Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Br J Dermatol ; 164(2): 308-15, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21039406

RESUMEN

BACKGROUND: Infantile haemangioma (IH) may present as a precursor area of pallor prior to the initial proliferative phase, which implies that the early lesion may be hypoxic. OBJECTIVES: To examine the effect of hypoxia on the expression and activity of two key molecular markers of IH, glucose transporter-1 (GLUT1) and indoleamine 2,3-dioxygenase (IDO). METHODS: IH endothelial cells express both haematopoietic and endothelial cell markers. CD14+ monocyte-derived endothelial-like cells have been employed in the study of IH and is the cell type used in this study. RESULTS: GLUT1 transcript, protein and activity levels were strongly induced by hypoxia and remained elevated following 2 days of normoxic recovery. IDO transcript levels were not affected by hypoxia, although IDO protein level was reduced fivefold and IDO activity >100-fold following 2 days of hypoxia. The protein and activity levels returned to normal following 2 days of normoxic recovery. CONCLUSIONS: The findings link the tissue hypoxia that precedes lesion development and the expression and/or activity of two key IH proteins. The early hypoxic insult may contribute to the elevated GLUT1 levels in IH lesions, while the very low IDO activity during the hypoxic phase may promote activation of immune cells in the lesion, which release cytokines that trigger IDO expression and activity and entry into the proliferative phase. Interestingly, IH lesion development shares some common features with ischaemia-reperfusion injury.


Asunto(s)
Hipoxia de la Célula/fisiología , Células Endoteliales/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Hemangioma/etiología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias Cutáneas/etiología , Biomarcadores de Tumor/metabolismo , Células Cultivadas , Humanos
2.
J Exp Med ; 193(12): 1341-9, 2001 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-11413189

RESUMEN

Plasma von Willebrand factor (vWF) is a multimeric protein that mediates adhesion of platelets to sites of vascular injury. Only the very large vWF multimers are effective in promoting platelet adhesion in flowing blood. A protein disulfide bond reductase in plasma reduces the average multimer size of vWF secreted by endothelial cells. This activity has been isolated from human endothelial cell conditioned medium and shown to be the trimeric glycoprotein, thrombospondin-1 (TSP-1). Incubation of purified TSP-1 with vWF resulted in formation of thiol-dependent complexes of TSP-1 and vWF, generation of new thiols in vWF, and reduction in the average multimer size of vWF. The ratio of the concentrations of TSP-1 and vWF in plasma reflected with average multimer size of vWF. The higher the plasma TSP-1/vWF molar ratio, the smaller the average vWF multimer size. In addition, administration of TSP-1 to mice resulted in reduction in the average multimer size of plasma vWF. Interaction of TSP-1 with vWF is mediated by TSP-1 type 1 properdin domains and the vWF A3 domain. These results indicate that TSP-1 regulates the multimeric size and therefore hemostatic activity of vWF.


Asunto(s)
Trombospondina 1/metabolismo , Factor de von Willebrand/metabolismo , Adulto , Anciano , Animales , Sitios de Unión , Preescolar , Medios de Cultivo Condicionados/química , Disulfuros/química , Endotelio Vascular/química , Femenino , Humanos , Masculino , Ratones , Modelos Biológicos , Peso Molecular , Oxidorreductasas/sangre , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Estructura Terciaria de Proteína , Púrpura Trombocitopénica Trombótica/sangre , Trombospondina 1/sangre , Trombospondina 1/química , Factor de von Willebrand/química
3.
Cell Mol Life Sci ; 66(9): 1570-9, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19337691

RESUMEN

L-xylulose reductase (XR) is involved in water re-absorption and cellular osmoregulation. The crystal structure of human XR complemented with site-directed mutagenesis (Cys138Ala) indicated that the disulfide bond in the active site between Cys138 and Cys150 is unstable and may affect the reactivity of the enzyme. The effects of reducing agents on the activities of the wild-type and mutant enzymes indicated the reversibility of disulfide-bond formation, which resulted in three-fold decrease in catalytic efficiency. Furthermore, the addition of cysteine (>2 mM) inactivated human XR and was accompanied by a 10-fold decrease in catalytic efficiency. TOF-MS analysis of the inactivated enzyme showed the S-cysteinylation of Cys138 in the wild-type and Cys150 in the mutant enzymes. Thus, the action of human XR may be regulated by cellular redox conditions through reversible disulfide-bond formation and by S-cysteinylation.


Asunto(s)
Deshidrogenasas del Alcohol de Azúcar/química , Sitios de Unión , Cristalografía por Rayos X , Cisteína/química , Humanos , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Oxidación-Reducción , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Deshidrogenasas del Alcohol de Azúcar/genética , Deshidrogenasas del Alcohol de Azúcar/fisiología
4.
J Thromb Haemost ; 16(6): 1198-1210, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29569428

RESUMEN

Essentials Procoagulant platelets can be detected using GSAO in human whole blood. Stable coronary artery disease is associated with a heightened procoagulant platelet response. Agonist-induced procoagulant platelet response is not inhibited by aspirin alone. Collagen plus thrombin induced procoagulant platelet response is partially resistant to clopidogrel. SUMMARY: Background Procoagulant platelets are a subset of highly activated platelets with a critical role in thrombin generation. Evaluation of their clinical utility in thrombotic disorders, such as coronary artery disease (CAD), has been thwarted by the lack of a sensitive and specific whole blood assay. Objectives We developed a novel assay, utilizing the cell death marker, GSAO [(4-(N-(S-glutathionylacetyl)amino)phenylarsonous acid], and the platelet activation marker, P-selectin, to identify procoagulant platelets in whole blood by flow cytometry. Patients/Methods Using this assay, we characterized the procoagulant platelet population in healthy controls and a cohort of patients undergoing elective coronary angiography. Results In patients with CAD, compared with patients without CAD, there was a heightened procoagulant platelet response to thrombin (25.2% vs. 12.2%), adenosine diphosphate (ADP) (7.8% vs. 2.7%) and thrombin plus collagen (27.2% vs. 18.3%). The heightened procoagulant platelet potential in CAD patients was not associated with other markers of platelet function, including aggregation, dense granule release and activation of α2b ß3 integrin. Although dual antiplatelet therapy (DAPT) was associated with partial suppression of procoagulant platelets, this inhibitory effect on a patient level could not be predicted by aggregation response to ADP and was not fully suppressed by clopidogrel. Conclusions We report for the first time that procoagulant platelets can be efficiently detected in a few microliters of whole blood using the cell death marker, GSAO, and the platelet activation marker, P-selectin. A heightened procoagulant platelet response may provide insight into the thrombotic risk of CAD and help identify a novel target for antiplatelet therapies in CAD.


Asunto(s)
Arsenicales/sangre , Coagulación Sanguínea , Plaquetas/metabolismo , Enfermedad de la Arteria Coronaria/sangre , Citometría de Flujo , Glutatión/análogos & derivados , Selectina-P/sangre , Activación Plaquetaria , Pruebas de Función Plaquetaria/métodos , Anciano , Aspirina/farmacología , Biomarcadores/sangre , Coagulación Sanguínea/efectos de los fármacos , Plaquetas/efectos de los fármacos , Estudios de Casos y Controles , Clopidogrel/farmacología , Angiografía Coronaria , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Resistencia a Medicamentos , Femenino , Glutatión/sangre , Humanos , Masculino , Persona de Mediana Edad , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Valor Predictivo de las Pruebas
5.
J Thromb Haemost ; 16(11): 2322-2335, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30207066

RESUMEN

Essentials Nitric oxide synthesis controls protein disulfide isomerase (PDI) function. Nitric oxide (NO) modulation of PDI controls endothelial thrombogenicity. S-nitrosylated PDI inhibits platelet function and thrombosis. Nitric oxide maintains vascular quiescence in part through inhibition of PDI. SUMMARY: Background Protein disulfide isomerase (PDI) plays an essential role in thrombus formation, and PDI inhibition is being evaluated clinically as a novel anticoagulant strategy. However, little is known about the regulation of PDI in the vasculature. Thiols within the catalytic motif of PDI are essential for its role in thrombosis. These same thiols bind nitric oxide (NO), which is a potent regulator of vessel function. To determine whether regulation of PDI represents a mechanism by which NO controls vascular quiescence, we evaluated the effect of NO on PDI function in endothelial cells and platelets, and thrombus formation in vivo. Aim To assess the effect of S-nitrosylation on the regulation of PDI and other thiol isomerases in the vasculature. Methods and results The role of endogenous NO in PDI activity was evaluated by incubating endothelium with an NO scavenger, which resulted in exposure of free thiols, increased thiol isomerase activity, and enhanced thrombin generation on the cell membrane. Conversely, exposure of endothelium to NO+ carriers or elevation of endogenous NO levels by induction of NO synthesis resulted in S-nitrosylation of PDI and decreased surface thiol reductase activity. S-nitrosylation of platelet PDI inhibited its reductase activity, and S-nitrosylated PDI interfered with platelet aggregation, α-granule release, and thrombin generation on platelets. S-nitrosylated PDI also blocked laser-induced thrombus formation when infused into mice. S-nitrosylated ERp5 and ERp57 were found to have similar inhibitory activity. Conclusions These studies identify NO as a critical regulator of vascular PDI, and show that regulation of PDI function is an important mechanism by which NO maintains vascular quiescence.


Asunto(s)
Células Endoteliales/metabolismo , Óxido Nítrico/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Trombosis/metabolismo , Músculos Abdominales/metabolismo , Animales , Plaquetas/citología , Plaquetas/metabolismo , Membrana Celular/metabolismo , Factor Xa/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos C57BL , Selectina-P/metabolismo , Activación Plaquetaria , Agregación Plaquetaria , Proteínas Recombinantes/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Trombina/metabolismo
6.
J Thromb Haemost ; 4(12): 2533-41, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17002656

RESUMEN

Allosteric disulfide bonds control protein function by mediating conformational change when they undergo reduction or oxidation. The known allosteric disulfide bonds are characterized by a particular bond geometry, the -RHStaple. A number of thrombosis and thrombolysis proteins contain one or more disulfide bonds of this type. Tissue factor (TF) was the first hemostasis protein shown to be controlled by an allosteric disulfide bond, the Cys186-Cys209 bond in the membrane-proximal fibronectin type III domain. TF exists in three forms on the cell surface: a cryptic form that is inert, a coagulant form that rapidly binds factor VIIa to initiate coagulation, and a signaling form that binds FVIIa and cleaves protease-activated receptor 2, which functions in inflammation, tumor progression and angiogenesis. Reduction and oxidation of the Cys186-Cys209 disulfide bond is central to the transition between the three forms of TF. The redox state of the bond appears to be controlled by protein disulfide isomerase and NO. Plasmin(ogen), vitronectin, glycoprotein 1balpha, integrin beta(3) and thrombomodulin also contain -RHStaple disulfides, and there is circumstantial evidence that the function of these proteins may involve cleavage/formation of these disulfide bonds.


Asunto(s)
Sitio Alostérico , Disulfuros/química , Disulfuros/metabolismo , Fibrinólisis , Trombosis/metabolismo , Animales , Proteínas Sanguíneas/química , Proteínas Sanguíneas/metabolismo , Activación Enzimática , Fibrinolisina/química , Fibrinolisina/metabolismo , Glicoproteínas/química , Glicoproteínas/metabolismo , Humanos , Inmunoglobulinas/química , Inmunoglobulinas/metabolismo , Integrina beta3/química , Integrina beta3/metabolismo , Oxidación-Reducción , Plasminógeno/química , Plasminógeno/metabolismo , Unión Proteica , Conformación Proteica , Trombomodulina/química , Trombomodulina/metabolismo , Tromboplastina/química , Tromboplastina/metabolismo , Trombosis/sangre , Vitronectina/química , Vitronectina/metabolismo
7.
Biochim Biophys Acta ; 843(3): 159-63, 1985 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-2415166

RESUMEN

A general counterpart of the Scatchard analysis has been developed which takes into account the valence of the ligand. Its use is first demonstrated by application to binding data obtained by exclusion chromatography of mixtures of Dextran T2000 and concanavalin A (a bivalent ligand) on a column of porous glass beads (Glyceryl-CPG 170) equilibrated at 5 degrees C with phosphate-chloride buffer (pH 5.5), I 0.5. A recycling partition equilibrium study with Sephadex G-100 as gel phase then provides a quantitative evaluation of the interaction between haemoglobin and a monoclonal mouse antihaemoglobin antibody preparation in 0.1 M phosphate (pH 7.0) in order to emphasize the ability of the present analysis to consider collectively binding results obtained with a range of acceptor concentrations. Finally, the use of the generalized Scatchard analysis to assess acceptor site homogeneity is illustrated by reappraisal of results for the binding of glyceraldehyde-3-phosphate dehydrogenase to erythrocyte membranes.


Asunto(s)
Sitios de Unión de Anticuerpos , Sitios de Unión , Ligandos , Animales , Anticuerpos Monoclonales/análisis , Reacciones Antígeno-Anticuerpo , Cromatografía de Afinidad , Concanavalina A , Dextranos , Membrana Eritrocítica/metabolismo , Gliceraldehído-3-Fosfato Deshidrogenasas/sangre , Hemoglobinas/inmunología , Humanos , Matemática , Ratones , Unión Proteica
8.
Biochim Biophys Acta ; 1073(3): 609-13, 1991 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-2015284

RESUMEN

The technique of competitive chromogenic substrate hydrolysis is used to examine the inhibitory effects of sucrose and glycerol on the inactivation of thrombin by antithrombin III. This inhibition is attributed to the existence of a slight increase in volume/asymmetry associated with formation of the thrombin-antithrombin complex that subsequently undergoes covalent modification in an irreversible inactivation step. Partial reversal of the equilibrium step is thus considered to result from the effects of molecular crowding in the highly concentrated environment that is generated by the inclusion of these small insert solutes.


Asunto(s)
Antitrombina III/metabolismo , Péptido Hidrolasas/metabolismo , Trombina/metabolismo , Antitrombina III/efectos de los fármacos , Glicerol/farmacología , Cinética , Modelos Químicos , Péptido Hidrolasas/efectos de los fármacos , Sacarosa/farmacología , Termodinámica , Trombina/efectos de los fármacos
9.
Biochim Biophys Acta ; 1388(2): 478-88, 1998 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-9858782

RESUMEN

Thrombospondin-1 is a matrix protein that inhibits proliferation, motility and sprouting of endothelial cells in vitro and angiogenesis in vivo. One mechanism by which thrombospondin-1 may influence endothelial cell biology is through interaction with the endothelial cell alphav beta3 integrin receptor. This interaction is mediated via a cryptic Arg-Gly-Asp sequence in the C-terminal Ca2+-binding region of thrombospondin-1. Exposure of the Arg-Gly-Asp sequence is controlled by disulfide interchange events in the Ca2+-binding loops and C-globular domain. Limited reduction of thrombospondin-1 by dithiothreitol exposes the Arg-Gly-Asp sequence which can bind to the alphav beta3 integrin receptor and support endothelial cell spreading (X. Sun, K. Skorstengaard, D.F. Mosher, J. Cell Biol. 118 (1992) 693-701). Our aim was to identify possible physiological reductants that can mediate Arg-Gly-Asp exposure. We now report that protein disulfide isomerase, which is known to catalyze disulfide interchange in thrombospondin-1 and change its enzyme inhibitory properties and its binding to monoclonal antibodies, was secreted by bovine aortic endothelial cells and deposited on the cell surface. There was an average of approximately 2.2 fg of protein disulfide isomerase on the surface of a bovine aortic endothelial cell. Treatment of thrombospondin-1 with purified protein disulfide isomerase enhanced adhesion of endothelial cells to thrombospondin-1 in an Arg-Gly-Asp-dependent manner through the alphav beta3 integrin receptor and supported cell spreading. Both Ca2+-depleted and Ca2+-replete thrombospondin-1 were substrates for protein disulfide isomerase. These results suggest that endothelial cell derived protein disulfide isomerase may regulate Arg-Gly-Asp-dependent binding of thrombospondin-1.


Asunto(s)
Oligopéptidos/química , Proteína Disulfuro Isomerasas/análisis , Trombospondina 1/química , Animales , Aorta/enzimología , Plaquetas/metabolismo , Bovinos , Adhesión Celular/fisiología , Membrana Celular/enzimología , Células Cultivadas , Disulfuros/metabolismo , Ditiotreitol/metabolismo , Endotelio Vascular/enzimología , Glutatión/metabolismo , Humanos , Inmunohistoquímica , Microscopía de Contraste de Fase
10.
Biochim Biophys Acta ; 1296(2): 138-44, 1996 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-8814219

RESUMEN

Thrombospondin 1 (TSP1) is a multidomain glycoprotein from platelets and cells which functions in cell-cell and cell-matrix interactions. The structure of TSP1 is regulated by sulfhydryl-disulfide interchange in the carboxy-terminal Ca2(+)-binding loops and globular domain which markedly influence its interaction with cell surface integrins and its inhibition of neutrophil enzymes. We have identified murine monoclonal antibodies that recognized different disulfide-bonded forms of TSP1, made by preparing TSP1 in buffers containing either 0.1 mM or 2 mM Ca2+. Antibody HB8432 recognizes TSP1 prepared in buffers containing either 0.1 or 2 mM Ca2+, while antibodies D4.6 and A65M recognized only TSP1 prepared in buffers containing 0.1 mM Ca2+. The antibodies recognize these different TSP1 preparations either adsorbed to plastic or extracellular matrix. Immunohistochemistry of human rheumatoid synovial tissue using HB8432 resulted in staining of numerous blood vessel walls and matrix cells, while D4.6 and A65M stained a subset of the HB8432 positive blood vessels and only occasionally stained matrix cells. These results suggested that different disulfide-bonded forms of TSP1 were being expressed in different areas of inflamed tissue.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Cistina/química , Epítopos/inmunología , Glicoproteínas de Membrana/inmunología , Adsorción , Animales , Especificidad de Anticuerpos , Reacciones Antígeno-Anticuerpo , Artritis Reumatoide/inmunología , Artritis Reumatoide/metabolismo , Colágeno , Combinación de Medicamentos , Epítopos/química , Humanos , Laminina , Glicoproteínas de Membrana/análisis , Glicoproteínas de Membrana/química , Ratones , Plásticos , Unión Proteica , Estructura Terciaria de Proteína , Proteoglicanos , Conejos , Membrana Sinovial/química , Trombospondinas
11.
Mol Immunol ; 24(7): 797-801, 1987 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-3657807

RESUMEN

A procedure is devised for simple graphical evaluation of the association constant for antibody-antigen interactions from data obtained by conventional solid-phase immunoassay for antigen. Application of the method to situations involving a univalent antigen is illustrated by means of ELISA data for the interactions of paraquat with its elicited murine monoclonal antibody, and the Fab fragment derived therefrom. Although the completely general situation with both antibody and antigen multivalent is not amenable to study by the present procedure, the quantitative expression is readily modified to accommodate antigen multivalency provided that the univalent Fab fragment may be substituted for the multivalent antibody (IgG or IgM) as partitioning species in the solid-phase immunoassay.


Asunto(s)
Reacciones Antígeno-Anticuerpo , Paraquat/inmunología , Animales , Anticuerpos Monoclonales , Ensayo de Inmunoadsorción Enzimática , Fragmentos Fab de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C
12.
Protein Sci ; 9(12): 2436-45, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11206065

RESUMEN

It has been proposed that certain cell-surface proteins undergo redox reactions, that is, transfer of hydrogens and electrons between closely spaced cysteine thiols that can lead to reduction, formation, or interchange of disulfide bonds. This concept was tested using a membrane-impermeable trivalent arsenical to identify closely spaced thiols in cell-surface proteins. We attached the trivalent arsenical, phenylarsenoxide, to the thiol of reduced glutathione to produce 4-(N-(S-glutathionylacetyl)amino)phenylarsenoxide (GSAO). GSAO bound tightly to synthetic, peptide, and protein dithiols like thioredoxin, but not to monothiols. To identify cell-surface proteins that contain closely spaced thiols, we attached a biotin moiety through a spacer arm to the primary amino group of the gamma-glutamyl residue of GSAO (GSAO-B). Incorporation of GSAO-B into proteins was assessed by measuring the biotin using streptavidin-peroxidase. Up to 12 distinct proteins were labeled with GSAO-B on the surface of endothelial and fibrosarcoma cells. The pattern of labeled proteins differed between the different cell types. Protein disulfide isomerase was one of the proteins on the endothelial and fibrosarcoma cell surface that incorporated GSAO-B. These findings demonstrate that the cell-surface environment can support the existence of closely spaced protein thiols and suggest that at least some of these thiols are redox active.


Asunto(s)
Proteínas de la Membrana/química , Compuestos de Sulfhidrilo/química , Animales , Arsenicales/metabolismo , Biotina/metabolismo , Electroforesis en Gel de Poliacrilamida , Endotelio/química , Endotelio/citología , Fibrosarcoma/química , Fibrosarcoma/patología , Glutatión/análogos & derivados , Glutatión/metabolismo , Humanos , Indicadores y Reactivos , Mamíferos , Proteínas de la Membrana/metabolismo , Oxidación-Reducción , Proteína Disulfuro Isomerasas/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Tiorredoxinas/metabolismo
13.
Thromb Haemost ; 84(3): 506-13, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11019979

RESUMEN

The haemostatic activity of plasma von Willebrand factor (vWF) is a function of multimer size. Only the large vWF multimers are effective in promoting platelet adhesion to a site of vascular injury. We observed that the conditioned medium of cultured human umbilical vein, human microvascular and bovine aortic endothelial cells contained an activity which reduced the average multimer size of plasma or purified vWF. The average multimer size of vWF produced endogenously by human umbilical vein endothelial cells was similarly reduced following secretion. The reducing activity was ablated by pre-treatment with heat or the thiol blocking agents. iodoacetamide, N-ethylmaleimide or E-64, but not by a range of specific serine-, cysteine-, aspartic-, or metalloproteinase inhibitors. Reduction in vWF multimer size was associated with formation of new thiols in vWF and there was no evidence for additional proteolytic processing of vWF. The reducing activity was associated with a protein with an anionic pi that binds heparin and contains reactive thiol(s). These results suggested that the interchain disulfide bonds that link the vWF homodimers near the N-termini were being reduced by a vWF reductase secreted by endothelial cells. In support of this hypothesis, incubation of vWF with the protein reductants, protein disulfide isomerase and thioredoxin, resulted in formation of new thiols in vWF and reduction in the average multimer size of vWF. These findings may have consequences for control of vWF haemostatic activity.


Asunto(s)
Endotelio Vascular/citología , Factor de von Willebrand/química , Factor de von Willebrand/metabolismo , Animales , Aorta , Bovinos , Disulfuros/metabolismo , Endotelio Vascular/química , Endotelio Vascular/efectos de los fármacos , Calor , Humanos , Maleimidas/farmacología , Oxidación-Reducción , Oxidorreductasas/química , Polímeros/metabolismo , Compuestos de Sulfhidrilo/metabolismo , Trombina/farmacología , Factores de Tiempo , Venas Umbilicales
14.
Thromb Haemost ; 70(2): 342-5, 1993 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-8236146

RESUMEN

Antiphospholipid antibodies, defined either by lupus anticoagulant (LA) activity or positive anticardiolipin immunoabsorbent assay (ACA) are associated with a predisposition to thromboses, recurrent fetal loss or thrombocytopenia. The mechanisms for these predispositions remain undefined. We have enriched immunoglobulin fractions from two patient plasmas to obtain antibodies with LA activity but no ACA, or conversely, with ACA positivity but no LA, in order to investigate in vitro characteristics which might explain a thrombotic propensity. beta 2-glycoprotein I (beta 2-GPI), the plasma cofactor required for ACA binding to negatively charged phospholipid, has previously been shown to inhibit prothrombinase generation in the presence of activated platelets (8). We now report that beta 2-GPI, at physiological concentrations, inhibits the generation of factor Xa in the presence of activated gel-filtered platelets. Further, ACA interferes with this inhibition, resulting in protracted, unopposed factor Xa generation. This interference with beta 2-GPI, a natural anticoagulant component of plasma, is potentially prothrombotic. LA immunoglobulins behave differently and inhibit factor Xa generation in a manner similar to beta 2-GPI. These findings provide the basis for a previously unsuspected mechanism for thrombosis in patients with aPL.


Asunto(s)
Anticuerpos Anticardiolipina/farmacología , Síndrome Antifosfolípido/sangre , Enfermedades Autoinmunes/sangre , Plaquetas/metabolismo , Factor Xa/biosíntesis , Glicoproteínas/antagonistas & inhibidores , Inhibidor de Coagulación del Lupus/farmacología , Adulto , Anticuerpos Anticardiolipina/aislamiento & purificación , Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/inmunología , Enfermedades Autoinmunes/inmunología , Factores de Coagulación Sanguínea/metabolismo , Femenino , Humanos , Inhibidor de Coagulación del Lupus/aislamiento & purificación , Masculino , Persona de Mediana Edad , Trombosis/etiología , beta 2 Glicoproteína I
15.
Ann N Y Acad Sci ; 667: 64-9, 1992 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-1309073

RESUMEN

Thrombospondin is a large, trimeric glycoprotein secreted by activated platelets and growing cells. Thrombospondin copolymerizes with fibrin during blood coagulation and deposits in extracellular matrix. We found that thrombospondin is a slow (rate constant approximately 6.3 x 10(3) M-1 sec-1), tight-binding (Kd < 10(-9) M) inhibitor of plasmin as determined by loss of amidolytic activity, loss of ability to degrade fibrinogen, and decreased lysis zones in fibrin plate assays (Biochemistry 31: 265-269, 1992). Thrombospondin also slowly inhibits urokinase plasminogen activator. The lysis zone when urokinase is put on fibrin plates made from whole plasma is less if thrombospondin is present. The stoichiometry of inhibition is approximately one mole plasmin:one mole thrombospondin trimer, a somewhat surprising result considering the trimeric nature of thrombospondin. These results indicate that thrombospondin is an important regulator of fibrinolysis and degradation of extracellular matrix, particularly when these processes are initiated by urokinase and even when other inhibitors of fibrinolysis are present.


Asunto(s)
Plaquetas/fisiología , Fibrinólisis/fisiología , Glicoproteínas de Membrana/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Fibrinólisis/efectos de los fármacos , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/farmacología , Trombospondinas , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
16.
Thromb Res ; 79(4): 353-61, 1995 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-7482438

RESUMEN

The adherent human hepatoma cell line Hep G2 exhibits receptor mediated endocytosis and catabolism of tissue-type plasminogen activator.plasminogen activator inhibitor type-1 (t-PA.PAI-1) complexes formed when exogenous t-PA combines with endogenous PAI-1 in the extracellular matrix. To determine whether the other major PA, urokinase (u-PA), which also complexes with PAI-1, is metabolised via the same mechanism, 125I-labelled high (hmw) and low (lmw) molecular weight forms of u-PA were incubated with Hep G2 cells at 4 degrees C for 2 hr in the absence and presence of a 100-fold excess of unlabelled ligand in order to detect specific binding. Both hmw and lmw 125I-u-PA formed complexes with PAI-1 and these bound specifically and with high affinity (apparent Kd 3.9 and 4.1 nM, with Bmax 78 x 10(3) and 83 x 10(3) binding sites/cell respectively). Binding by each form of radiolabelled u-PA was inhibited in a dose-dependent fashion by unlabelled t-PA, hmw-u-PA, lmw-u-PA, and by monoclonal anti-PAI-1 antibody. At 37 degrees C, bound hmw and lmw 125I-u-PA.PAI-1 complexes were internalised and degraded rapidly. These findings indicate that the specificity of the previously described receptor which mediates PAI-1 dependent catabolism of t-PA by Hep G2 cells extends to complexes of u-PA with this inhibitor.


Asunto(s)
Inhibidor 1 de Activador Plasminogénico/metabolismo , Activador de Tejido Plasminógeno/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Anticuerpos Monoclonales , Sitios de Unión , Línea Celular , Endocitosis , Humanos , Cinética , Peso Molecular , Inhibidor 1 de Activador Plasminogénico/inmunología , Receptores de Superficie Celular/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Células Tumorales Cultivadas , Activador de Plasminógeno de Tipo Uroquinasa/química
17.
Biophys Chem ; 75(3): 187-99, 1998 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-9894338

RESUMEN

Kinetic evidence is presented for the interaction of prothrombin with several distinctive topological regions of the thrombin molecule. Modulations of thrombin catalytic activity on the protein substrates prothrombin and prethrombin 1 are demonstrated that involve the fragment 1 and fragment 2 portions. The inhibitory effects are demonstrably non-competitive. In addition to exhibiting non-competitive inhibition, fragment 2 is capable of enhancing proteolysis by thrombin; and therefore to react with a second region of the enzyme. On the basis of the crystallographic studies of the complex between fragment 2 and thrombin (Arni et al., Biochemistry 32 (1992) 4727), this activating site is proposed to be associated with exosite II. The allosteric switch between procoagulant and anticoagulant activities identified from studies by Di Cera (Dang et al., Proc. Natl. Acad. Sci USA 92 (1995) 5977) could be 'thrown' by a macromolecular effector that is generated during thrombin formation--a plausible mechanism for switching that deserves further investigation.


Asunto(s)
Protrombina/química , Protrombina/metabolismo , Trombina/química , Trombina/metabolismo , Sitio Alostérico , Dominio Catalítico , Fenómenos Químicos , Química Física , Compuestos Cromogénicos/metabolismo , Precursores Enzimáticos/química , Precursores Enzimáticos/metabolismo , Humanos , Técnicas In Vitro , Cinética , Sustancias Macromoleculares , Oligopéptidos/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Especificidad por Sustrato
18.
J Thromb Haemost ; 11 Suppl 1: 277-84, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23809131

RESUMEN

Tissue factor (TF) is a transmembrane cofactor that binds and promotes the catalytic activity of factor (F) VIIa. The TF/VIIa complex activates FX by limited proteolysis to initiate blood coagulation and helps provide the thrombin burst that is important for a stable thrombus. TF is present both in the extravascular compartment, where it functions as a hemostatic envelope, and the intravascular compartment, where it contributes to thrombus formation, particularly when endothelial disruption is minimal. The regulation of its cofactor function appears to differ in the two compartments. Intravascular TF derives predominately from leucocytes, with either monocytes or neutrophils implicated in different models of thrombosis. This TF exists mostly in a non-coagulant or cryptic form and acute events lead to local decryption of TF and FX activation. A variety of experimental observations imply that decryption of leucocyte surface TF involves both a dithiol/disulfide switch and exposure of phosphatidylserine. The dithiol/disulfide switch appears to involve the Cys186-Cys209 disulfide bond in the membrane-proximal domain of TF, although this has not been demonstrated in vivo. Activation of a purinergic receptor or complement has recently been observed to decrypt TF on myeloid cells and a dithiol/disulfide switch and the oxidoreductase, protein disulfide isomerase, have been implicated in both systems. The molecular mechanism of action of protein disulfide isomerase in TF encryption/decryption, though, remains to be determined.


Asunto(s)
Tromboplastina/metabolismo , Coagulación Sanguínea , Humanos
19.
Cell Death Dis ; 4: e473, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23348587

RESUMEN

Traumatic brain injury is a major public health concern and is characterised by both apoptotic and necrotic cell death in the lesion. Anatomical imaging is usually used to assess traumatic brain injuries and there is a need for imaging modalities that provide complementary cellular information. We sought to non-invasively image cell death in a mouse model of traumatic brain injury using a near-infrared fluorescent conjugate of a synthetic heat shock protein-90 alkylator, 4-(N-(S-glutathionylacetyl) amino) phenylarsonous acid (GSAO). GSAO labels both apoptotic and necrotic cells coincident with loss of plasma membrane integrity. The optical GSAO specifically labelled apoptotic and necrotic cells in culture and did not accumulate in healthy organs or tissues in the living mouse body. The conjugate is a very effective imager of cell death in brain lesions. The optical GSAO was detected by fluorescence intensity and GSAO bound to dying/dead cells was detected from prolongation of the fluorescence lifetime. An optimal signal-to-background ratio was achieved as early as 3 h after injection of the probe and the signal intensity positively correlated with both lesion size and probe concentration. This optical GSAO offers a convenient and robust means to non-invasively image apoptotic and necrotic cell death in brain and other lesions.


Asunto(s)
Apoptosis , Arsenicales/química , Lesiones Encefálicas/diagnóstico , Glutatión/análogos & derivados , Proteínas HSP90 de Choque Térmico/química , Alquilación , Animales , Arsenicales/farmacología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacología , Glutatión/química , Glutatión/farmacología , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Células Jurkat , Ratones , Espectroscopía Infrarroja Corta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA