/química , Antioxidantes/química , Xilanos/química , Zea mays/química , Células 3T3 , Animales , Antiinfecciosos/farmacología , Anticoagulantes/metabolismo , Antioxidantes/metabolismo , Biopelículas/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células HeLa , Humanos , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/fisiología , Ratones , Tiempo de Tromboplastina Parcial , Extractos Vegetales/química , Extractos Vegetales/farmacología , Staphylococcus epidermidis/efectos de los fármacos , Staphylococcus epidermidis/fisiología , Superóxidos/química , Tromboplastina/química , Tromboplastina/metabolismo , Xilanos/farmacología , Zea mays/metabolismo
9.
Clin Appl Thromb Hemost ; 15(2): 220-4, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18385150

RESUMEN

Background. Monitoring of oral anticoagulant therapy (OAT) is usually accomplished by measuring prothrombin time and the international normalized ratio (INR). However, thromboplastins have different responsiveness and sensitivity to vitamin K-dependent coagulation factors depletion. Several studies have shown INR variation when low sensitive thromboplastins are used. This study compared INR variability between two laboratories using highly sensitive thromboplastins. Methods. A total of 237 plasmas were tested, half of them from patients under OAT. Samples were tested simultaneously in two laboratories: in laboratory A, a Behring Coagulation Timer instrument and a human recombinant thromboplastin (Innovin, Dade Behring) (ISI 1.01) were used. In laboratory B, a Thrombolyzer Compact (Behnk Elektronik) and a rabbit brain thromboplastin (Simplastin Excel S, Organon Teknika) with an ISI of 1.30 were used. Statistical analysis was carried out according to the method of Bland and Altman. Results. Even though high correlation coefficients were obtained when comparing both laboratories, Bland-Altman analysis showed a variation of INR between laboratories ranging from -0.77 to +1.07. After logarithmic transformation of data, these values yielded a variation of the INR either 25% below or 44% above. Conclusions. These results are clearly inadequate for clinical use because such a variation would most probably induce the clinician to make a change in warfarin dose. Standardization of instruments, reagents, and controls is warranted to decrease this variation.


Asunto(s)
Relación Normalizada Internacional/normas , Anticoagulantes/administración & dosificación , Coagulación Sanguínea/efectos de los fármacos , Femenino , Humanos , Relación Normalizada Internacional/métodos , Masculino , Tiempo de Protrombina , Proteínas Recombinantes/química , Tromboplastina/química
10.
Biochem Soc Trans ; 35(Pt 3): 555-8, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17511650

RESUMEN

Coagulation proteases are involved in a highly orchestrated proteolytic cascade which is essential for haemostasis and blood clotting. In particular, the initiator of the coagulation cascade, Factor VIIa, binds to its cofactor, tissue factor, and its substrate, Factor X, via exosite interactions to form a ternary catalytic complex named extrinsic Xase. These exosite interactions have also been shown to allosterically induce the active conformation of the catalytic site of Factor VIIa. We have developed a direct continuous fluorescence polarization-based extrinsic Xase assay, which has been used to screen in excess of 1 million structurally diverse low-molecular-mass compounds as a potential starting point for the development of anticoagulants. The primary screen hits were categorized with deconvolution assays into either active-site or exosite inhibitors. The latter category of hits displayed both competitive and uncompetitive modalities of inhibition with respect to Factor X activation. An uncompetitive mechanism of action is of particular interest as it offers a hypothetical inhibitory advantage in the context of inhibiting a proteolytic cascade such as the blood coagulation pathway.


Asunto(s)
Factor VIIa/antagonistas & inhibidores , Regulación Alostérica , Sitios de Unión , Evaluación Preclínica de Medicamentos , Factor VIIa/química , Factor VIIa/metabolismo , Factor X/química , Factor X/metabolismo , Polarización de Fluorescencia/métodos , Humanos , Técnicas In Vitro , Cinética , Modelos Moleculares , Complejos Multiproteicos , Tromboplastina/química , Tromboplastina/metabolismo
11.
Thromb Haemost ; 91(1): 16-27, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14691564

RESUMEN

The contribution of the factor Va C1 domain (fVa-C1) to assembly of the prothrombinase complex has not been previously investigated. The homologous fVa-C2 domain contains a binding site for phosphatidylserine (PS) that includes the indole moieties of Trp(2063)/Trp(2064) at the apex of spike-1. In order to investigate the structure and function of fVa-C1 a molecular model was constructed based on the structure of fVa-C2. The aromatic and hydrophobic side chains of Tyr (1956) /Leu (1957) in fVa-C1 are located at the predicted apex of spike-3. Exposed charged and hydrophobic residues in fVa-C1 were changed to alanine in clusters of 1-3 mutations per construct. The resultant 20 mutants were expressed in COS cells and screened for binding to immobilized PS and prothrombinase activity on phospholipid vesicles containing either 25% or 5% PS. Two mutants, (Y1956,L1957)A, and (R2023,R2027)A showed both decreased binding to immobilized PS and a selective decrease in prothrombinase activity on membranes containing 5% PS. The interaction of purified (Y1956,L1957)A with phospholipid vesicles was studied using fluorescence resonance energy transfer and prothrombinase assays. The affinity of (Y1956,L1957)A binding to 25% PS membranes was reduced 12-fold compared to rHFVa. Prothrombin activation in the presence of (Y1956,L1957)A was markedly impaired on phos-pholipid vesicles containing 10% or less PS. We conclude that solvent exposed hydrophobic and aromatic amino acids in both fVa-C1 and fVa-C2 contribute to the interaction of factor V with PS membranes.


Asunto(s)
Membrana Celular/metabolismo , Factor V/química , Alanina/química , Secuencia de Aminoácidos , Animales , Células COS , Medios de Cultivo Condicionados/farmacología , ADN Complementario/metabolismo , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Transferencia Resonante de Energía de Fluorescencia , Humanos , Cinética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación , Fosfatidilserinas/química , Fosfolípidos/química , Unión Proteica , Estructura Terciaria de Proteína , Tromboplastina/química , Tromboplastina/metabolismo
12.
Thromb Haemost ; 83(2): 282-9, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10739387

RESUMEN

Phosphatidylserine (PhtdSer) is an anionic aminophospholipid necessary for the development of optimal tissue factor (TF) activity at the cell surface. This study investigates the implication of a restricted lipid environment with respect to PhtdSer availability on TF expression and activity. K562 cells, showing a reduced ability to externalize PhtdSer, were transfected with human TF cDNA. PhtdSer exposure and TF activity were examined in transfected cells and compared to monocytic THP-1 cells expressing constitutive and inducible TF or megakaryocytic HEL cells showing a high PhtdSer externalization potency. TF expression was evidenced by flow cytometry and its activity measured using functional assays. PhtdSer exposure was monitored by enzymatic prothrombinase assay. One clone (DC9) expressed a stable amount of TF antigen without global modification of its membrane status. Despite a noticeable TF expression level, clone DC9 presented only a weak TF activity even after ionophore stimulation. The apparent Km, relative to factor X (FX) activation by TF-factor VIIa (FVIIa) complex, was 335 nM versus 70 nM for THP-1 cells. The velocity of the reaction was found 3-fold slower in DC9 than THP-1 cells. Ionophore treatment resulting in slightly enhanced amounts of available PhtdSer abolished this difference. The DC9 clone appears suitable for further investigations on the biology of TF expressed at the surface of cells where the contribution of PhtdSer is significantly attenuated. Such cells should enable further assessment of the role of TF as a receptor coupled to intracellular signaling pathways and its fate during apoptotic cell death.


Asunto(s)
Membrana Celular/química , Fosfolípidos/farmacología , Tromboplastina/efectos de los fármacos , Tromboplastina/metabolismo , Antígenos de Superficie/biosíntesis , Pruebas de Coagulación Sanguínea , Células Clonales , ADN Complementario , Factor VIIa/metabolismo , Factor VIIa/farmacología , Factor X/efectos de los fármacos , Factor X/metabolismo , Humanos , Células K562 , Cinética , Fosfatidilserinas/química , Fosfatidilserinas/metabolismo , Fosfatidilserinas/farmacología , Fosfolípidos/química , Tromboplastina/química , Volumetría , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA